Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 7(4): e21899, 2012.
Article in English | MEDLINE | ID: mdl-22514596

ABSTRACT

Diabetic conditions are associated with modified brain function, namely with cognitive deficits, through largely undetermined processes. More than understanding the underlying mechanism, it is important to devise novel strategies to alleviate diabetes-induced cognitive deficits. Caffeine (a mixed antagonist of adenosine A(1) and A(2A) receptors) emerges as a promising candidate since caffeine consumption reduces the risk of diabetes and effectively prevents memory deficits caused by different noxious stimuli. Thus, we took advantage of a novel animal model of type 2 diabetes to investigate the behavioural, neurochemical and morphological modifications present in the hippocampus and tested if caffeine consumption might prevent these changes. We used a model closely mimicking the human type 2 diabetes condition, NONcNZO10/LtJ mice, which become diabetic at 7-11 months when kept under an 11% fat diet. Caffeine (1 g/l) was applied in the drinking water from 7 months onwards. Diabetic mice displayed a decreased spontaneous alternation in the Y-maze accompanied by a decreased density of nerve terminal markers (synaptophysin, SNAP25), mainly glutamatergic (vesicular glutamate transporters), and increased astrogliosis (GFAP immunoreactivity) compared to their wild type littermates kept under the same diet. Furthermore, diabetic mice displayed up-regulated A(2A) receptors and down-regulated A(1) receptors in the hippocampus. Caffeine consumption restored memory performance and abrogated the diabetes-induced loss of nerve terminals and astrogliosis. These results provide the first evidence that type 2 diabetic mice display a loss of nerve terminal markers and astrogliosis, which is associated with memory impairment; furthermore, caffeine consumption prevents synaptic dysfunction and astrogliosis as well as memory impairment in type 2 diabetes.


Subject(s)
Caffeine/therapeutic use , Diabetes Mellitus, Experimental/complications , Hippocampus/drug effects , Hippocampus/metabolism , Memory Disorders/drug therapy , Memory Disorders/etiology , Animals , Blotting, Western , Caffeine/blood , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Immunohistochemistry , Insulin/blood , Male , Memory Disorders/blood , Mice
2.
Eur J Neurosci ; 34(1): 12-21, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21615561

ABSTRACT

Adenosine neuromodulation depends on a balanced activation of inhibitory A1 (A1R) and facilitatory A(2A) receptors (A(2A) R). Both A1 R and A(2A) R modulate hippocampal glutamate release and NMDA-dependent long-term potentiation (LTP) but ageing affects the density of both A1 R and A(2A) R. We tested the effects of selective A1 R and A(2A) R antagonists in the modulation of synaptic transmission and plasticity in rat hippocampal slices from three age groups (young adults, 2-3 month; middle-aged adults, 6-8 months; aged, 18-20 months). The selective A(2A) R antagonist SCH58261 (50 nm) attenuated LTP in all age groups, with a larger effect in aged (-63 ± 7%) than in middle-aged adults (-36 ± 9%) or young adult rats (-36 ± 9%). In contrast, the selective A1 R antagonist DPCPX (50 nm) increased LTP magnitude in young adult rats (+42 ± 6%), but failed to affect LTP magnitude in the other age groups. Finally, in the continuous presence of DPCPX, SCH58261 caused a significantly larger inhibition of LTP amplitude in aged (-71 ± 45%) than middle-aged (-28 ±9%) or young rats (-11 ± 2%). Accordingly, aged rats displayed an increased expression of A(2A) R mRNA in the hippocampus and a higher number of glutamatergic nerve terminals equipped with A(2A) R in aged (67 ± 6%) compared with middle-aged (34 ± 7%) and young rats (25 ± 5%). The results show an enhanced A(2A) R-mediated modulation of LTP in aged rats, in accordance with the age-associated increased expression and density of A(2A) R in glutamatergic terminals. This age-associated gain of function of A(2A) R modulating synaptic plasticity may underlie the ability of A(2A) R antagonists to prevent memory dysfunction in aged animals.


Subject(s)
Aging/physiology , Hippocampus/physiology , Long-Term Potentiation/physiology , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine/metabolism , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Male , Memory Disorders/physiopathology , N-Methylaspartate/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Pyrimidines/pharmacology , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Triazoles/pharmacology , Xanthines/pharmacology
3.
J Neurochem ; 117(1): 100-11, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21235574

ABSTRACT

The blockade of adenosine A(2A) receptors (A2AR) affords a robust neuroprotection in different noxious brain conditions. However, the mechanisms underlying this general neuroprotection are unknown. One possible mechanism could be the control of neuroinflammation that is associated with brain damage, especially because A2AR efficiently control peripheral inflammation. Thus, we tested if the intracerebroventricular injection of a selective A2AR antagonist (SCH58261) would attenuate the changes in the hippocampus triggered by intraperitoneal administration of lipopolysaccharide (LPS) that induces neuroinflammation through microglia activation. LPS administration triggers an increase in inflammatory mediators like interleukin-1ß that causes biochemical changes (p38 and c-jun N-terminal kinase phosphorylation and caspase 3 activation) contributing to neuronal dysfunction typified by decreased long-term potentiation, a form of synaptic plasticity. Long-term potentiation, measured 30 min after the tetanus, was significantly lower in LPS-treated rats compared with control-treated rats, while SCH58261 attenuated the LPS-induced change. The LPS-induced increases in phosphorylation of c-jun N-terminal kinase and p38 and activation of caspase 3 were also prevented by SCH58261. Significantly, SCH58261 also prevented the LPS-induced recruitment of activated microglial cells and the increase in interleukin-1ß concentration in the hippocampus, indicating that A2AR activation is a pivotal step in mediating the neuroinflammation triggered by LPS. These results indicate that A2AR antagonists prevent neuroinflammation and support the hypothesis that this mechanism might contribute for the ability of A2AR antagonists to control different neurodegenerative diseases known to involve neuroinflammation.


Subject(s)
Hippocampus/pathology , Inflammation Mediators/physiology , Neural Inhibition/physiology , Neurons/pathology , Receptor, Adenosine A2A/physiology , Animals , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/physiology , Male , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Receptor, Adenosine A2A/metabolism , Resorcinols/pharmacology , Resorcinols/therapeutic use , Synapses/drug effects , Synapses/metabolism , Synapses/pathology
4.
Biochim Biophys Acta ; 1808(5): 1380-99, 2011 May.
Article in English | MEDLINE | ID: mdl-21145878

ABSTRACT

Adenosine acts in parallel as a neuromodulator and as a homeostatic modulator in the central nervous system. Its neuromodulatory role relies on a balanced activation of inhibitory A(1) receptors (A1R) and facilitatory A(2A) receptors (A2AR), mostly controlling excitatory glutamatergic synapses: A1R impose a tonic brake on excitatory transmission, whereas A2AR are selectively engaged to promote synaptic plasticity phenomena. This neuromodulatory role of adenosine is strikingly similar to the role of adenosine in the control of brain disorders; thus, A1R mostly act as a hurdle that needs to be overcame to begin neurodegeneration and, accordingly, A1R only effectively control neurodegeneration if activated in the temporal vicinity of brain insults; in contrast, the blockade of A2AR alleviates the long-term burden of brain disorders in different neurodegenerative conditions such as ischemia, epilepsy, Parkinson's or Alzheimer's disease and also seem to afford benefits in some psychiatric conditions. In spite of this qualitative agreement between neuromodulation and neuroprotection by A1R and A2AR, it is still unclear if the role of A1R and A2AR in the control of neuroprotection is mostly due to the control of glutamatergic transmission, or if it is instead due to the different homeostatic roles of these receptors related with the control of metabolism, of neuron-glia communication, of neuroinflammation, of neurogenesis or of the control of action of growth factors. In spite of this current mechanistic uncertainty, it seems evident that targeting adenosine receptors might indeed constitute a novel strategy to control the demise of different neurological and psychiatric disorders.


Subject(s)
Brain Diseases/metabolism , Brain Diseases/pathology , Nerve Degeneration , Neuroprotective Agents/pharmacology , Neurotransmitter Agents/physiology , Receptors, Purinergic P1/metabolism , Animals , Humans
5.
J Alzheimers Dis ; 20 Suppl 1: S95-116, 2010.
Article in English | MEDLINE | ID: mdl-20182043

ABSTRACT

Caffeine, the most widely consumed psychoactive drug, enhances attention/vigilance, stabilizes mood, and might also independently enhance cognitive performance. Notably, caffeine displays clearer and more robust beneficial effects on memory performance when memory is perturbed by stressful or noxious stimuli either in human or animal studies. Thus, caffeine restores memory performance in sleep-deprived or aged human individuals, a finding replicated in rodent animal models. Likewise, in animal models of Alzheimer's disease (AD), caffeine alleviates memory dysfunction, which is in accordance with the tentative inverse correlation between caffeine intake and the incidence of AD in different (but not all) cohorts. Caffeine also affords beneficial effects in animal models of conditions expected to impair memory performance such as Parkinson's disease, chronic stress, type 2 diabetes, attention deficit and hyperactivity disorder, early life convulsions, or alcohol-induced amnesia. Thus, caffeine should not be viewed as a cognitive enhancer but instead as a cognitive normalizer. Interestingly, these beneficial effects of caffeine on stress-induced memory disturbance are mimicked by antagonists of adenosine A2A receptors. This prominent role of A2A receptors in preventing memory deterioration is probably related to the synaptic localization of this receptor in limbic areas and its ability to control glutamatergic transmission, especially NMDA receptor-dependent plasticity, and to control apoptosis, brain metabolism, and the burden of neuroinflammation. This opens the real and exciting possibility that caffeine consumption might be a prophylactic strategy and A2A receptor antagonists may be a novel therapeutic option to manage memory dysfunction both in AD and in other chronic neurodegenerative disorders where memory deficits occur.


Subject(s)
Caffeine/administration & dosage , Central Nervous System Stimulants/administration & dosage , Disease Models, Animal , Memory Disorders/prevention & control , Animals , Brain/drug effects , Brain/physiology , Brain Diseases/drug therapy , Brain Diseases/physiopathology , Drug Administration Schedule , Humans , Mental Processes/drug effects , Purinergic P1 Receptor Antagonists , Receptors, Purinergic P1/metabolism
6.
J Neurochem ; 112(2): 453-62, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19878534

ABSTRACT

Seizures early in life cause long-term behavioral modifications, namely long-term memory deficits in experimental animals. Since caffeine and adenosine A(2A) receptor (A(2A)R) antagonists prevent memory deficits in adult animals, we now investigated if they also prevented the long-term memory deficits caused by a convulsive period early in life. Administration of kainate (KA, 2 mg/kg) to 7-days-old (P7) rats caused a single period of self-extinguishable convulsions which lead to a poorer memory performance in the Y-maze only when rats were older than 90 days, without modification of locomotion or anxiety-like behavior in the elevated-plus maze. In accordance with the relationship between synaptotoxicity and memory dysfunction, the hippocampus of these adult rats treated with kainate at P7 displayed a lower density of synaptic proteins such as SNAP-25 and syntaxin (but not synaptophysin), as well as vesicular glutamate transporters type 1 (but not vesicular GABA transporters), with no changes in PSD-95, NMDA receptor subunits (NR1, NR2A, NR2B) or alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor subunits (GluR1, GluR2) compared with controls. Caffeine (1 g/L) or the A(2A)R antagonist, KW6002 (3 mg/kg) applied in the drinking water from P21 onwards, prevented these memory deficits in P90 rats treated with KA at P7, as well as the accompanying synaptotoxicity. These results show that a single convulsive episode in early life causes a delayed memory deficit in adulthood accompanied by a glutamatergic synaptotoxicity that was prevented by caffeine or adenosine A(2A)R antagonists.


Subject(s)
Adenosine A2 Receptor Antagonists , Caffeine/administration & dosage , Memory Disorders/prevention & control , Neurotoxicity Syndromes/prevention & control , Phosphodiesterase Inhibitors/administration & dosage , Purines/administration & dosage , Analysis of Variance , Animals , Animals, Newborn , Disease Models, Animal , Disease Progression , Drug Administration Schedule , Glial Fibrillary Acidic Protein/metabolism , Kainic Acid , Memory Disorders/etiology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Pyrimidines/pharmacology , Qa-SNARE Proteins/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/chemically induced , Seizures/complications , Synapses/drug effects , Synapses/metabolism , Synaptophysin/metabolism , Synaptosomal-Associated Protein 25/metabolism , Triazoles/pharmacology , Tritium/metabolism , Xanthines/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...