Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Neurobiol Aging ; 36(1): 323-33, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25086838

ABSTRACT

Fischer 344 × Brown Norway F1 (F344 × BN-F1) hybrid rats express greater longevity with improved health relative to aging rodents of other strains; however, few behavioral reports have thoroughly evaluated cognition across the F344 × BN-F1 lifespan. Consequently, this study evaluated spatial reference memory in F344 × BN-F1 rats at 6, 18, 24, or 28 months of age in the Morris water maze. Reference memory decrements were observed between 6 and 18 months and 18 and 24 months. At 28 months, spatial learning was not worse than 24 months, but swim speed was significantly slower. Reliable individual differences revealed that ∼50% of 24- to 28-month-old rats performed similarly to 6 months, whereas others were spatial learning impaired. Aged rats were impaired at learning within daily training sessions but not impaired at retaining information between days of training. Aged rats were also slower to learn to escape onto the platform, regardless of strategy. In summary, these data clarify the trajectory of cognitive decline in aging F344 × BN-F1 rats and elucidate relevant behavioral parameters.


Subject(s)
Aging/psychology , Spatial Memory/physiology , Animals , Behavior, Animal/physiology , Cognition/physiology , Cues , Male , Models, Animal , Rats , Rats, Inbred F344 , Spatial Learning/physiology
2.
Neuropharmacology ; 70: 63-73, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23347951

ABSTRACT

Normal aging may limit the signaling efficacy of certain GPCRs by disturbing the function of specific Gα-subunits and leading to deficient modulation of intracellular functions that subserve synaptic plasticity, learning and memory. Evidence suggests that Gαq/11 is more sensitive to the effects of aging relative to other Gα-subunits, including Gαo. To test this hypothesis, the functionality of Gαq/11 and Gαo were compared in the hippocampus of young (6 months) and aged (24 months) F344 × BNF1 hybrid rats assessed for spatial learning ability. Basal GTPγS-binding to Gαq/11 was significantly elevated in aged rats relative to young and but not reliably associated with spatial learning. mAChR stimulation of Gαq/11 with oxotremorine-M produced equivocal GTPγS-binding between age groups although values tended to be lower in the aged hippocampus and were inversely related to basal activity. Downstream Gαq/11 function was measured in hippocampal subregion CA1 by determining changes in [Ca(2+)]i after mAChR and mGluR (DHPG) stimulation. mAChR-stimulated peak change in [Ca(2+)]i was lower in aged CA1 relative to young while mGluR-mediated integrated [Ca(2+)]i responses tended to be larger in aged. GPCR modulation of [Ca(2+)]i was observed to depend on intracellular stores to a greater degree in aged than young. In contrast, measures of Gαo-mediated GTPγS-binding were stable across age, including basal, mAChR-, GABABR (baclofen)-stimulated levels. Overall, the data indicate that aging selectively modulates the activity of Gαq/11 within the hippocampus leading to deficient modulation of [Ca(2+)]i following stimulation of mAChRs but these changes are not related to spatial learning.


Subject(s)
Aging/metabolism , Aging/psychology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Hippocampus/metabolism , Animals , Baclofen/pharmacology , CA1 Region, Hippocampal/metabolism , Calcium/metabolism , Excitatory Amino Acid Agonists/pharmacology , GABA-B Receptor Agonists/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Hippocampus/drug effects , Learning , Male , Muscarinic Agonists/pharmacology , Oxotremorine/analogs & derivatives , Oxotremorine/pharmacology , Rats , Resorcinols/pharmacology , Spatial Behavior
3.
Neurobiol Aging ; 33(6): 1124.e1-12, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22169202

ABSTRACT

Gamma aminobutyric acid (GABA)(B) receptors (GABA(B)Rs) have been linked to a wide range of physiological and cognitive processes and are of interest for treating a number of neurodegenerative and psychiatric disorders. As many of these diseases are associated with advanced age, it is important to understand how the normal aging process impacts GABA(B)R expression and signaling. Thus, we investigated GABA(B)R expression and function in the prefrontal cortex (PFC) and hippocampus of young and aged rats characterized in a spatial learning task. Baclofen-stimulated GTP-binding and GABA(B)R1 and GABA(B)R2 proteins were reduced in the prefrontal cortex of aged rats but these reductions were not associated with spatial learning abilities. In contrast, hippocampal GTP-binding was comparable between young and aged rats but reduced hippocampal GABA(B)R1 expression was observed in aged rats with spatial learning impairment. These data demonstrate marked regional differences in GABA(B)R complexes in the adult and aged brain and could have implications for both understanding the role of GABAergic processes in normal brain function and the development of putative interventions that target this system.


Subject(s)
Aging/metabolism , Guanosine Triphosphate/metabolism , Hippocampus/metabolism , Prefrontal Cortex/metabolism , Receptors, GABA-B/metabolism , Animals , Male , Maze Learning/physiology , Protein Binding/physiology , Rats , Rats, Inbred F344
4.
Behav Brain Res ; 227(1): 258-64, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22085876

ABSTRACT

Behavioral flexibility, the ability to modify responses due to changing task demands, is detrimentally affected by aging with a shift towards increased cognitive rigidity. The neurobiological basis of this cognitive deficit is not clear although striatal cholinergic neurotransmission has been implicated. To investigate the possible association between striatal acetylcholine signaling with age-related changes in behavioral flexibility, young, middle-aged, and aged F344 X Brown Norway F1 rats were assessed using an attentional set-shifting task that includes two tests of behavioral flexibility: reversal learning and an extra-dimensional shift. Rats were also assessed in the Morris water maze to compare potential fronto-striatal-dependent deficits with hippocampal-dependent deficits. Behaviorally characterized rats were then assessed for acetylcholine muscarinic signaling within the striatum using oxotremorine-M-stimulated [(35)S]GTPγS binding and [(3)H]AFDX-384 receptor binding autoradiography. The results showed that by old age, cognitive deficits were pronounced across cognitive domains, suggesting deterioration of both hippocampal and fronto-striatal regions. A significant decline in oxotremorine-M-stimulated [(35)S]GTPγS binding was limited to the dorsomedial striatum of aged rats when compared to young and middle-aged rats. There was no effect of age on striatal [(3)H]AFDX-384 receptor binding. These results suggest that a decrease in M2/M4 muscarinic receptor coupling is involved in the age-associated decline in behavioral flexibility.


Subject(s)
Aging/pathology , Cognition Disorders/pathology , Corpus Striatum/metabolism , GTP-Binding Proteins/metabolism , Receptor, Muscarinic M2/metabolism , Receptor, Muscarinic M4/metabolism , Animals , Attention , Autoradiography , Corpus Striatum/drug effects , Corpus Striatum/pathology , Food Deprivation/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Hippocampus/drug effects , Hippocampus/metabolism , Isotopes/pharmacokinetics , Male , Maze Learning/physiology , Parasympatholytics/pharmacokinetics , Pirenzepine/analogs & derivatives , Pirenzepine/pharmacokinetics , Protein Binding/drug effects , Rats , Rats, Inbred F344 , Reversal Learning/physiology , Set, Psychology
5.
J Gerontol A Biol Sci Med Sci ; 66(5): 521-33, 2011 May.
Article in English | MEDLINE | ID: mdl-21393424

ABSTRACT

Fish oil (FO) mediates a number of cardioprotective benefits in patients with cardiovascular disease. In the absence of cardiovascular disease, however, the effects of FO on cardiac structure and function are not clear. In addition, it is not known if an effective dosing strategy for attenuating age-related cardiac dysfunction is also effective at limiting cognitive dysfunction. Therefore, we determined if 4 months of FO supplementation in aged rats would lessen age-related cardiac dysfunction while concomitantly preventing the cognitive decline that is normally observed in this population. The results indicate that FO initiated late in life modifies diastolic function in a small but positive way by attenuating the age-related increases in filling pressure, posterior wall thickness, and interstitial collagen without mitigating age-related deficits in memory or increases in brain inflammation. These data raise the possibility that FO supplementation for purposes of cardiac and brain protection may need to occur earlier in the life span.


Subject(s)
Aging/physiology , Brain/pathology , Diastole/drug effects , Dietary Fats, Unsaturated/pharmacology , Fish Oils/pharmacology , Memory/drug effects , Aging/drug effects , Animals , Body Weight/drug effects , Brain/drug effects , Cell Count , Cognition/drug effects , Cognition/physiology , Diastole/physiology , Encephalitis , Fatty Acids, Omega-3/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Immunohistochemistry , Male , Motor Activity/drug effects , Motor Activity/physiology , Organ Size/drug effects , Rats , Vision, Ocular/drug effects , Vision, Ocular/physiology
6.
Brain Res ; 1385: 307-16, 2011 Apr 18.
Article in English | MEDLINE | ID: mdl-21338580

ABSTRACT

Fractionated partial or whole-brain irradiation (fWBI) is a widely used, effective treatment for primary and metastatic brain tumors, but it also produces radiation-induced brain injury, including cognitive impairment. Radiation-induced neural changes are particularly problematic for elderly brain tumor survivors who also experience age-dependent cognitive impairment. Accordingly, we investigated i] radiation-induced cognitive impairment, and ii] potential biomarkers of radiation-induced brain injury in a rat model of aging. Fischer 344 x Brown Norway rats received fractionated whole-brain irradiation (fWBI rats, 40 Gy, 8 fractions over 4 weeks) or sham-irradiation (Sham-IR rats) at 12 months of age; all analyses were performed at 26-30 months of age. Spatial learning and memory were measured using the Morris water maze (MWM), hippocampal metabolites were measured using proton magnetic resonance spectroscopy ((1)H MRS), and hippocampal glutamate receptor subunits were evaluated using Western blots. Young rats (7-10 months old) were included to control for age effects. The results revealed that both Sham-IR and fWBI rats exhibited age-dependent impairments in MWM performance; fWBI induced additional impairments in the reversal MWM. (1)H MRS revealed age-dependent decreases in neuronal markers, increases in glial markers, but no detectable fWBI-dependent changes. Western blot analysis revealed age-dependent, but not fWBI-dependent, glutamate subunit declines. Although previous studies demonstrated fWBI-induced changes in cognition, glutamate subunits, and brain metabolites in younger rats, age-dependent changes in these parameters appear to mask their detection in old rats, a phenomenon also likely to occur in elderly fWBI patients >70 years of age.


Subject(s)
Aging/metabolism , Aging/radiation effects , Brain Injuries/metabolism , Cognition Disorders/metabolism , Hippocampus/metabolism , Radiation Injuries/metabolism , Aging/psychology , Animals , Brain Injuries/diagnosis , Brain Injuries/psychology , Cognition Disorders/diagnosis , Cognition Disorders/psychology , Male , Radiation Injuries/diagnosis , Radiation Injuries/psychology , Random Allocation , Rats , Rats, Inbred BN , Rats, Inbred F344
7.
Neurobiol Aging ; 32(12): 2322.e1-4, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20561717

ABSTRACT

Degeneration of the cholinergic neurons in the basal forebrain and elevation of inflammatory markers are well-established hallmarks of Alzheimer's disease; however, the interplay of these processes in normal aging is not extensively studied. Consequently, we conducted a neuroanatomical investigation to quantify cholinergic neurons and activated microglia in the medial septum/vertical diagonal band (MS/VDB) of young (6 months) and aged (28 months) Fisher 344 × Brown Norway F(1) rats. Aged rats in this study were impaired relative to the young animals in spatial learning ability as assessed in the Morris water maze. Stereological analysis revealed no difference between aged and young rats in the total numbers of cholinergic neurons, demonstrating that loss of cholinergic neurons is not a necessary condition to observe impaired spatial learning in aged rats. In this same region, the total number of activated microglia was substantially greater in aged rats relative to young rats. Jointly, these data demonstrate that aging is characterized by an increase in the basal inflammatory state within the MS/VDB, but this inflammation is not associated with cholinergic neuron death.


Subject(s)
Aging , Brain Chemistry , Cholinergic Neurons/chemistry , Nerve Degeneration , Aging/pathology , Animals , Cholinergic Neurons/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Male , Nerve Degeneration/pathology , Rats , Rats, Inbred BN , Rats, Inbred F344
8.
J Neurosci ; 29(45): 14271-86, 2009 Nov 11.
Article in English | MEDLINE | ID: mdl-19906975

ABSTRACT

M(1) muscarinic acetylcholine receptors (mAChRs) may represent a viable target for treatment of disorders involving impaired cognitive function. However, a major limitation to testing this hypothesis has been a lack of highly selective ligands for individual mAChR subtypes. We now report the rigorous molecular characterization of a novel compound, benzylquinolone carboxylic acid (BQCA), which acts as a potent, highly selective positive allosteric modulator (PAM) of the rat M(1) receptor. This compound does not directly activate the receptor, but acts at an allosteric site to increase functional responses to orthosteric agonists. Radioligand binding studies revealed that BQCA increases M(1) receptor affinity for acetylcholine. We found that activation of the M(1) receptor by BQCA induces a robust inward current and increases spontaneous EPSCs in medial prefrontal cortex (mPFC) pyramidal cells, effects which are absent in acute slices from M(1) receptor knock-out mice. Furthermore, to determine the effect of BQCA on intact and functioning brain circuits, multiple single-unit recordings were obtained from the mPFC of rats that showed BQCA increases firing of mPFC pyramidal cells in vivo. BQCA also restored discrimination reversal learning in a transgenic mouse model of Alzheimer's disease and was found to regulate non-amyloidogenic APP processing in vitro, suggesting that M(1) receptor PAMs have the potential to provide both symptomatic and disease modifying effects in Alzheimer's disease patients. Together, these studies provide compelling evidence that M(1) receptor activation induces a dramatic excitation of PFC neurons and suggest that selectively activating the M(1) mAChR subtype may ameliorate impairments in cognitive function.


Subject(s)
Carboxylic Acids/pharmacology , Cholinergic Agents/pharmacology , Learning Disabilities/drug therapy , Neurons/drug effects , Prefrontal Cortex/drug effects , Quinolones/pharmacology , Reversal Learning/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , Female , Humans , In Vitro Techniques , Learning Disabilities/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/physiology , Prefrontal Cortex/physiology , Prefrontal Cortex/physiopathology , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M1/metabolism , Reversal Learning/physiology
9.
Curr Alzheimer Res ; 5(4): 385-91, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18690835

ABSTRACT

The medial temporal lobe-dependent memory loss associated with Alzheimer's disease (AD) is often accompanied by a loss of prefrontal cortex-dependent cognitive domains that fall under the broad category of executive function. In this study, we examined the relationship between one type of prefrontal-dependent executive function, discrimination reversal-learning, and levels of the amyloid beta protein (Abeta) of 40 and 42 residues in a transgenic mouse model (Tg2576) of the over-expression of the familial AD mutant form of the amyloid precursor protein (APPsw). Tg2576 and their non-transgenic (NTg) littermates were assessed at 3 and 6 months of age when there is little to no amyloid plaque deposition. After reversal-learning assessment, Abeta40 and Abeta42 were quantified in the prefrontal cortex and hippocampus. Tg2576 mice were impaired in reversal-learning at 6 but not 3 months of age when compared to the NTg group. Coincidently, there was a corresponding approximately 3-fold increase of Abeta42 levels in the prefrontal cortex of 6- compared to 3-month-old Tg2576 mice. In addition, the prefrontal cortex contained higher levels of Abeta42 compared to the hippocampus at both 3 and 6 months of age, regardless of genotype, indicating a high vulnerability of this brain region to Abeta42 accumulation. These data suggest that the early emergence of reversal-learning deficits in the Tg2576 mouse may be due to the localized increase of Abeta42 in the prefrontal cortex.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Learning Disabilities/etiology , Learning Disabilities/pathology , Peptide Fragments/metabolism , Prefrontal Cortex/metabolism , Reversal Learning/physiology , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Discrimination Learning , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Humans , Linear Models , Mice , Mice, Inbred C57BL , Mice, Transgenic , Odorants
10.
BMC Neurosci ; 8: 54, 2007 Jul 24.
Article in English | MEDLINE | ID: mdl-17650315

ABSTRACT

BACKGROUND: Long-term use of non-steroidal anti-inflammatory drugs (NSAIDs) is associated with a reduced incidence of Alzheimer's disease (AD). We and others have shown that certain NSAIDs reduce secretion of Abeta42 in cell culture and animal models, and that the effect of NSAIDs on Abeta42 is independent of the inhibition of cyclooxygenase by these compounds. Since Abeta42 is hypothesized to be the initiating pathologic molecule in AD, the ability of these compounds to lower Abeta42 selectively may be associated with their protective effect. We have previously identified R-flurbiprofen (tarenflurbil) as a selective Abeta42 lowering agent with greatly reduced cyclooxygenase activity that shows promise for testing this hypothesis. In this study we report the effect of chronic R-flurbiprofen treatment on cognition and Abeta loads in Tg2576 APP mice. RESULTS: A four-month preventative treatment regimen with R-flurbiprofen (10 mg/kg/day) was administered to young Tg2576 mice prior to robust plaque or Abeta pathology. This treatment regimen improved spatial learning as assessed by the Morris water maze, indicated by an increased spatial bias during the third probe trial and an increased utilization of a place strategy to solve the water maze. These results are consistent with an improvement in hippocampal- and medial temporal lobe-dependent memory function. A modest, though not statistically significant, reduction in formic acid-soluble levels of Abeta was also observed. To determine if R-flurbiprofen could reverse cognitive deficits in Tg2576 mice where plaque pathology was already robust, a two-week therapeutic treatment was given to older Tg2576 mice with the same dose of R-flurbiprofen. This approach resulted in a significant decrease in Abeta plaque burden but no significant improvement in spatial learning. CONCLUSION: We have found that chronic administration of R-flurbiprofen is able to attenuate spatial learning deficits if given prior to plaque deposition in Tg2576 mice. Given its ability to selectively target Abeta42 production and improve cognitive impairments in transgenic APP mice, as well as promising data from a phase 2 human clinical trial, future studies are needed to investigate the utility of R-flurbiprofen as an AD therapeutic and its possible mechanisms of action.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/metabolism , Brain/drug effects , Encephalitis/drug therapy , Flurbiprofen/therapeutic use , Learning Disabilities/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Brain/physiopathology , Cyclooxygenase Inhibitors/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Encephalitis/metabolism , Encephalitis/physiopathology , Female , Flurbiprofen/analogs & derivatives , Learning Disabilities/genetics , Learning Disabilities/physiopathology , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/drug therapy , Memory Disorders/genetics , Memory Disorders/physiopathology , Mice , Mice, Transgenic , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/biosynthesis , Plaque, Amyloid/drug effects , Plaque, Amyloid/metabolism , Stereoisomerism , Treatment Outcome
11.
Neurobiol Aging ; 28(3): 440-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-16504344

ABSTRACT

Tg2576 mice, a transgenic model of amyloid pathology associated with Alzheimer's disease (AD), develop measurable levels of soluble amyloid beta1-40 and 1-42 by 6 months of age and amyloid plaque deposition in cortex, hippocampus and amygdala by 10 months of age. To investigate whether non-hippocampal learning strategies would predominate coincident with the age-related increase in Abeta load in the hippocampal region, we measured learning strategies in the T-maze and a redundant cued version of the water maze. Each of these tasks can be solved using either hippocampal or non-hippocampal learning strategies and has proved sensitive to hippocampal disruption in other settings. The results revealed subtle differences in T-maze and water maze performance in Tg2576 mice compared to controls. Surprisingly, however, Tg2576 mice were not impaired relative to non-transgenic littermates on any measures of hippocampal dependent behavior assessed in these tasks. These data suggest that the medial temporal lobe retains considerable function in 15-month-old Tg2576 mice despite significant Abeta pathology.


Subject(s)
Amyloid beta-Peptides/genetics , Learning/physiology , Space Perception/physiology , Analysis of Variance , Animals , Behavior, Animal/physiology , Female , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
Neurobiol Aging ; 28(8): 1248-57, 2007 Aug.
Article in English | MEDLINE | ID: mdl-16828204

ABSTRACT

To understand the relationship between amyloid-beta and cognitive decline in Alzheimer's disease, we evaluated cortical and hippocampal function in a transgenic mouse model of amyloid over-expression in Alzheimer's disease, the Tg2576 mouse. Tg2576 mice and their non-transgenic littermates were assessed at both 6 and 14 months of age in a battery of cognitive tests: attentional set-shifting, water maze spatial reference memory and T-maze working memory. Spatial reference memory was not affected by Tg status at either age. Working memory was only affected by age, with 6-month-old mice performing better than 14-month-old ones. Older mice were also significantly impaired on reversal learning and on the intra- and extra-dimensional shift in attentional set-shifting. A significant transgene effect was apparent in reversal learning, with Tg2576 mice requiring more trials to reach criterion at 6 months old. These data indicate that the effects of normal aging in C57B6xSJL F1 mice are most pronounced on putative frontal cortex-dependent tasks and that increasing Abeta load only affects discrimination reversal learning in our study.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Protein Precursor/genetics , Discrimination Learning/physiology , Learning Disabilities/etiology , Reversal Learning/physiology , Space Perception/physiology , Age Factors , Alzheimer Disease/genetics , Analysis of Variance , Animals , Attention/physiology , Behavior, Animal , Disease Models, Animal , Female , Learning Disabilities/genetics , Longitudinal Studies , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuropsychological Tests
13.
Neurobiol Aging ; 28(4): 619-26, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16600436

ABSTRACT

The present study examined muscarinic receptor/G-protein coupling in the hippocampus and the prefrontal cortex of young and aged Long-Evans rats characterized for spatial learning ability in the Morris water maze. In a highly sensitive time-resolved fluorometry GTP-Eu binding assay, muscarinic-mediated GTP-Eu binding was severely blunted in hippocampus (-32%) and prefrontal cortex (-34%) as a consequence of aging. Furthermore, the magnitude of decreased muscarinic-mediated GTP-Eu binding was significantly correlated with the severity of spatial learning impairment in hippocampus and prefrontal cortex of aged rats and was specifically decreased in the subset of aged rats that were spatial learning impaired when compared to the aged unimpaired and the young rats. Western blot data indicated a preservation of the membrane-bound M1 receptor and the Galphaq/11 protein in both brain regions. These data demonstrate that muscarinic signaling is severely impaired as a consequence of normal aging in a manner that is closely associated with age-related cognitive decline.


Subject(s)
Aging/physiology , Guanosine Triphosphate/metabolism , Hippocampus/metabolism , Memory Disorders , Prefrontal Cortex/metabolism , Receptors, Muscarinic/physiology , Space Perception/physiology , Age Factors , Analysis of Variance , Animals , Behavior, Animal , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Hippocampus/drug effects , Male , Maze Learning/physiology , Memory Disorders/metabolism , Memory Disorders/pathology , Memory Disorders/physiopathology , Muscarinic Agonists/pharmacology , Oxotremorine/analogs & derivatives , Oxotremorine/pharmacology , Prefrontal Cortex/drug effects , Protein Binding/drug effects , Rats , Rats, Long-Evans
14.
Radiat Res ; 166(6): 892-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17149974

ABSTRACT

Whole-brain irradiation is used for the treatment of brain tumors, but can it also induce neural changes, with progressive dementia occurring in 20-50% of long-term survivors. The present study investigated whether 45 Gy of whole-brain irradiation delivered to 12-month-old Fischer 344 x Brown Norway rats as nine fractions over 4.5 weeks leads to impaired Morris water maze (MWM) performance 12 months later. Compared to sham-irradiated rats, the irradiated rats demonstrated impaired MWM performance. The relative levels of the NR1 and NR2A but not the NR2B subunits of the NMDA receptor were significantly higher in hippocampal CA1 of irradiated rats compared to control rats. No significant differences were detected for these NMDA subunits in CA3 or dentate gyrus. Further analysis of CA1 revealed that the relative levels of the GluR1 and GluR2 subunits of the AMPA receptor and synaptophysin were not altered by whole-brain irradiation. In summary, a clinically relevant regimen of fractionated whole-brain irradiation led to significant impairments in spatial learning and reference memory and alterations in the relative levels of subunits of the NMDA, but not the AMPA, receptors in hippocampal CA1. These findings suggest for the first time that radiation-induced cognitive impairments may be associated with alterations in glutamate receptor composition.


Subject(s)
Brain Injuries/metabolism , Hippocampus/physiology , Hippocampus/radiation effects , Memory Disorders/etiology , Memory Disorders/metabolism , Radiation Injuries/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain Injuries/etiology , Male , Organ Specificity , Protein Subunits/metabolism , Radiation Dosage , Radiation Injuries/etiology , Rats , Rats, Inbred F344
15.
Neuroreport ; 17(17): 1819-23, 2006 Nov 27.
Article in English | MEDLINE | ID: mdl-17164671

ABSTRACT

Cholinergic degeneration occurs in several neurodegenerative diseases. To investigate whether normal aging causes selective neurodegeneration, we compared counts of cholinergic neurons in the medial septum/vertical limb of the diagonal band and pedunculopontine and laterodorsal tegmental nuclei of the brainstem in young and aged Long-Evans rats characterized for their spatial learning ability in the Morris water maze. A subset of aged rats (aged-unimpaired) learned the spatial learning task as young rats, whereas another group (age-impaired) showed poorer learning than young animals. In the medial septum/diagonal band, there was a significant loss (-23%, P < 0.02) of cholinergic neurons in aged-impaired animals compared with young subjects. In the brainstem, there were no significant differences in cholinergic cell number in any group. This selective loss of cholinergic neurons may, in part, account for the cognitive deficits observed in aging and, considering previous findings in this model, may be related to oxidative stress.


Subject(s)
Acetylcholine/metabolism , Aging/pathology , Neurons/metabolism , Prosencephalon/cytology , Reticular Formation/cytology , Aging/physiology , Animals , Behavior, Animal , Cell Count/methods , Cell Death/physiology , Choline O-Acetyltransferase/metabolism , Immunohistochemistry , Learning Disabilities/pathology , Learning Disabilities/physiopathology , Male , Maze Learning/physiology , Neurons/cytology , Rats , Rats, Long-Evans , Space Perception/physiology
16.
Curr Alzheimer Res ; 3(3): 247-57, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16842102

ABSTRACT

We have pursued an interdisciplinary research program to develop novel behavioral assessment tools for evaluating specific memory impairments following damage to the medial temporal lobe, including the hippocampus and associated structures that show pathology early in the course of Alzheimer's disease (AD). Our approach uses computational models to identify the functional consequences of hippocampal-region damage, leading to testable predictions in both rodents and humans. Our modeling argues that hippocampal-region dysfunction may selectively impair the ability to generalize when familiar information is presented in novel recombinations. Previous research has shown that specific reductions in hippocampal volume in non-demented elderly individuals correlate with future development of AD. In two previous studies, we tested non-demented elderly with and without mild hippocampal atrophy (HA) on stimulus-response learning tasks. Individuals with and without HA could learn the initial information, but the HA group was selectively impaired on transfer tests where familiar features and objects were recombined. This suggests that such generalization deficits may be behavioral markers of HA, and an early indicator of risk for subsequent cognitive decline. Converging support for the relevance of these tasks to aging and Alzheimer's disease comes from our recent fMRI studies of individuals with mild cognitive impairment (MCI). Activity in the hippocampus declines with progressive training on these tasks, suggesting that the hippocampus is important for learning new stimulus representations that support subsequent transfer. Individuals with HA may be able to learn, but in a more hippocampal-independent fashion that does not support later transfer. Ultimately, this line of research could lead to a novel battery of behavioral tests sensitive to very mild hippocampal atrophy and risk for decline to AD, allowing early diagnosis and also allowing researchers to test new Alzheimer's drugs that target individuals in the earliest stages of the disease - before significant cognitive decline. A new mouse version of one of our tasks shows promise for translating these paradigms into rodents, allowing for future studies of therapeutic interventions in transgenic mouse models of AD.


Subject(s)
Aging/physiology , Alzheimer Disease/diagnosis , Brain Mapping/methods , Cognition Disorders/diagnosis , Geriatric Assessment/methods , Hippocampus/physiology , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Animals , Association Learning/physiology , Atrophy/diagnosis , Cognition Disorders/physiopathology , Computer Simulation , Discrimination Learning/physiology , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Magnetic Resonance Imaging , Mice , Models, Neurological , Neural Networks, Computer , Neuropsychological Tests , Predictive Value of Tests , Reference Values , Risk Assessment , Temporal Lobe/physiology , Transfer, Psychology/physiology
17.
Eur J Neurosci ; 18(12): 3335-42, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14686906

ABSTRACT

Aged Long-Evans rats exhibit deficits in attentional set shifting, an aspect of executive function, relative to adult rats. Impairments in set shifting and spatial learning are uncorrelated in aged rats, indicating a possible dissociation of the effects of ageing in prefrontal versus hippocampal systems. Ionotropic glutamate receptor binding was assessed using an in vitro autoradiography method in young and aged rats. The rats had been tested on a set-shifting task that measured attentional set shifts and reversal learning, as well as in a spatial learning task in the Morris water maze. [3H]Kainate, [3H]AMPA and NMDA-displaceable [3H]glutamate receptor binding were quantified in orbital cortex, cingulate cortex, medial frontal cortex, dorsolateral and dorsomedial striatum. Age-related decreases in [3H]kainate binding were apparent in all regions measured. Similarly, NMDA-displaceable [3H]glutamate binding was decreased in the aged rats in all the regions measured except for the medial frontal area where no age effects were observed. [3H]AMPA receptor binding was preserved with age in all the regions measured. Lower levels of [3H]kainate binding in the cingulate cortex were significantly correlated with poorer set-shifting performance, whereas higher levels of NMDA binding in the dorsomedial striatum were correlated with poorer set-shifting performance. There were no significant correlations between the levels of ionotropic glutamate receptors and performance in the reversal task or spatial learning in the Morris water maze. These results indicate that age-related behavioural deficits in attentional set shifting are selectively associated with neurobiological alterations in the cingulate cortex and dorsomedial striatum.


Subject(s)
Aging/metabolism , Cognition Disorders/metabolism , Frontal Lobe/metabolism , Neostriatum/metabolism , Receptors, Glutamate/metabolism , Aging/psychology , Animals , Attention/physiology , Binding, Competitive/physiology , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Down-Regulation/physiology , Excitatory Amino Acid Agonists/pharmacology , Frontal Lobe/physiopathology , Glutamic Acid/metabolism , Gyrus Cinguli/metabolism , Gyrus Cinguli/physiopathology , Maze Learning/physiology , Neostriatum/physiopathology , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Radioligand Assay , Rats , Rats, Long-Evans , Set, Psychology , Space Perception/physiology , Tritium
18.
Learn Mem ; 10(6): 520-4, 2003.
Article in English | MEDLINE | ID: mdl-14657263

ABSTRACT

The effects of age on cue learning, spatial reference memory, and strategy preference were assessed in B6 x SJL F1 mice by using the Morris water maze. This mouse strain is of particular interest because it is the background strain for a common transgenic model of Alzheimer's disease, the Tg2576 mouse, which develops plaques and other neurobiological markers of pathology beginning at 8 mo and increasing in severity with advanced age. In the current study, 12- and 23-mo-old C57B6 x SJL F1 mice were serially trained in cue and place versions of the Morris water maze task. At the completion of training, mice received a strategy probe test in which place (hidden) and cue (visible) strategies were in competition. Cue and spatial learning ability was maintained between 12 and 23 mo of age; however, on the strategy preference probe test, the 23-mo-old mice exhibited a significant bias toward the selection of a cue strategy. There was no relationship between strategy preference in the probe test and spatial learning ability, but the 23-mo-old mice did exhibit a strong trend toward shorter latencies during visible platform training, possibly reflecting the enhanced function of striatal-based neural systems in aging. These data demonstrate that 23-mo-old C57B6 x SJL F1 mice are capable of effective place learning, but if a place strategy is pitted against the use of a cue strategy, the use of a cue strategy predominates in the aged mice. The strategy preference observed here may reflect an emergence of differential processing in underlying brain circuitry with age in the B6 x SJL F1 mouse strain.


Subject(s)
Aging/physiology , Cues , Maze Learning/physiology , Memory/physiology , Animals , Female , Learning/physiology , Male , Mice , Mice, Inbred Strains , Space Perception/physiology , Water
SELECTION OF CITATIONS
SEARCH DETAIL
...