Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Neurobiol Aging ; 53: 112-121, 2017 05.
Article in English | MEDLINE | ID: mdl-28254590

ABSTRACT

Aging is accompanied by increased neuroinflammation, synaptic dysfunction, and cognitive deficits both in rodents and humans, yet the onset and progression of these deficits throughout the life span remain unknown. These aging-related deficits affect the quality of life and present challenges to our aging society. Here, we defined age-dependent and progressive impairments of synaptic and cognitive functions and showed that reducing astrocyte-related neuroinflammation through anti-inflammatory drug treatment in aged mice reverses these events. By comparing young (3 months), middle-aged (18 months), aged (24 months), and advanced-aged wild-type mice (30 months), we found that the levels of an astrocytic marker, glial fibrillary acidic protein, progressively increased after 18 months of age, which preceded the decreases of the synaptic marker PSD-95. Hippocampal long-term potentiation was also suppressed in an age-dependent manner, where significant deficits were observed after 24 months of age. Fear conditioning tests demonstrated that associative memory in the context and cued conditions was decreased starting at the ages of 18 and 30 months, respectively. When the mice were tested on hidden platform water maze, spatial learning memory was significantly impaired after 24 months of age. Importantly, subacute treatment with the anti-inflammatory drug ibuprofen suppressed astrocyte activation and restored synaptic plasticity and memory function in advanced-aged mice. These results support the critical contribution of aging-related inflammatory responses to hippocampal-dependent cognitive function and synaptic plasticity, in particular during advanced aging. Our findings provide strong evidence that suppression of neuroinflammation could be a promising treatment strategy to preserve cognition during aging.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cognition/drug effects , Cognition/physiology , Cognitive Aging/physiology , Cognitive Aging/psychology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Ibuprofen/pharmacology , Ibuprofen/therapeutic use , Neuronal Plasticity/drug effects , Animals , Astrocytes/pathology , Biomarkers/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/psychology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/physiopathology , Humans , Inflammation/drug therapy , Inflammation/pathology , Inflammation/physiopathology , Long-Term Potentiation , Male , Mice , Molecular Targeted Therapy , Spatial Learning/drug effects , Spatial Learning/physiology
2.
Eur J Neurosci ; 41(10): 1372-80, 2015 May.
Article in English | MEDLINE | ID: mdl-25864922

ABSTRACT

The Reelin signaling pathway is implicated in processes controlling synaptic plasticity and hippocampus-dependent learning and memory. A single direct in vivo application of Reelin enhances long-term potentiation, increases dendritic spine density and improves associative and spatial learning and memory. Angelman syndrome (AS) is a neurological disorder that presents with an overall defect in synaptic function, including decreased long-term potentiation, reduced dendritic spine density, and deficits in learning and memory, making it an attractive model in which to examine the ability of Reelin to recover synaptic function and cognitive deficits. In this study, we investigated the effects of Reelin administration on synaptic plasticity and cognitive function in a mouse model of AS and demonstrated that bilateral, intraventricular injections of Reelin recover synaptic function and corresponding hippocampus-dependent associative and spatial learning and memory. Additionally, we describe alteration of the Reelin profile in tissue from both the AS mouse and post-mortem human brain.


Subject(s)
Angelman Syndrome/physiopathology , Angelman Syndrome/psychology , Cell Adhesion Molecules, Neuronal/administration & dosage , Extracellular Matrix Proteins/administration & dosage , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Nerve Tissue Proteins/administration & dosage , Serine Endopeptidases/administration & dosage , Angelman Syndrome/drug therapy , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/metabolism , Dendritic Spines/drug effects , Disease Models, Animal , Extracellular Matrix Proteins/metabolism , Female , HEK293 Cells , Hippocampus/physiopathology , Hippocampus/ultrastructure , Humans , Injections, Intraventricular , Male , Mice , Motor Activity/drug effects , Nerve Tissue Proteins/metabolism , Reelin Protein , Serine Endopeptidases/metabolism , Spatial Learning/drug effects , Spatial Memory/drug effects
3.
J Psychopharmacol ; 27(4): 386-95, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23104248

ABSTRACT

The lipoprotein receptor ligand Reelin is important for the processes of normal synaptic plasticity, dendritic morphogenesis, and learning and memory. Heterozygous reeler mice (HRM) show many neuroanatomical, biochemical, and behavioral features that are associated with schizophrenia. HRM show subtle morphological defects including reductions in dendritic spine density, altered synaptic plasticity and behavioral deficits in associative learning and memory and pre-pulse inhibition. The present studies test the hypothesis that in vivo elevation of Reelin levels can rescue synaptic and behavioral phenotypes associated with HRM. We demonstrate that a single in vivo injection of Reelin increases GAD67 expression and alters dendritic spine morphology. In parallel we observed enhancement of hippocampal synaptic function and associative learning and memory. Reelin supplementation also increases pre-pulse inhibition. These results suggest that characteristics of HRM, similar to those observed in schizophrenia, are sensitive to Reelin levels and can be modified with Reelin supplementation in male and female adults.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Gait Disorders, Neurologic/metabolism , Learning Disabilities/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity , Schizophrenia/metabolism , Serine Endopeptidases/metabolism , Animals , Cell Adhesion Molecules, Neuronal/genetics , Crosses, Genetic , Dendritic Spines/metabolism , Dendritic Spines/pathology , Extracellular Matrix Proteins/genetics , Female , Gait Disorders, Neurologic/etiology , Glutamate Decarboxylase/metabolism , Heterozygote , Hippocampus/metabolism , Learning , Learning Disabilities/etiology , Male , Mice , Mice, Neurologic Mutants , Nerve Tissue Proteins/genetics , Neural Inhibition , Neurons/metabolism , Reelin Protein , Schizophrenia/pathology , Schizophrenia/physiopathology , Sensory Gating , Serine Endopeptidases/genetics , Synaptic Transmission
4.
J Neurosci ; 31(45): 16241-50, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22072675

ABSTRACT

The protective/neurotoxic role of fractalkine (CX3CL1) and its receptor CX3C chemokine receptor 1 (CX3CR1) signaling in neurodegenerative disease is an intricate and highly debated research topic and it is becoming even more complicated as new studies reveal discordant results. It appears that the CX3CL1/CX3CR1 axis plays a direct role in neurodegeneration and/or neuroprotection depending on the CNS insult. However, all the above studies focused on the role of CX3CL1/CX3CR1 signaling in pathological conditions, ignoring the relevance of CX3CL1/CX3CR1 signaling under physiological conditions. No approach to date has been taken to decipher the significance of defects in CX3CL1/CX3CR1 signaling in physiological condition. In the present study we used CX3CR1⁻/⁻, CX3CR1⁺/⁻, and wild-type mice to investigate the physiological role of CX3CR1 receptor in cognition and synaptic plasticity. Our results demonstrate for the first time that mice lacking the CX3CR1 receptor show contextual fear conditioning and Morris water maze deficits. CX3CR1 deficiency also affects motor learning. Importantly, mice lacking the receptor have a significant impairment in long-term potentiation (LTP). Infusion with IL-1ß receptor antagonist significantly reversed the deficit in cognitive function and impairment in LTP. Our results reveal that under physiological conditions, disruption in CX3CL1 signaling will lead to impairment in cognitive function and synaptic plasticity via increased action of IL-1ß.


Subject(s)
Cognition Disorders/pathology , Hippocampus/pathology , Hippocampus/physiopathology , Long-Term Potentiation/genetics , Receptors, Interleukin-8A/deficiency , Analysis of Variance , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Biophysics , Bromodeoxyuridine/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cerebellum/metabolism , Cognition Disorders/genetics , Conditioning, Psychological/physiology , Cytokines/metabolism , Disease Models, Animal , Electric Stimulation , Enzyme-Linked Immunosorbent Assay/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Exploratory Behavior/physiology , Fear/physiology , Gene Expression Regulation/genetics , In Vitro Techniques , Interleukin 1 Receptor Antagonist Protein/pharmacology , Long-Term Potentiation/physiology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microglia/pathology , Motor Activity/genetics , Neurogenesis/genetics , Patch-Clamp Techniques , Rotarod Performance Test
5.
Learn Mem ; 18(9): 558-64, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21852430

ABSTRACT

Apolipoprotein receptors belong to an evolutionarily conserved surface receptor family that has intimate roles in the modulation of synaptic plasticity and is necessary for proper hippocampal-dependent memory formation. The known lipoprotein receptor ligand Reelin is important for normal synaptic plasticity, dendritic morphology, and cognitive function; however, the in vivo effect of enhanced Reelin signaling on cognitive function and synaptic plasticity in wild-type mice is unknown. The present studies test the hypothesis that in vivo enhancement of Reelin signaling can alter synaptic plasticity and ultimately influence processes of learning and memory. Purified recombinant Reelin was injected bilaterally into the ventricles of wild-type mice. We demonstrate that a single in vivo injection of Reelin increased activation of adaptor protein Disabled-1 and cAMP-response element binding protein after 15 min. These changes correlated with increased dendritic spine density, increased hippocampal CA1 long-term potentiation (LTP), and enhanced performance in associative and spatial learning and memory. The present study suggests that an acute elevation of in vivo Reelin can have long-term effects on synaptic function and cognitive ability in wild-type mice.


Subject(s)
Brain/cytology , Cell Adhesion Molecules, Neuronal/pharmacology , Cognition/drug effects , Dendritic Spines/drug effects , Extracellular Matrix Proteins/pharmacology , Nerve Tissue Proteins/pharmacology , Neuronal Plasticity/drug effects , Neurons/ultrastructure , Serine Endopeptidases/pharmacology , Action Potentials/drug effects , Animals , CREB-Binding Protein/metabolism , Conditioning, Psychological/drug effects , Dendritic Spines/ultrastructure , Excitatory Postsynaptic Potentials/drug effects , Exploratory Behavior/drug effects , Fear/drug effects , Fear/psychology , HEK293 Cells/cytology , Humans , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Reelin Protein , Silver Staining/methods
6.
Hum Mol Genet ; 20(5): 1000-7, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21159798

ABSTRACT

Hippocampal neurogenesis is the lifelong production of new neurons in the central nervous system (CNS), and affects many physiological and pathophysiological conditions, including neurobehavioral disorders. The early postnatal stage is the most prominent neurogenesis period; however, the functional role of neurogenesis in this developing stage has not been well characterized. To understand the role of hippocampal neurogenesis in the postnatal developing period, we analyzed secretin, a neuropeptide, which is expressed significantly higher in the development stage. Secretin is a pleiotropic neuropeptide hormone that belongs to the secretin/VIP/glucagon peptide family. Although secretin was originally isolated in the gastrointestinal system, it has been found that secretin itself acts as a neuropeptide in the CNS. Here, we report a new function of secretin as a survival factor for neural progenitor cells in the hippocampus. We found that secretin-deficient mice exhibit decreased numbers of BrdU-labeled new neurons and dramatically increased apoptosis of doublecortin-positive neural progenitor cells in the subgranular zone of the dentate gyrus (DG) during the early postnatal period. Furthermore, we found that reduced survival of neural progenitor cells leads to decreased volume of DG, reduced long-term potentiation and impaired spatial learning ability in adults. Our studies demonstrate that secretin has important implications for neurogenesis in postnatal development, and affects neurobehavioral function in the adult mouse.


Subject(s)
Neurons/cytology , Neurons/metabolism , Secretin/deficiency , Stem Cells/cytology , Stem Cells/metabolism , Animals , Apoptosis , Cell Survival , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Secretin/genetics
7.
Neuron Glia Biol ; 4(3): 259-70, 2008 Aug.
Article in English | MEDLINE | ID: mdl-19674510

ABSTRACT

Low-density-lipoprotein receptors (LDLRs) are an evolutionarily ancient surface protein family with the ability to activate a diversity of extracellular signals across the cellular membrane in the adult central nervous system (CNS). Their intimate roles in modulating synaptic plasticity and their necessity in hippocampal-dependent learning and memory have only recently come to light. Two known LDLR ligands, specifically apolipoprotein E (apoE) and reelin, have been the most widely investigated in this regard. Most of our understanding of synaptic plasticity comes from investigation of both pre- and postsynaptic alterations. Therefore, it is interesting to note that neurons and glia that do not contribute to the synaptic junction in question can secrete signaling molecules that affect synaptic plasticity. Notably, reelin and apoE have been shown to modulate hippocampal long-term potentiation in general, and affect NMDA receptor and AMPA receptor regulation specifically. Furthermore, these receptors and signaling molecules have significant roles in neuronal degenerative diseases such as Alzheimer's disease. The recent production of recombinant proteins, knockout and transgenic mice for receptors and ligands and the development of human ApoE targeted replacement mice have significantly expanded our understanding of the roles LDLRs and their ligands have in certain disease states and the accompanying initiation of specific signaling pathways. This review describes the role LDLRs, apoE and reelin have in the regulation of hippocampal synaptic plasticity.


Subject(s)
Apolipoproteins E/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Memory/physiology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Serine Endopeptidases/metabolism , Signal Transduction/physiology , Synapses/physiology , Animals , Apolipoproteins E/genetics , Brain/cytology , Brain/embryology , Brain/growth & development , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Humans , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Models, Biological , Nerve Tissue Proteins/genetics , Neuronal Plasticity/physiology , Neurons/cytology , Reelin Protein , Serine Endopeptidases/genetics
8.
Brain Res ; 1151: 20-31, 2007 Jun 02.
Article in English | MEDLINE | ID: mdl-17433272

ABSTRACT

Excessive glutamate (Glu) stimulation of the NMDA-R is a widely recognized trigger for Ca(2+)-mediated excitotoxicity. Primary neurons typically show a large increase in vulnerability to excitotoxicity with increasing days in vitro (DIV). This enhanced vulnerability has been associated with increased expression of the NR2B subunit or increased NMDA-R current, but the detailed age-courses of these variables in primary hippocampal neurons have not been compared in the same study. Further, it is not clear whether the NMDA-R is the only source of excess Ca(2+). Here, we used primary hippocampal neurons to examine the age dependence of the increase in excitotoxic vulnerability with changes in NMDA-R current, and subunit expression. We also tested whether L-type voltage-gated Ca(2+) channels (L-VGCCs) contribute to the enhanced vulnerability. The EC(50) for Glu toxicity decreased by approximately 10-fold between 8-9 and 14-15 DIV, changing little thereafter. Parallel experiments found that during the same period both amplitude and duration of NMDA-R current increased dramatically; this was associated with an increase in protein expression of the NR1 and NR2A subunits, but not of the NR2B subunit. Compared to MK-801, ifenprodil, a selective NR2B antagonist, was less effective in protecting older than younger neurons from Glu insult. Conversely, nimodipine, an L-VGCC antagonist, protected older but not younger neurons. Our results indicate that enhanced excitotoxic vulnerability with age in culture was associated with a substantial increase in NMDA-R current, concomitant increases in NR2A and NR1 but not NR2B subunit expression, and with apparent recruitment of L-VGCCs into the excitotoxic process.


Subject(s)
Aging/physiology , Hippocampus/cytology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calcium/metabolism , Calcium Channels, L-Type , Cell Survival/drug effects , Cells, Cultured , Dizocilpine Maleate/pharmacology , Embryo, Mammalian , Female , Gene Expression/drug effects , Glutamic Acid/toxicity , L-Lactate Dehydrogenase/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , N-Methylaspartate/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Patch-Clamp Techniques/methods , Pregnancy , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...