Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
Int J Mol Sci ; 23(2)2022 Jan 06.
Article En | MEDLINE | ID: mdl-35054778

Myotonic dystrophy type 1 (DM1) is a severe neuromuscular disease mediated by a toxic gain of function of mutant RNAs. The neuropsychological manifestations affect multiple domains of cognition and behavior, but their etiology remains elusive. Transgenic DMSXL mice carry the DM1 mutation, show behavioral abnormalities, and express low levels of GLT1, a critical regulator of glutamate concentration in the synaptic cleft. However, the impact of glutamate homeostasis on neurotransmission in DM1 remains unknown. We confirmed reduced glutamate uptake in the DMSXL hippocampus. Patch clamp recordings in hippocampal slices revealed increased amplitude of tonic glutamate currents in DMSXL CA1 pyramidal neurons and DG granule cells, likely mediated by higher levels of ambient glutamate. Unexpectedly, extracellular GABA levels and tonic current were also elevated in DMSXL mice. Finally, we found evidence of synaptic dysfunction in DMSXL mice, suggestive of abnormal short-term plasticity, illustrated by an altered LTP time course in DG and in CA1. Synaptic dysfunction was accompanied by RNA foci accumulation in localized areas of the hippocampus and by the mis-splicing of candidate genes with relevant functions in neurotransmission. Molecular and functional changes triggered by toxic RNA may induce synaptic abnormalities in restricted brain areas that favor neuronal dysfunction.


Hippocampus/metabolism , Myotonic Dystrophy/physiopathology , Myotonin-Protein Kinase/physiology , Neuronal Plasticity , Neurotransmitter Agents/metabolism , RNA Splicing , Animals , Disease Models, Animal , Excitatory Amino Acid Transporter 2 , Hippocampus/physiology , Homeostasis , Mice , Mice, Transgenic , Myotonic Dystrophy/metabolism , Myotonin-Protein Kinase/genetics , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , RNA/metabolism , Synaptic Transmission
2.
Prog Neurobiol ; 206: 102139, 2021 11.
Article En | MEDLINE | ID: mdl-34391810

Alzheimer's disease is the most common form of dementia characterized by intracellular aggregates of hyperphosphorylated Tau protein and extracellular accumulation of amyloid ß (Aß) peptides. We previously demonstrated that the purinergic receptor P2X7 (P2X7) plays a major role in Aß-mediated neurodegeneration but the relationship between P2X7 and Tau remained overlooked. Such a link was supported by cortical upregulation of P2X7 in patients with various type of frontotemporal lobar degeneration, including mutation in the Tau-coding gene, MAPT, as well as in the brain of a Tauopathy mouse model (THY-Tau22). Subsequent phenotype analysis of P2X7-deficient Tau mice revealed the instrumental impact of this purinergic receptor. Indeed, while P2X7-deficiency had a moderate effect on Tau pathology itself, we observed a significant reduction of microglia activation and of Tau-related inflammatory mediators, particularly CCL4. Importantly, P2X7 deletion ultimately rescued synaptic plasticity and memory impairments of Tau mice. Altogether, the present data support a contributory role of P2X7 dysregulation on processes governing Tau-induced brain anomalies. Due to the convergent role of P2X7 blockade in both Aß and Tau background, P2X7 inhibitors might prove to be ideal candidate drugs to curb the devastating cognitive decline in Alzheimer's disease and Tauopathies.


Alzheimer Disease , Receptors, Purinergic P2X7/deficiency , Tauopathies , Alzheimer Disease/genetics , Amyloid beta-Peptides , Animals , Cognition , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Tauopathies/genetics , tau Proteins/genetics
3.
Neuropharmacology ; 164: 107902, 2020 03 01.
Article En | MEDLINE | ID: mdl-31811873

Vesicular glutamate transporters (VGLUT1-3) mediate the uptake of glutamate into synaptic vesicles. VGLUTs are pivotal actors of excitatory transmission and of almost all brain functions. Their implication in various pathologies has been clearly documented. Despite their functional importance, the pharmacology of VGLUTs is limited to a few dyes such as Trypan Blue, Rose Bengal or Brilliant Yellow type. Here, we report the design and evaluation of new potent analogs based on Trypan Blue scaffold. Our best compound, named LSP5-2157, has an EC50 of 50 nM on glutamate vesicular uptake. Using a 3D homology model of VGLUT1 and docking experiments, we determined its putative binding subdomains within vesicular glutamate transporters and validated the structural requirement for VGLUT inhibition. To better estimate the specificity and potency of LSP5-2157, we also investigated its ability to block glutamatergic transmission in autaptic hippocampal cells. Neither glutamate receptors nor GABAergic transmission or transmission machinery were affected by LSP5-2157. Low doses of compound reversibly reduce glutamatergic neurotransmission in hippocampal autpases. LSP5-2157 had a low and depressing effect on synaptic efficacy in hippocampal slice. Furthermore, LSP5-2157 had no effect on NMDA-R- mediated fEPSP but reduce synaptic plasticity induced by 3 trains of 100 Hz. Finally, LSP5-2157 had the capacity to inhibit VGLUT3-dependent auditory synaptic transmission in the guinea pig cochlea. In this model, it abolished the compound action potential of auditory nerve at high concentration showing the limited permeation of LSP5-2157 in an in-vivo model. In summary, the new ligand LSP5-2157, has a high affinity and specificity for VGLUTs and shows some permeability in isolated neuron, tissue preparations or in vivo in the auditory system. These findings broaden the field of VGLUTs inhibitors and open the way to their use to assess glutamatergic functions in vitro and in vivo.


Vesicular Glutamate Transport Proteins/antagonists & inhibitors , Action Potentials/drug effects , Animals , Cochlea/drug effects , Cochlear Nerve/drug effects , Excitatory Postsynaptic Potentials/drug effects , Guinea Pigs , Hippocampus/cytology , Hippocampus/drug effects , Mice , Mice, Inbred C57BL , Models, Molecular , Neurons/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Vesicular Glutamate Transport Proteins/metabolism
4.
Sci Rep ; 9(1): 20138, 2019 12 27.
Article En | MEDLINE | ID: mdl-31882891

The prevalence of cognitive decline is increasing as the ageing population is considerably growing. Restricting this age-associated process has become a challenging public health issue. The age-related increase in oxidative stress plays a major role in cognitive decline, because of its harmful effect on functional plasticity of the brain, such as long-term potentiation (LTP). Here, we show that citrulline (Cit) has powerful antioxidant properties that can limit ex vivo oxidative stress-induced LTP impairment in the hippocampus. We also illustrate that a three-month Cit supplementation has a protective effect on LTP in aged rats in vivo. The identification of a Cit oxidation byproduct in vitro suggests that the antioxidant properties of Cit could result from its own oxidation. Cit supplementation may be a promising preventive nutritional approach to limit age-related cognitive decline.


Aging , Citrulline/pharmacology , Long-Term Potentiation/drug effects , Aging/metabolism , Animals , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Mice , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Rats
5.
Neurochem Res ; 44(3): 692-702, 2019 Mar.
Article En | MEDLINE | ID: mdl-30426348

Aging, as the major risk factor of Alzheimer's disease (AD), may increase susceptibility to neurodegenerative diseases through many gradual molecular and biochemical changes. Extracellular glutamate homeostasis and extrasynaptic glutamate N-methyl-D-aspartate receptors (NMDAR) are among early synaptic targets of oligomeric amyloid ß (Aßo), one of the AD related synaptotoxic protein species. In this study, we asked for the effects of Aßo on long-term depression (LTD), a form of synaptic plasticity dependent on extrasynaptic NMDAR activation, and on a tonic current (TC) resulting from the activation of extrasynaptic NMDAR by ambient glutamate in hippocampal slices from young (3-6-month-old) and aged (24-28-month-old) Sprague-Dawley rats. Aßo significantly enhanced the magnitude of LTD and the amplitude of TC in aged slices compared to young ones. TBOA, a glutamate transporter inhibitor, also significantly increased LTD magnitude and TC amplitude in slices from aged rats, suggesting either an age-related weakness of the glutamate clearance system and/or a facilitated extrasynaptic NMDAR activation. From our present data, we hypothesize that senescence-related impairment of the extrasynaptic environment may be a vector of vulnerability of the aged hippocampus to neurodegenerative promotors such as Aßo.


Aging , Amyloid beta-Peptides/metabolism , Excitatory Postsynaptic Potentials/physiology , Hippocampus/metabolism , Long-Term Potentiation/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Amino Acid Transport System X-AG/metabolism , Animals , Neuronal Plasticity/physiology , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/metabolism
6.
Neuroscience ; 396: 175-186, 2019 01 01.
Article En | MEDLINE | ID: mdl-30472430

Significant alterations in glutamatergic neurotransmission have been reported in major depressive disorder (MDD) that could underlie psychiatric traits. Studies were mainly interested in synaptic dysfunction in the prefrontal cortex, a key structure involved in depressive-like behavior, however hippocampus has been shown to be important in MDD. As cognitive deficits such as hippocampus-memory process were observed in MDD, we investigated in a mild hypoglutamatergic model behaviors related to depression and memory, synaptic transmission parameters and glutamatergic state specifically in the hippocampus. We thus characterized these phenotypes in adult male mice partially depleted in glutaminase type 1 or GLS1 (GLS1 HET), the enzyme responsible for glutamate synthesis in neurons, that we previously characterized as displaying moderate lower levels of glutamate in brain. We showed that GLS1 mutant mice display AMPA-R-mediated response deficits after prolonged repetitive stimulation with electrophysiological recording and inability to sustain glutamate release by microdialysis experiments with no consequences on behavioral spatial learning performances. However, their ability to escape from unpleasant but repeated escapable condition was attenuated whereas they were more immobile in the unescapable situation in the FST during re-test. These results show that GLS1 mutant mice display moderate impairments of hippocampal glutamatergic neurotransmission and moderate changes in adaptive behaviors that have been shown to participate to the development of depressive-like state.


Avoidance Learning/physiology , Glutamic Acid/physiology , Glutaminase/physiology , Hippocampus/physiology , Immobility Response, Tonic/physiology , Spatial Learning/physiology , Synaptic Transmission/physiology , Animals , Corticosterone/blood , Electric Stimulation , Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Glutaminase/genetics , Long-Term Potentiation/physiology , Male , Mice , Microdialysis , Mutation , Restraint, Physical/physiology
7.
EMBO Mol Med ; 10(11)2018 11.
Article En | MEDLINE | ID: mdl-30275019

Chromatin acetylation, a critical regulator of synaptic plasticity and memory processes, is thought to be altered in neurodegenerative diseases. Here, we demonstrate that spatial memory and plasticity (LTD, dendritic spine formation) deficits can be restored in a mouse model of tauopathy following treatment with CSP-TTK21, a small-molecule activator of CBP/p300 histone acetyltransferases (HAT). At the transcriptional level, CSP-TTK21 re-established half of the hippocampal transcriptome in learning mice, likely through increased expression of neuronal activity genes and memory enhancers. At the epigenomic level, the hippocampus of tauopathic mice showed a significant decrease in H2B but not H3K27 acetylation levels, both marks co-localizing at TSS and CBP enhancers. Importantly, CSP-TTK21 treatment increased H2B acetylation levels at decreased peaks, CBP enhancers, and TSS, including genes associated with plasticity and neuronal functions, overall providing a 95% rescue of the H2B acetylome in tauopathic mice. This study is the first to provide in vivo proof-of-concept evidence that CBP/p300 HAT activation efficiently reverses epigenetic, transcriptional, synaptic plasticity, and behavioral deficits associated with Alzheimer's disease lesions in mice.


Enzyme Activators/pharmacology , Memory , Neuronal Plasticity/drug effects , Tauopathies/physiopathology , p300-CBP Transcription Factors/metabolism , Acetylation/drug effects , Animals , Disease Models, Animal , Epigenesis, Genetic/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Histones/metabolism , Inflammation/pathology , Memory/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Tauopathies/genetics , Transcriptome/drug effects , Transcriptome/genetics , Transgenes
8.
Acta Neuropathol ; 135(6): 839-854, 2018 06.
Article En | MEDLINE | ID: mdl-29696365

Alzheimer's disease (AD) is associated with a progressive loss of synapses and neurons. Studies in animal models indicate that morphological alterations of dendritic spines precede synapse loss, increasing the proportion of large and short ("stubby") spines. Whether similar alterations occur in human patients, and what their functional consequences could be, is not known. We analyzed biopsies from AD patients and APP x presenilin 1 knock-in mice that were previously shown to present a loss of pyramidal neurons in the CA1 area of the hippocampus. We observed that the proportion of stubby spines and the width of spine necks are inversely correlated with synapse density in frontal cortical biopsies from non-AD and AD patients. In mice, the reduction in the density of synapses in the stratum radiatum was preceded by an alteration of spine morphology, with a reduction of their length and an enlargement of their neck. Serial sectioning examined with electron microscopy allowed us to precisely measure spine parameters. Mathematical modeling indicated that the shortening and widening of the necks should alter the electrical compartmentalization of the spines, leading to reduced postsynaptic potentials in spine heads, but not in soma. Accordingly, there was no alteration in basal synaptic transmission, but long-term potentiation and spatial memory were impaired. These results indicate that an alteration of spine morphology could be involved in the early cognitive deficits associated with AD.


Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Dendritic Spines/pathology , Dendritic Spines/physiology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Computer Simulation , Disease Models, Animal , Female , Frontal Lobe/pathology , Frontal Lobe/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Imaging, Three-Dimensional , Male , Membrane Potentials/physiology , Mice, Transgenic , Microscopy, Electron , Middle Aged , Models, Neurological , Presenilin-1/genetics , Presenilin-1/metabolism , Synapses/pathology , Tissue Culture Techniques
9.
Cereb Cortex ; 28(11): 3976-3993, 2018 11 01.
Article En | MEDLINE | ID: mdl-29048465

The treatment of Alzheimer's disease (AD) remains challenging and requires a better in depth understanding of AD progression. Particularly, the link between amyloid protein precursor (APP) processing and Tau pathology development remains poorly understood. Growing evidences suggest that APP processing and amyloid-ß (Aß) release are upstream of Tau pathology but the lack of animal models mimicking the slow progression of human AD raised questions around this mechanism. Here, we described that an AD-like ßAPP processing in adults wild-type rats, yielding to human APP, ßCTF and Aß levels similar to those observed in AD patients, is sufficient to trigger gradual Tauopathy. The Tau hyperphosphorylation begins several months before the formation of both amyloid plaques and tangle-like aggregates in aged rats and without associated inflammation. Based on a longitudinal characterization over 30 months, we showed that extrasynaptic and emotional impairments appear before long-term potentiation deficits and memory decline and so before Aß and Tau aggregations. These compelling data allowed us to (1) experimentally confirm the causal relationship between ßAPP processing and Tau pathology in vivo and without Tau transgene overexpression, (2) support the amyloidogenic cascade and (3) propose a 4-step hypothesis of prodromal AD progression.


Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , tau Proteins/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Animals , Disease Progression , Female , Genetic Vectors , Humans , Long-Term Potentiation , Male , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism , Presenilin-1/genetics , Protein Aggregation, Pathological/metabolism , Rats, Wistar
10.
Curr Alzheimer Res ; 14(1): 30-39, 2017.
Article En | MEDLINE | ID: mdl-27539596

BACKGROUND: The early phase of Alzheimer`s disease (AD) involves the disruption of finely tuned neuronal circuitry in brain regions associated with learning and memory. This tuning is obtained from the delicate balance of excitatory and inhibitory inputs which regulate cortical network function. This homeostatic plasticity provides a dynamic basis for appropriate information transfer in the brain. Excitatory synaptic transmission is driven mainly by glutamatergic synapses whereas inhibitory synaptic transmission involves GABAergic and glycinergic signaling. GABAergic cells, responsible for inhibitory transmission in adult brain, have recently become the subject of study in AD research. The discovery that GABAergic interneurons are targets of the amyloid-beta (Aß) peptide suggest that deregulation of the excitatory/inhibitory balance contributes to changes in cortical regulation, possibly with consequences for the development of the pathology. Thus, understanding the molecular details involved in GABAergic alterations may provide insight into the pathogenesis of AD. OBJECTIVE: Here, we review recent discoveries illustrating the concept of early alterations to the inhibitory circuits in AD and consider their functional implications for GABAergic components at membrane, cellular and microcircuit levels. CONCLUSION: We look at approaches that may lead to new hypotheses, animal models and therapeutic strategies based on GABAergic cells in AD with particular interest in microcircuits.


Alzheimer Disease/metabolism , Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Humans , Neurons/drug effects , Neurons/pathology
11.
Mol Neurodegener ; 11: 5, 2016 Jan 12.
Article En | MEDLINE | ID: mdl-26759118

BACKGROUND: Alzheimer's disease (AD) is the most frequent form of dementia in the elderly and no effective treatment is currently available. The mechanisms triggering AD onset and progression are still imperfectly dissected. We aimed at deciphering the modifications occurring in vivo during the very early stages of AD, before the development of amyloid deposits, neurofibrillary tangles, neuronal death and inflammation. Most current AD models based on Amyloid Precursor Protein (APP) overproduction beginning from in utero, to rapidly reproduce the histological and behavioral features of the disease within a few months, are not appropriate to study the early steps of AD development. As a means to mimic in vivo amyloid APP processing closer to the human situation in AD, we used an adeno-associated virus (AAV)-based transfer of human mutant APP and Presenilin 1 (PS1) genes to the hippocampi of two-month-old C57Bl/6 J mice to express human APP, without significant overexpression and to specifically induce its amyloid processing. RESULTS: The human APP, ßCTF and Aß42/40 ratio were similar to those in hippocampal tissues from AD patients. Three months after injection the murine Tau protein was hyperphosphorylated and rapid synaptic failure occurred characterized by decreased levels of both PSD-95 and metabolites related to neuromodulation, on proton magnetic resonance spectroscopy ((1)H-MRS). Astrocytic GLT-1 transporter levels were lower and the tonic glutamatergic current was stronger on electrophysiological recordings of CA1 hippocampal region, revealing the overstimulation of extrasynaptic N-methyl D-aspartate receptor (NMDAR) which precedes the loss of long-term potentiation (LTP). These modifications were associated with early behavioral impairments in the Open-field, Y-maze and Morris Mater Maze tasks. CONCLUSIONS: Altogether, this demonstrates that an AD-like APP processing, yielding to levels of APP, ßCTF and Aß42/Aß40 ratio similar to those observed in AD patients, are sufficient to rapidly trigger early steps of the amyloidogenic and Tau pathways in vivo. With this strategy, we identified a sequence of early events likely to account for disease onset and described a model that may facilitate efforts to decipher the factors triggering AD and to evaluate early neuroprotective strategies.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Hippocampus/metabolism , Aged , Aged, 80 and over , Aging , Animals , Disease Models, Animal , Disease Progression , Female , Humans , Long-Term Potentiation/physiology , Male , Mice, Inbred C57BL , Plaque, Amyloid/metabolism , tau Proteins/metabolism
12.
Sci Rep ; 5: 15862, 2015 Oct 29.
Article En | MEDLINE | ID: mdl-26511387

Chemokines are signaling molecules playing an important role in immune regulations. They are also thought to regulate brain development, neurogenesis and neuroendocrine functions. While chemokine upsurge has been associated with conditions characterized with cognitive impairments, their ability to modulate synaptic plasticity remains ill-defined. In the present study, we specifically evaluated the effects of MIP1-α/CCL3 towards hippocampal synaptic transmission, plasticity and spatial memory. We found that CCL3 (50 ng/ml) significantly reduced basal synaptic transmission at the Schaffer collateral-CA1 synapse without affecting NMDAR-mediated field potentials. This effect was ascribed to post-synaptic regulations, as CCL3 did not impact paired-pulse facilitation. While CCL3 did not modulate long-term depression (LTD), it significantly impaired long-term potentiation (LTP), an effect abolished by Maraviroc, a CCR5 specific antagonist. In addition, sub-chronic intracerebroventricular (icv) injections of CCL3 also impair LTP. In accordance with these electrophysiological findings, we demonstrated that the icv injection of CCL3 in mouse significantly impaired spatial memory abilities and long-term memory measured using the two-step Y-maze and passive avoidance tasks. These effects of CCL3 on memory were inhibited by Maraviroc. Altogether, these data suggest that the chemokine CCL3 is an hippocampal neuromodulator able to regulate synaptic plasticity mechanisms involved in learning and memory functions.


Chemokine CCL3/pharmacology , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Long-Term Synaptic Depression/drug effects , Memory/drug effects , Neurotransmitter Agents/pharmacology , Synaptic Transmission/drug effects , Animals , Chemokine CCL3/metabolism , Male , Mice , Neurotransmitter Agents/metabolism
13.
Hum Mol Genet ; 24(21): 5965-76, 2015 Nov 01.
Article En | MEDLINE | ID: mdl-26358780

Alzheimer's disease (AD) is characterized by both amyloid and Tau pathologies. The amyloid component and altered cholesterol metabolism are closely linked, but the relationship between Tau pathology and cholesterol is currently unclear. Brain cholesterol is synthesized in situ and cannot cross the blood-brain barrier: to be exported from the central nervous system into the blood circuit, excess cholesterol must be converted to 24S-hydroxycholesterol by the cholesterol 24-hydroxylase encoded by the CYP46A1 gene. In AD patients, the concentration of 24S-hydroxycholesterol in the plasma and the cerebrospinal fluid are lower than in healthy controls. The THY-Tau22 mouse is a model of AD-like Tau pathology without amyloid pathology. We used this model to investigate the potential association between Tau pathology and CYP46A1 modulation. The amounts of CYP46A1 and 24S-hydroxycholesterol in the hippocampus were lower in THY-Tau22 than control mice. We used an adeno-associated virus (AAV) gene transfer strategy to increase CYP46A1 expression in order to investigate the consequences on THY-Tau22 mouse phenotype. Injection of the AAV-CYP46A1 vector into the hippocampus of THY-Tau22 mice led to CYP46A1 and 24S-hydroxycholesterol content normalization. The cognitive deficits, impaired long-term depression and spine defects that characterize the THY-Tau22 model were completely rescued, whereas Tau hyperphosphorylation and associated gliosis were unaffected. These results argue for a causal link between CYP46A1 protein content and memory impairments that result from Tau pathology. Therefore, CYP46A1 may be a relevant therapeutic target for Tauopathies and especially for AD.


Memory Disorders/enzymology , Steroid Hydroxylases/metabolism , Tauopathies/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cholesterol 24-Hydroxylase , Disease Models, Animal , Gliosis/metabolism , Hippocampus/enzymology , Humans , Hydroxycholesterols/metabolism , Memory Disorders/genetics , Mice , Mice, Transgenic , Phosphorylation , Steroid Hydroxylases/genetics , Tauopathies/genetics , tau Proteins
14.
J Alzheimers Dis ; 48(4): 927-35, 2015.
Article En | MEDLINE | ID: mdl-26402095

This study shows a decrease in soluble amyloid-ß protein precursor-α (sAßPPα) levels, but no change in sAßPPß, in the rat hippocampus during healthy aging, associated with the weaker expression of N-methyl-D-aspartate receptor (NMDAR)-dependent long-term potentiation (LTP) in the CA1 area of hippocampal slices. Exogenous application of recombinant sAßPPα increases NMDAR activation in aged animals and could rescue the age-related LTP deficits described. In contrast, it does not affect basal synaptic transmission or glutamate release. These results indicate that improving synaptic sAßPPα availability at synapses helps in reducing the functional NMDAR-related deregulation of hippocampal networks linked to aging.


Aging/physiology , Amyloid beta-Protein Precursor/metabolism , Hippocampus/physiology , Long-Term Potentiation/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/administration & dosage , Animals , Blotting, Western , Electric Stimulation , Electrophoresis, Polyacrylamide Gel , Excitatory Postsynaptic Potentials/physiology , Microelectrodes , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Tissue Culture Techniques
15.
Aging Cell ; 12(1): 76-84, 2013 Feb.
Article En | MEDLINE | ID: mdl-23113887

Epidemiological data suggest that a poor ω3 status favoured by the low ω3/ω6 polyunsaturated fatty acids ratio in western diets contributes to cognitive decline in the elderly, but mechanistic evidence is lacking. We therefore explored the impact of ω3 deficiency on the evolution of glutamatergic transmission in the CA1 of the hippocampus during aging by comparing 4 groups of rats aged 6-22 months fed ω3-deficient or ω3/ω6-balanced diets from conception to sacrifice: Young ω3 Balanced (YB) or Deficient (YD), Old ω3 Balanced (OB) or Deficient (OD) rats. ω3 Deficiency induced a 65% decrease in the amount of docosahexaenoic acid (DHA, the main ω3 in cell membranes) in brain phospholipids, but had no impact on glutamatergic transmission and astroglial function in young rats. Aging induced a 10% decrease in brain DHA, a 35% reduction of synaptic efficacy (fEPSP/PFV) due to decreased presynaptic glutamate release and a 30% decrease in the astroglial glutamate uptake associated with a marked astrogliosis (+100% GFAP). The ω3 deficiency further decreased these hallmarks of aging (OD vs. OB rats: -35% fEPSP/PFV P < 0.05, -15% astroglial glutamate uptake P < 0.001, +30% GFAP P < 0.01). This cannot be attributed to aggravation of the brain DHA deficit because the brains of OD rats had more DHA than those of YD rats. Thus, ω3 deficiency worsens the age-induced degradation of glutamatergic transmission and its associated astroglial regulation in the hippocampus.


Astrocytes/metabolism , CA1 Region, Hippocampal/metabolism , Fatty Acids, Omega-3/metabolism , Glutamic Acid/metabolism , Synapses/metabolism , Animals , Astrocytes/cytology , CA1 Region, Hippocampal/cytology , Cellular Senescence/physiology , Fatty Acids, Omega-3/administration & dosage , Female , Male , Rats , Rats, Wistar
16.
Article En | MEDLINE | ID: mdl-23230430

Somatostatin is highly expressed in mammalian brain and is involved in many brain functions such as motor activity, sleep, sensory, and cognitive processes. Five somatostatin receptors have been described: sst(1), sst(2) (A and B), sst(3), sst(4), and sst(5), all belonging to the G-protein-coupled receptor family. During the recent years, numerous studies contributed to clarify the role of somatostatin systems, especially long-range somatostatinergic interneurons, in several functions they have been previously involved in. New advances have also been made on the alterations of somatostatinergic systems in several brain diseases and on the potential therapeutic target they represent in these pathologies.

17.
J Alzheimers Dis ; 32(1): 183-96, 2012.
Article En | MEDLINE | ID: mdl-22785392

Alzheimer's disease is characterized by the loss of memory and synaptic damage. Evidence is accumulating for a causal role of soluble oligomeric species of amyloid-ß peptide (Aßo) in the impairment of synaptic plasticity and cognition but the precise mechanisms underlying these effects are still not clear. Synaptic plasticity such as long-term potentiation is thought to underlie learning and memory. While the effect of Aß on long-term potentiation is well documented, a more general understanding of Aß action on various aspects of plasticity involving synaptic and extrasynaptic receptors and the nature of the mechanisms involved in its effects are lacking. Using a combination of electrophysiological and biochemical techniques in mouse hippocampal slices, we show here that Aßo drastically affects synaptic plasticities induced by high stimulation frequencies through the involvement of extrasynaptic glutamate receptors. Experiments on hippocampal slices as well as on cultured cortical neurons show that Aßo potentiates extrasynaptic NMDA receptors-mediated responses. Pharmacological characterization indicates that GluN2B-containing NMDARs are involved in these responses. When synaptic and extrasynaptic glutamate receptor-mediated effects are dissociated using cortical neurons in culture, it appears that Aßo has differential effects on these two receptors types. We conclude that the pool of extrasynaptic GluN2B-containing NMDARs is a major target of Aßo in the hippocampus. During high frequency stimulation, Aßo dramatically impairs long-term neuronal responses.


Amyloid beta-Peptides/toxicity , Hippocampus/pathology , Neuronal Plasticity/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Synapses/drug effects , Amyloid beta-Peptides/chemistry , Animals , Calcium Signaling/physiology , Cells, Cultured , Data Interpretation, Statistical , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Long-Term Potentiation/drug effects , Mice , Mice, Inbred C57BL , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/physiology , Synaptic Transmission/physiology
18.
Aging Cell ; 11(2): 336-44, 2012 Apr.
Article En | MEDLINE | ID: mdl-22230264

Oxidative stress (OS) resulting from an imbalance between antioxidant defenses and the intracellular accumulation of reactive oxygen species (ROS) contributes to age-related memory deficits. While impaired synaptic plasticity in neuronal networks is thought to underlie cognitive deficits during aging, whether this process is targeted by OS and what the mechanisms involved are still remain open questions. In this study, we investigated the age-related effects of the reducing agent N-acetyl-L-cysteine (L-NAC) on the activation of the N-methyl-D-aspartate receptor (NMDA-R) by its co-agonist D-serine, because alterations in this mechanism contribute greatly to synaptic plasticity deficits in aged animals. Long-term dietary supplementation with L-NAC prevented oxidative damage in the hippocampus of aged rats. Electrophysiological recordings in the CA1 of hippocampal slices indicated that NMDA-R-mediated synaptic potentials and theta-burst-induced long-term potentiation (LTP) were depressed in aged animals, deficits that could be reversed by exogenous D-serine. Chronic treatment with L-NAC, but not acute application of the reducing agent, restored potent D-serine-dependent NMDA-R activation and LTP induction in aged rats. In addition, it is also revealed that the age-related decrease in D-serine levels and in the expression of the synthesizing enzyme serine racemase, which underlies the decrease in NMDA-R activation by the amino acid, was rescued by long-term dietary treatment with L-NAC. Our results indicate that protecting redox status in aged animals could prevent injury to the cellular mechanisms underlying cognitive aging, in part by maintaining potent NMDA-R activation through the D-serine-dependent pathway.


Aging , Hippocampus/metabolism , Neuronal Plasticity , Oxidative Stress , Receptors, N-Methyl-D-Aspartate/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Lysine/analogs & derivatives , Lysine/pharmacology , Male , Neuronal Plasticity/drug effects , Rats , Rats, Wistar , Serine/metabolism
19.
Neurobiol Aging ; 33(6): 1126.e1-14, 2012 Jun.
Article En | MEDLINE | ID: mdl-22206845

In Alzheimer's disease, amyloid beta peptide (Aß) accumulation is associated with hippocampal network dysfunction. Intrahippocampal injections of Aß induce aberrant inhibitory septohippocampal (SH) network activity in vivo and impairment of memory processing. In the present study, we observed, after hippocampal Aß treatment, a selective loss of neurons projecting to the medial septum (MS) and containing calbindin (CB) and/or somatostatin (SOM). Other GABAergic neuronal subpopulations were not altered. Thus, the present study identifies hippocamposeptal neuron populations as specific targets for Aß deposits. We observed that in Aß-treated rats but not in controls, glutamate agonist application induced rhythmic bursting in 55% of the slow-firing neurons in the medial septum. This suggests that hippocampal Aß can trigger modifications of the septohippocampal pathway via the alteration of a specific neuronal population. Long-range hippocamposeptal GABA/calbindin neurons, targets of hippocampal amyloid deposits, are implicated in supporting network synchronization. By identifying this target, we contribute to the understanding of the mechanisms underlying deleterious effects of Aß, one of the main agents of dementia in Alzheimer's disease.


Amyloid beta-Peptides/toxicity , Hippocampus/pathology , Neurons/pathology , Action Potentials/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/administration & dosage , Animals , Hippocampus/metabolism , Hippocampus/physiology , Male , Neural Pathways/metabolism , Neural Pathways/pathology , Neural Pathways/physiology , Neurons/physiology , Rats , Rats, Sprague-Dawley
20.
Mech Ageing Dev ; 132(5): 240-8, 2011 May.
Article En | MEDLINE | ID: mdl-21530571

INTRODUCTION: Effects of 3-month continuous environmental enrichment (EE) on cognitive abilities and on theta burst-related synaptic plasticity of CA1 hippocampal neuronal networks have been assessed in 6- and 20-month old NMRI female mice. RESULTS: EE decreased anxiety-like behavior and improved learning and memory performances in adult but not in aged mice. Electrophysiological results in CA1 hippocampal slices showed that basal synaptic transmission was not affected by EE in adult mice whereas it was partially improved in aged animals, even though not sufficient to rescue the decrease related to aging. Besides, no effect of EE on N-methyl-d-aspartate receptor activation and theta-burst-induced long-term potentiation was found in adult or aged animals. DISCUSSION: These results indicate that continuous EE is able to improve cognitive abilities in adult NMRI female mice, that does not correlate with changes in theta burst-related synaptic plasticity within neuronal networks. In addition, the lack of effects in aged animals suggests the existence of a critical delay for the beneficial effects of EE on cognitive aging.


Aging/physiology , Hippocampus/physiology , Learning/physiology , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Animals , Female , Hippocampus/cytology , Mice
...