Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
J Intellect Disabil Res ; 65(12): 1085-1096, 2021 12.
Article in English | MEDLINE | ID: mdl-34786786

ABSTRACT

BACKGROUND: Individuals with Down syndrome (DS) are at high risk for dementia, specifically Alzheimer's disease. However, many measures regularly used for the detection of dementia in the general population are not suitable for individuals with DS due in part to floor effects. Some measures, including the Severe Impairment Battery (SIB), Brief Praxis Test (BPT) and Dementia Scale for People with Learning Disabilities (DLD), have been used in clinical trials and other research with this population. Validity research is limited, particularly regarding the use of such tools for detection of prodromal dementia in the DS population. The current project presents baseline cross-sectional SIB, BPT and DLD performance in order to characterise their predictive utility in discriminating normal cognition, possible dementia and probable dementia in adult DS. METHOD: Baseline SIB, BPT and DLD performances from 100 individuals (no dementia = 68, possible dementia = 16 & probable dementia = 16) were examined from a longitudinal cohort of aging individuals with DS. Receiver operating characteristic curves investigated the accuracy of these measures in relation to consensus dementia diagnoses, diagnoses which demonstrated high percent agreement with the examining neurologist's independent diagnostic impression. RESULTS: The SIB and BPT exhibited fair discrimination ability for differentiating no/possible versus probable dementia [area under the curve (AUC) = 0.61 and 0.66, respectively]. The DLD exhibited good discrimination ability for differentiating no versus possible/probable dementia (AUC = 0.75) and further demonstrated better performance of the DLD Cognitive subscale compared with the DLD Social subscale (AUC = 0.77 and 0.67, respectively). CONCLUSIONS: Results suggest that the SIB, BPT and DLD are able to reasonably discriminate consensus dementia diagnoses in individuals with DS, supporting their continued use in the clinical assessment of dementia in DS. The general performance of these measures suggests that further work in the area of test development is needed to improve on the AUCs for dementia status discrimination in this unique population. At present, however, the current findings suggest that the DLD may be the best option for reliable identification of prodromal dementia in this population, reinforcing the importance of including informant behaviour ratings in assessment of cognition for adults with DS.


Subject(s)
Alzheimer Disease , Dementia , Down Syndrome , Learning Disabilities , Adult , Cross-Sectional Studies , Dementia/diagnosis , Down Syndrome/complications , Down Syndrome/diagnosis , Humans , Neuropsychological Tests
2.
J Intellect Disabil Res ; 64(12): 934-945, 2020 12.
Article in English | MEDLINE | ID: mdl-32996650

ABSTRACT

BACKGROUND: People with Down syndrome (DS) develop Alzheimer's disease (AD) at an earlier age of onset than those with sporadic AD. AD neuropathology is typically present in DS by 40 years of age with an onset of dementia approximately 10 years later. This early onset is due to the overexpression of amyloid precursor protein from the third copy of chromosome 21. Cerebrovascular neuropathology is thought to contribute in 40-60% of cases sporadic AD. However, the vascular contribution to dementia in people with DS has been relatively unexplored. We hypothesised that vascular perfusion is compromised in older adults with DS relative to younger individuals and is further exacerbated in those with dementia. METHOD: Cerebral blood flow (CBF) was measured using pulsed arterial spin labelling in 35 cognitively characterised adults with DS (26-65 years). DS participants were also compared with 15 control subjects without DS or dementia (26-65 years). Linear regression evaluated the difference in CBF across groups and diagnosis along with assessing the association between CBF and cognitive measures within the DS cohort. RESULTS: Cerebral blood flow was significantly lower among DS participants with probable AD compared with controls (P = 0.02) and DS participants with no dementia (P = 0.01). Within the DS cohort, CBF was significantly associated with the Severe Impairment Battery (SIB) measure and the Dementia Questionnaire for People with Learning Disabilities (DLD) rating (F3,25  = 5.13; P = 0.007). Both the SIB (ß = 0.74; t = 2.71; P = 0.01) and DLD (ß = -0.96; t = -3.87; P < 0.001) indicated greater impairment as global CBF decreased. Age was significantly associated with CBF among participants with DS. There was a non-linear effect of age, whereby CBF declined more rapidly after 45 years of age. CONCLUSIONS: This preliminary study of CBF in DS indicates that cerebrovascular pathology may be a significant contributor to dementia in DS. CBF was associated with diagnosis, cognition and age. Notably, CBF decreases at a greater rate after age 45 and may represent a significant prodromal event in AD progression.


Subject(s)
Aging/physiology , Cerebrovascular Circulation/physiology , Dementia/epidemiology , Down Syndrome/epidemiology , Adult , Aged , Comorbidity , Dementia/physiopathology , Down Syndrome/physiopathology , Female , Humans , Longitudinal Studies , Male , Middle Aged , United States/epidemiology
3.
Neuroimage Clin ; 11: 728-735, 2016.
Article in English | MEDLINE | ID: mdl-27330972

ABSTRACT

To determine if proton magnetic resonance spectroscopy ((1)H-MRS) detect differences in dementia status in adults with Down syndrome (DS), we used (1)H-MRS to measure neuronal and glial metabolites in the posterior cingulate cortex in 22 adults with DS and in 15 age- and gender-matched healthy controls. We evaluated associations between (1)H-MRS results and cognition among DS participants. Neuronal biomarkers, including N-acetylaspartate (NAA) and glutamate-glutamine complex (Glx), were significantly lower in DS patients with Alzheimer's should probably be changed to Alzheimer (without ' or s) through ms as per the new naming standard disease (DSAD) when compared to non-demented DS (DS) and healthy controls (CTL). Neuronal biomarkers therefore appear to reflect dementia status in DS. In contrast, all DS participants had significantly higher myo-inositol (MI), a putative glial biomarker, compared to CTL. Our data indicate that there may be an overall higher glial inflammatory component in DS compared to CTL prior to and possibly independent of developing dementia. When computing the NAA to MI ratio, we found that presence or absence of dementia could be distinguished in DS. NAA, Glx, and NAA/MI in all DS participants were correlated with scores from the Brief Praxis Test and the Severe Impairment Battery. (1)H-MRS may be a useful diagnostic tool in future longitudinal studies to measure AD progression in persons with DS. In particular, NAA and the NAA/MI ratio is sensitive to the functional status of adults with DS, including prior to dementia.


Subject(s)
Aspartic Acid/analogs & derivatives , Dementia/etiology , Dementia/metabolism , Down Syndrome/complications , Gyrus Cinguli/metabolism , Proton Magnetic Resonance Spectroscopy , Activities of Daily Living , Adult , Analysis of Variance , Aspartic Acid/metabolism , Dementia/psychology , Down Syndrome/pathology , Female , Glutamic Acid/metabolism , Glutamine/metabolism , Gyrus Cinguli/pathology , Humans , Inositol/metabolism , Male , Middle Aged , Neuropsychological Tests , Psychiatric Status Rating Scales , Surveys and Questionnaires
4.
Vet Pathol ; 53(2): 327-48, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26869150

ABSTRACT

According to the WHO, the proportion of people over 60 years is increasing and expected to reach 22% of total world's population in 2050. In parallel, recent animal demographic studies have shown that the life expectancy of pet dogs and cats is increasing. Brain aging is associated not only with molecular and morphological changes but also leads to different degrees of behavioral and cognitive dysfunction. Common age-related brain lesions in humans include brain atrophy, neuronal loss, amyloid plaques, cerebrovascular amyloid angiopathy, vascular mineralization, neurofibrillary tangles, meningeal osseous metaplasia, and accumulation of lipofuscin. In aging humans, the most common neurodegenerative disorder is Alzheimer's disease (AD), which progressively impairs cognition, behavior, and quality of life. Pathologic changes comparable to the lesions of AD are described in several other animal species, although their clinical significance and effect on cognitive function are poorly documented. This review describes the commonly reported age-associated neurologic lesions in domestic and laboratory animals and the relationship of these lesions to cognitive dysfunction. Also described are the comparative interspecies similarities and differences to AD and other human neurodegenerative diseases including Parkinson's disease and progressive supranuclear palsy, and the spontaneous and transgenic animal models of these diseases.


Subject(s)
Aging/pathology , Animals, Domestic , Animals, Laboratory , Cat Diseases/pathology , Dog Diseases/pathology , Neurodegenerative Diseases/veterinary , Alzheimer Disease/pathology , Alzheimer Disease/veterinary , Animals , Brain/pathology , Cats , Cerebral Amyloid Angiopathy/pathology , Cerebral Amyloid Angiopathy/veterinary , Disease Models, Animal , Dogs , Humans , Neurodegenerative Diseases/pathology , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Plaque, Amyloid/veterinary , Quality of Life
6.
J Comp Pathol ; 149(2-3): 237-41, 2013.
Article in English | MEDLINE | ID: mdl-23660088

ABSTRACT

Several subtypes of canine meningioma are recognized. This report describes four canine meningiomas with previously unreported features. The four affected dogs were of different breeds. Three of the affected dogs were male and aged 7-10 years. In one dog, age and gender were not recorded. Meningiomas were located intracranially (three dogs) or within the vertebral canal (one dog). Two meningiomas resembled gemistocytic astrocytomas, while one had focal features of a rhabdoid tumour; these three meningiomas also contained amyloid deposits. The fourth tumour, a secretory meningioma, was rich in amianthoid fibres (i.e. unusual collagen deposits containing giant collagen fibres). All of these features are also described in human meningiomas.


Subject(s)
Dog Diseases/pathology , Meningeal Neoplasms/veterinary , Meningioma/veterinary , Animals , Biomarkers, Tumor/analysis , Dogs , Female , Immunohistochemistry , Male , Meningeal Neoplasms/pathology , Meningioma/pathology
7.
Exp Neurol ; 220(1): 171-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19703441

ABSTRACT

Dogs develop cognitive decline and a progressive accumulation of oxidative damage. In a previous longitudinal study, we demonstrated that aged dogs treated with either an antioxidant diet or with behavioral enrichment show cognitive improvement. The antioxidant diet included cellular antioxidants (vitamins E and C, fruits and vegetables) and mitochondrial cofactors (lipoic acid and carnitine). Behavioral enrichment consisted of physical exercise, social enrichment, and cognitive training. We hypothesized that the antioxidant treatment improved neuronal function through increased mitochondrial function. Thus, we measured reactive oxygen species (ROS) production and bioenergetics in mitochondria isolated from young, aged, and treated aged animals. Aged canine brain mitochondria show significant increases in ROS production and a reduction in NADH-linked respiration. Mitochondrial function (ROS and NADH-linked respiration) was improved selectively in aged dogs treated with an antioxidant diet. In contrast, behavioral enrichment had no effect on any mitochondrial parameters. These results suggest that an antioxidant diet improves cognition by maintaining mitochondrial homeostasis, which may be an independent molecular pathway not engaged by behavioral enrichment.


Subject(s)
Aging/metabolism , Brain/metabolism , Food, Formulated , Memory Disorders/diet therapy , Memory Disorders/metabolism , Mitochondria/metabolism , Aging/drug effects , Aging/pathology , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/drug effects , Brain/physiopathology , Coenzymes/pharmacology , Coenzymes/therapeutic use , Cognitive Behavioral Therapy/methods , Disease Models, Animal , Dogs , Environment, Controlled , Exercise Therapy/methods , Female , Humans , Male , Memory Disorders/physiopathology , Mitochondria/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Physical Conditioning, Animal/methods , Treatment Outcome
8.
Neuroimage ; 39(3): 1324-32, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18006337

ABSTRACT

BACKGROUND: We report functional and structural brain indicators that may precede the onset of dementia in individuals with Down's syndrome (DS). METHODS: Middle-aged adults with DS (n=19), a group known to be at high risk for dementia, were studied with (1) positron emission tomography (PET) to determine cerebral glucose metabolic rate (GMR), (2) structural magnetic resonance imaging (MRI) to determine gray matter volume (GM), and (3) ratings of potential dementia indicators based on a structured interview of caregiver observations designed to evaluate individuals with low intelligence. RESULTS: Although none of the participants showed clinical signs of dementia, ratings of dementia indicators were correlated to both functional and structural imaging. The strongest correlations (p<.05, corrected for multiple comparisons) included the combination of higher GMR and decreased GM volume in parts of the temporal cortex, including the parahippocampus/hippocampus, in the thalamus, caudate, and frontal lobe (BA 47). INTERPRETATION: The combination of increased GMR overlapping with less gray matter in these areas may be consistent with a compensatory brain response to an early stage of the disease process.


Subject(s)
Dementia/pathology , Down Syndrome/pathology , Adult , Brain Chemistry/physiology , Cognition/physiology , Data Interpretation, Statistical , Dementia/diagnostic imaging , Dementia/metabolism , Down Syndrome/diagnostic imaging , Down Syndrome/metabolism , Emotions/physiology , Female , Follow-Up Studies , Glucose/metabolism , Humans , Individuality , Kinetics , Magnetic Resonance Imaging , Male , Middle Aged , Positron-Emission Tomography , Predictive Value of Tests , Psychiatric Status Rating Scales , Risk
9.
J Small Anim Pract ; 48(10): 546-53, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17617164

ABSTRACT

With improvements in nutrition and veterinary medicine the life expectancy of pet cats is increasing. Accompanying this growing geriatric population there are increasing numbers of cats with signs of apparent senility. A recent study suggests that 28 per cent of pet cats aged 11 to 14 years develop at least one geriatric onset behavioural problem, and this increases to over 50 per cent for cats of 15 years of age or older. While behavioural changes may result from systemic illness, organic brain disease or true behavioural problems, the possibility of age-related cognitive dysfunction is often overlooked. Studies have revealed a number of changes in the brains of geriatric cats that showed signs of cognitive dysfunction, and potential causes include vascular insufficiency leading to hypoxia, increased free radical damage and the deposition of beta-amyloid plaques and/or the modification of other proteins. By recognising the importance of behavioural changes in old cats, investigating them fully for potentially treatable medical conditions, and instigating dietary and environmental modifications to meet their changing needs, we can make the lives of our geriatric cats much more comfortable and rewarding.


Subject(s)
Aging , Brain/pathology , Cat Diseases/pathology , Cognition Disorders/pathology , Animals , Behavior, Animal , Cats
10.
J Alzheimers Dis ; 11(1): 61-76, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17361036

ABSTRACT

Adults with Down syndrome (DS) develop Alzheimer disease (AD) pathology progressively with age but clinical signs of dementia are delayed by at least 10 years after the first signs of disease. Some individuals with DS do not develop dementia despite extensive AD neuropathology. Given the discordance between clinical decline and AD neuropathology, compensatory events may be of particular relevance for this group. Imaging studies using PET suggest compensatory increases in metabolic rate in vulnerable brain regions in DS prior to the development of dementia. Neurobiological studies of similarly aged DS autopsy cases provide further evidence of activation of plasticity mechanisms. Genes that are overexpressed in DS (APP, DSCAM, MNB/DYRK1A, and RCAN1) produce proteins critical for neuron and synapse growth, development and maintenance. We present the hypothesis that these genes may lead to developmental cognitive deficits but paradoxically with aging, may participate in molecular cascades supporting neuronal compensation. Enhancing or supporting compensatory mechanisms in aging individuals with DS may be beneficial as suggested by intervention studies in animal models. In combination, adults with DS may be a unique group of individuals well-suited for studies involving the manipulation or upregulation of compensatory responses as an approach to promote successful brain aging in the general population.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Down Syndrome/genetics , Down Syndrome/pathology , Neuronal Plasticity/genetics , Age Factors , Aged , Alzheimer Disease/therapy , Amyloid beta-Protein Precursor , Animals , Cell Adhesion Molecules , Chromosomes, Human, Pair 21/genetics , DNA-Binding Proteins , Down Syndrome/therapy , Energy Metabolism/genetics , Female , Gene Expression Regulation/physiology , Humans , Intracellular Signaling Peptides and Proteins , Male , Membrane Proteins , Middle Aged , Muscle Proteins , Neurons/pathology , Positron-Emission Tomography , Protease Nexins , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Receptors, Cell Surface , Synapses/pathology , Up-Regulation/genetics , Dyrk Kinases
11.
Neurobiol Aging ; 28(10): 1493-506, 2007 Oct.
Article in English | MEDLINE | ID: mdl-16904243

ABSTRACT

Aged individuals with Down syndrome (DS) develop Alzheimer's disease (AD) neuropathology by the age of 40 years. The purpose of the current study was to measure age-associated changes in APP processing in 36 individuals with DS (5 months-69 years) and in 26 controls (5 months-100 years). Alpha-secretase significantly decreased with age in DS, particularly in cases over the age of 40 years and was stable in controls. The levels of C-terminal fragments of APP reflecting alpha-secretase processing (CTF-alpha) decreased with age in both groups. In both groups, there was significant increase in beta-secretase activity with age. CTF-beta remained constant with age in controls suggesting compensatory increases in turnover/clearance mechanisms. In DS, young individuals had the lowest CTF-beta levels that may reflect rapid conversion of beta-amyloid (Abeta) to soluble pools or efficient CTF-beta clearance mechanisms. Treatments to slow or prevent AD in the general population targeting secretase activity may be more efficacious in adults with DS if combined with approaches that enhance Abeta degradation and clearance.


Subject(s)
Aging/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/metabolism , Brain/enzymology , Down Syndrome/enzymology , Adolescent , Adult , Aged , Aging/pathology , Biomarkers/metabolism , Brain/pathology , Brain/physiopathology , Child , Child, Preschool , Disease Progression , Down Syndrome/pathology , Down Syndrome/physiopathology , Female , Humans , Infant , Male , Middle Aged , Plaque, Amyloid/enzymology , Plaque, Amyloid/pathology , Reference Values , Up-Regulation/physiology
12.
Neurobiol Aging ; 26(5): 749-63, 2005 May.
Article in English | MEDLINE | ID: mdl-15708450

ABSTRACT

The current study describes both Abeta and tau abnormalities that accumulate in the brains of aged (16-21 years), but not young (<4 years) clinically characterized cats. Diffuse plaques that were morphologically different from what is typically observed in the human brain could be detected with 4G8 (Abeta17-24) or an Abeta1-42-specific antibody but not with N-terminal Abeta or an Abeta1-40-specific antibody. SELDI-TOF mass spectrometry experiments indicated that cat brain Abeta consisted almost entirely of Abeta1-42. Markers of tau hyperphosphorylation (AT8 and PHF-1) labeled a subset of neurons in two aged animals. In the hilus of the hippocampus, a subset of AT8 positive neurons showed a sprouting morphology similar to that observed in human brain. Western blot analysis with antibodies against hyperphosphorylated tau indicated that tau is hyperphosphorylated in the aged cat and contains many of the same epitopes found in Alzheimer's disease (AD) brain. Thus, the aged cat brain develops AD-related lesions with important morphological and biochemical differences compared to human brain.


Subject(s)
Aging/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , tau Proteins/metabolism , Animals , Behavior, Animal , Blotting, Western/methods , Brain/anatomy & histology , Brain/pathology , Cats , Dogs , Female , Humans , Immunohistochemistry/methods , Male , Mass Spectrometry/methods , Mice , Mice, Transgenic , Phosphorylation , Protein Array Analysis/methods , Rats
13.
Neurobiol Aging ; 26(1): 77-90, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15585348

ABSTRACT

The effectiveness of two interventions, dietary fortification with antioxidants and a program of behavioral enrichment, was assessed in a longitudinal study of cognitive aging in beagle dogs. A baseline protocol of cognitive testing was used to select four cognitively equivalent groups: control food-control experience (C-C), control food-enriched experience (C-E), antioxidant fortified food-control experience (A-C), and antioxidant fortified food-enriched experience(A-E). We also included two groups of young behaviorally enriched dogs, one receiving the control food and the other the fortified food. Discrimination learning and reversal was assessed after one year of treatment with a size discrimination task, and again after two years with a black/white discrimination task. The four aged groups were comparable at baseline. At one and two years, the aged combined treatment group showed more accurate learning than the other aged groups. Discrimination learning was significantly improved by behavioral enrichment. Reversal learning was improved by both behavioral enrichment and dietary fortification. By contrast, the fortified food had no effect on the young dogs. These results suggest that behavioral enrichment or dietary fortification with antioxidants over a long-duration can slow age-dependent cognitive decline, and that the two treatments together are more effective than either alone in older dogs.


Subject(s)
Antioxidants/pharmacology , Behavior, Animal/physiology , Environment , Food, Fortified , Learning/physiology , Age Factors , Aging/physiology , Analysis of Variance , Animals , Discrimination Learning/drug effects , Discrimination Learning/physiology , Dogs , Female , Learning/drug effects , Longitudinal Studies , Male , Reversal Learning/drug effects , Reversal Learning/physiology , Time Factors
14.
Neurology ; 61(12): 1673-9, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14694028

ABSTRACT

BACKGROUND: Adults with Down syndrome (DS) are at increased risk for dementia and provide an opportunity to identify patterns of brain activity that may precede dementia. Studies of early Alzheimer's disease (AD) and risk of AD show decreased function in posterior cingulate and temporal cortex as initial indicators of the disease process, but whether the origin and sequence of predementia brain changes are the same in DS is unknown. METHODS: The regional cerebral glucose metabolic rates (GMR) among middle-aged nondemented people with DS (n = 17), people with moderate AD (n = 10), and age-matched control subjects (n = 24) were compared using PET during a cognitive task. RESULTS: Statistical parametric mapping conjunction analyses showed that 1) both DS and AD groups had lower GMR than their respective controls primarily in posterior cingulate and 2) compared with respective controls, the subjects with DS had higher GMR in the same areas of inferior temporal/entorhinal cortex where the AD subjects had lower GMR. The same results were replicated after 1 year of follow-up. CONCLUSIONS: As the DS subjects were not clinically demented, inferior temporal/entorhinal cortex hypermetabolism may reflect a compensatory response early in disease progression. Compensatory responses may subsequently fail, leading to neurodegenerative processes that the authors anticipate will be detectable in vivo as future GMR decreases in inferior temporal/entorhinal cortex are accompanied by clinical signs of dementia.


Subject(s)
Alzheimer Disease/metabolism , Down Syndrome/metabolism , Glucose/metabolism , Temporal Lobe/metabolism , Adult , Aged , Alzheimer Disease/diagnosis , Alzheimer Disease/diagnostic imaging , Down Syndrome/diagnosis , Down Syndrome/diagnostic imaging , Entorhinal Cortex/diagnostic imaging , Entorhinal Cortex/metabolism , Female , Fluorodeoxyglucose F18/pharmacokinetics , Follow-Up Studies , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/metabolism , Humans , Male , Middle Aged , Neuropsychological Tests , Reference Values , Reproducibility of Results , Temporal Lobe/diagnostic imaging , Tomography, Emission-Computed
15.
J Neuropathol Exp Neurol ; 62(9): 917-26, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14533781

ABSTRACT

Aged individuals with Down syndrome (DS) develop senile plaques and neurofibrillary tangles consistent with Alzheimer disease (AD). Prior to or in parallel with AD pathology, compensatory growth responses may occur. Immunohistochemistry and confocal microscopy studies in the hippocampus from 15 individuals ranging in age from 5 months to 67 years compared markers of normal and abnormal tau accumulation (phosphorylated tau [AT8, MC-1], tau-1, N-terminal tau) with the extent and location of neuronal growth marker immunoreactivity (BDNF, GAP-43, MAP-2). In middle age (30-40 years), prior to entorhinal neuron loss, the earliest tau accumulation occurred in the outer molecular layer (OML), which was consistent with both pathological and compensatory fetal tau expression. These events were followed at a later age, associated with entorhinal neuron loss, by an increase in GAP-43. Hilar neurons exhibiting a sprouting morphology were also noted. Age-dependent observations in the DS brain in the current study parallel hippocampal compensatory responses described in entorhinal cortex lesion studies in rodents. Thus, compensatory growth responses may occur in DS prior to extensive AD pathology and may be one mechanism underlying observations in PET studies of hypermetabolism in the entorhinal cortex of individuals with DS.


Subject(s)
Down Syndrome/metabolism , Down Syndrome/pathology , tau Proteins/metabolism , Adolescent , Adult , Age Factors , Age of Onset , Aged , Child , Child, Preschool , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Female , Gene Expression Regulation/physiology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Infant , Male , Middle Aged , Phosphorylation
16.
Neuroscience ; 120(2): 291-300, 2003.
Article in English | MEDLINE | ID: mdl-12890502

ABSTRACT

Alzheimer's disease (AD) is associated with the accumulation of extracellular deposits of the beta-amyloid protein (Abeta). Abeta is a result of misprocessing of the beta-amyloid precursor protein (APP). Gamma-secretase is involved in APP misprocessing and one hypothesis holds that this secretase is identical to PS1. We tested this hypothesis by determining whether PS is co-localised with Abeta in situ. Using confocal analyses and a sensitive immunogold procedure we show that PS and Abeta are co-localised within discrete microdomains of neuronal plasma membranes in AD patients and in aged dogs, an established model of human brain aging. Our data indicate that APP misprocessing occurs in discrete plasma membrane domains of neurons and provide evidence that PS1 is critically involved in Abeta formation.


Subject(s)
Amyloid beta-Peptides/metabolism , Cell Membrane/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Aged , Aged, 80 and over , Animals , Cell Membrane/ultrastructure , Dogs , Enzyme-Linked Immunosorbent Assay/methods , Female , Fluorescent Antibody Technique/methods , Gangliosidosis, GM1/metabolism , Humans , Immunohistochemistry/methods , Male , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Microscopy, Immunoelectron/instrumentation , Microscopy, Immunoelectron/methods , Neurons/ultrastructure , Plaque, Amyloid/metabolism , Plaque, Amyloid/ultrastructure , Presenilin-1 , Presenilin-2 , Protein Structure, Tertiary/physiology
17.
Neurosci Biobehav Rev ; 26(6): 679-95, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12479842

ABSTRACT

The landmark discrimination learning test can be used to assess the ability to utilize allocentric spatial information to locate targets. The present experiments examined the role of various factors on performance of a landmark discrimination learning task in beagle dogs. Experiments 1 and 2 looked at the effects of age and food composition. Experiments 3 and 4 were aimed at characterizing the cognitive strategies used in performance on this task and in long-term retention. Cognitively equivalent groups of old and young dogs were placed into either a test group maintained on food enriched with a broad-spectrum of antioxidants and mitochondrial cofactors, or a control group maintained on a complete and balanced food formulated for adult dogs. Following a wash-in period, the dogs were tested on a series of problems, in which reward was obtained when the animal responded selectively to the object closest to a thin wooden block, which served as a landmark. In Experiment 1, dogs were first trained to respond to a landmark placed directly on top of coaster, landmark 0 (L0). In the next phase of testing, the landmark was moved at successively greater distances (1, 4 or 10 cm) away from the reward object. Learning varied as a function of age group, food group, and task. The young dogs learned all of the tasks more quickly than the old dogs. The aged dogs on the enriched food learned L0 significantly more rapidly than aged dogs on control food. A higher proportion of dogs on the enriched food learned the task, when the distance was increased to 1cm. Experiment 2 showed that accuracy decreased with increased distance between the reward object and landmark, and this effect was greater in old animals. Experiment 3 showed stability of performance, despite using a novel landmark, and new locations, indicating that dogs learned the landmark concept. Experiment 4 found age impaired long-term retention of the landmark task. These results indicate that allocentric spatial learning is impaired in an age-dependent manner in dogs, and that age also affects performance when the distance between the landmark and target is increased. In addition, these results both support a role of oxidative damage in the development of age-associated cognitive dysfunction and indicate that short-term administration of a food enriched with supplemental antioxidants and mitochondrial cofactors can partially reverse the deleterious effects of aging on cognition.


Subject(s)
Aging/physiology , Antioxidants/pharmacology , Cognition/physiology , Diet , Discrimination Learning/drug effects , Discrimination Learning/physiology , Spatial Behavior/drug effects , Spatial Behavior/physiology , Age Factors , Analysis of Variance , Animals , Blood Chemical Analysis/methods , Distance Perception/drug effects , Distance Perception/physiology , Dogs/physiology , Female , Male , Psychomotor Performance/physiology , Retention, Psychology/drug effects , Retention, Psychology/physiology , Task Performance and Analysis , Time Factors , Vitamin E/blood
18.
Neurobiol Aging ; 23(5): 737-45, 2002.
Article in English | MEDLINE | ID: mdl-12392778

ABSTRACT

Advanced age is accompanied by cognitive decline indicative of central nervous system dysfunction. One possibly critical causal factor is oxidative stress. Accordingly, we studied the effects of dietary antioxidants and age in a canine model of aging that parallels the key features of cognitive decline and neuropathology in humans. Old and young animals were placed on either a standard control food, or a food enriched with a broad spectrum of antioxidants and mitochondrial enzymatic cofactors. After 6 months of treatment, the animals were tested on four increasingly difficult oddity discrimination learning problems. The old animals learned more slowly than the young, making significantly more errors. However, this age-associated decline was reduced in the animals fed the enriched food, particularly on the more difficult tasks. These results indicate that maintenance on foods fortified with complex mixtures of antioxidants can partially counteract the deleterious effects of aging on cognition.


Subject(s)
Aging/metabolism , Animal Feed , Cognition Disorders/diet therapy , Cognition Disorders/metabolism , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Cognition Disorders/prevention & control , Conditioning, Psychological , Diet , Discrimination Learning , Dogs , Female , Male , Mitochondria/metabolism , Oxidative Stress , alpha-Tocopherol/blood , alpha-Tocopherol/pharmacology
19.
J Neurochem ; 82(2): 375-81, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12124438

ABSTRACT

We assayed levels of lipid peroxidation, protein carbonyl formation, glutamine synthetase (GS) activity and both oxidized and reduced glutathione to study the link between oxidative damage, aging and beta-amyloid (Abeta) in the canine brain. The aged canine brain, a model of human brain aging, naturally develops extensive diffuse deposits of human-type Abeta. Abeta was measured in immunostained prefrontal cortex from 19 beagle dogs (4-15 years). Increased malondialdehyde (MDA), which indicates increased lipid peroxidation, was observed in the prefrontal cortex and serum but not in cerebrospinal fluid (CSF). Oxidative damage to proteins (carbonyl formation) also increased in brain. An age-dependent decline in GS activity, an enzyme vulnerable to oxidative damage, and in the level of glutathione (GSH) was observed in the prefrontal cortex. MDA level in serum correlated with MDA accumulation in the prefrontal cortex. Although 11/19 animals exhibited Abeta, the extent of deposition did not correlate with any of the oxidative damage measures, suggesting that each form of neuropathology accumulates in parallel with age. This evidence of widespread oxidative damage and Abeta deposition is further justification for using the canine model for studying human brain aging and neurodegenerative diseases.


Subject(s)
Aging/metabolism , Prefrontal Cortex/metabolism , Aging/pathology , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Animals , Cerebrospinal Fluid/chemistry , Dogs , Glutamate-Ammonia Ligase/chemistry , Glutamate-Ammonia Ligase/metabolism , Glutathione/analysis , Glutathione/metabolism , Glutathione Disulfide/analysis , Glutathione Disulfide/metabolism , Humans , Lipid Peroxidation , Malondialdehyde/analysis , Malondialdehyde/metabolism , Oxidation-Reduction , Prefrontal Cortex/chemistry , Prefrontal Cortex/pathology , Proteins/chemistry , Proteins/metabolism , Time
20.
Neurobiol Dis ; 8(6): 1006-16, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11741396

ABSTRACT

Recent studies support the activation of apoptotic pathways in the Alzheimer's disease (AD) brain. Neurons committed to apoptosis may do so by either activation of a receptor-mediated pathway employing caspase-8 or through an alternative mitochondrial pathway involving oxidative stress. In the present study, the role of caspase-8 in the AD brain was examined by designing a caspase-cleavage site-directed antibody to one of the active fragments of caspase-8. In vitro analysis with this antibody, termed CASP-8p18, demonstrated that it recognized the active 18-kDa fragment of caspase-8 but not the precursor protein. In vivo immunohistochemical analysis using hippocampal tissue sections from AD or aged-matched control brains demonstrated CASP-8p18 immunolabeling of neurons in all AD cases, whereas little staining was observed in controls. These results were confirmed using a commercially available antibody that, like the CASP-8p18 antibody reacts only with the 18-kDa fragment of caspase-8 and not full-length caspase-8. As with CASP-8p18 antibody, the commercial antibody-labeled neurons in all AD cases, while showing a relative paucity of staining in representative control cases. Labeling of CASP-8p18 within tangle-bearing neurons was observed in double-labeling studies with AT8 or PHF-1, both markers for neurofibrillary tangles (NFTs). In addition, using a caspase-cleavage site-directed antibody that recognizes cleavage products of caspase-3 showed colocalization of this antibody with the CASP-8p18 antibody within NFTs. These results suggest a role for caspase-8 and the receptor-mediated apoptotic pathway as a mechanism leading to the activation of caspase-3 within neurons of the AD brain.


Subject(s)
Alzheimer Disease/enzymology , Apoptosis/physiology , Caspases/metabolism , Entorhinal Cortex/enzymology , Hippocampus/enzymology , Neurons/enzymology , Aged , Aging/metabolism , Aging/pathology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Antibodies, Monoclonal/metabolism , Antibody Specificity , Blotting, Western , Carrier Proteins/metabolism , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/immunology , Cell Compartmentation/immunology , Dogs , Entorhinal Cortex/pathology , Entorhinal Cortex/physiopathology , Female , HeLa Cells , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Immunohistochemistry , Male , Microfilament Proteins/metabolism , Middle Aged , Neurofibrillary Tangles/enzymology , Neurofibrillary Tangles/pathology , Neurons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...