Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Hippocampus ; 22(10): 1978-89, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22987676

ABSTRACT

Previous lesion studies have shown compromised complex object discrimination in rats, monkeys, and human patients with damage to the perirhinal cortical region (PRC) of the medial temporal lobe. These findings support the notion that the PRC is involved in object discrimination when pairs of objects have a high degree of overlapping features but not when object discrimination can be resolved on the basis of a single feature (e.g., size or color). Recent studies have demonstrated age-related functional changes to the PRC in animals (rats and monkeys) resulting in impaired complex object discrimination and object recognition. To date, no studies have compared younger and older humans using paradigms previously shown to engage the PRC. To investigate the influence of age on complex object discrimination in humans, the present study used an object matching paradigm for blob-like objects that have previously been shown to recruit the PRC. Difficulty was manipulated by varying the number of overlapping features between objects. Functional MRI data was acquired to determine the involvement of the PRC in the two groups during complex object discrimination. Results indicated that while young and older adults performed similarly on the easy version of the task, most older adults were impaired relative to young participants when the number of overlapping features increased. fMRI results suggest that older adults do not engage bilateral anterior PRC to the same extent as young adults. Specifically, complex object matching performance in older adults was predicted by the degree to which they engage left anterior PRC. These results provide evidence for human age-related changes in PRC function that impact complex object discrimination.


Subject(s)
Aging/physiology , Cerebral Cortex/physiology , Discrimination, Psychological/physiology , Pattern Recognition, Visual/physiology , Photic Stimulation/methods , Psychomotor Performance/physiology , Adolescent , Adult , Aged , Female , Humans , Male , Young Adult
2.
Phys Med Biol ; 52(23): 7055-71, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-18029993

ABSTRACT

We previously developed a noninvasive technique for the quantification of fluorodeoxyglucose (FDG) positron emission tomography (PET) images using an image-derived input function obtained from a manually drawn carotid artery region. Here, we investigate the use of independent component analysis (ICA) for more objective identification of the carotid artery and surrounding tissue regions. Using FDG PET data from 22 subjects, ICA was applied to an easily defined cubical region including the carotid artery and neighboring tissue. Carotid artery and tissue time activity curves and three venous samples were used to generate spillover and partial volume-corrected input functions and to calculate the parametric images of the cerebral metabolic rate for glucose (CMRgl). Different from a blood-sampling-free ICA approach, the results from our ICA approach are numerically well matched to those based on the arterial blood sampled input function. In fact, the ICA-derived input functions and CMRgl measurements were not only highly correlated (correlation coefficients >0.99) to, but also highly comparable (regression slopes between 0.92 and 1.09), with those generated using arterial blood sampling. Moreover, the reliability of the ICA-derived input function remained high despite variations in the location and size of the cubical region. The ICA procedure makes it possible to quantify FDG PET images in an objective and reproducible manner.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Carotid Arteries/metabolism , Fluorodeoxyglucose F18/pharmacokinetics , Image Interpretation, Computer-Assisted/methods , Positron-Emission Tomography/methods , Adult , Brain/blood supply , Carotid Arteries/diagnostic imaging , Computer Simulation , Female , Humans , Male , Models, Neurological , Models, Statistical , Principal Component Analysis , Radiopharmaceuticals/pharmacokinetics
3.
Neurobiol Aging ; 27(11): 1604-12, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16226349

ABSTRACT

This study examined the functionality of the medial temporal lobe (MTL) and posterior cingulate (PC) in mild cognitive impairment amnestic type (MCI), a syndrome that puts patients at greater risk for developing Alzheimer disease (AD). Functional MRI (fMRI) was used to identify regions normally active during encoding of novel items and recognition of previously learned items in a reference group of 77 healthy young and middle-aged adults. The pattern of activation in this group guided further comparisons between 14 MCI subjects and 14 age-matched controls. The MCI patients exhibited less activity in the PC during recognition of previously learned items, and in the right hippocampus during encoding of novel items, despite comparable task performance to the controls. Reduced fMRI signal change in the MTL supports prior studies implicating the hippocampus for encoding new information. Reduced signal change in the PC converges with recent research on its role in recognition in normal adults as well as metabolic decline in people with genetic or cognitive risk for AD. Our results suggest that a change in function in the PC may account, in part, for memory recollection failure in AD.


Subject(s)
Alzheimer Disease/physiopathology , Cognition Disorders/physiopathology , Aged , Alzheimer Disease/psychology , Atrophy , Case-Control Studies , Cognition Disorders/pathology , Female , Gyrus Cinguli/physiopathology , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging/methods , Male , Matched-Pair Analysis , Memory , Temporal Lobe/pathology , Temporal Lobe/physiopathology
4.
Neurobiol Aging ; 24(1): 85-94, 2003.
Article in English | MEDLINE | ID: mdl-12493554

ABSTRACT

We used volumetric MRI and analysis of areas under receiver operating characteristic (ROC) curves to directly compare the extent of hippocampus-amygdala formation (HAF) and corpus callosum atrophy in patients with Alzheimer's disease (AD) in different clinical stages of dementia. Based on neuropathological studies, we hypothesized that HAF atrophy, representing allocortical neuronal degeneration, would precede atrophy of corpus callosum, representing loss of neocortical association neurons, in early AD. HAF and corpus callosum sizes were significantly reduced in 27 AD patients (37% and 16%, respectively) compared to 28 healthy controls. In mildly- and moderately-demented AD patients, the ROC derived index of atrophy was greater for HAF volume than for total corpus callosum area. The index of atrophy of posterior corpus callosum was not significantly different from HAF at mild, moderate or severe stages of dementia. In conclusion, these findings suggest a characteristic regional pattern of allocortical and neocortical neurodegeneraton in AD. Our data indicate that neuronal loss in parietotemporal cortex (represented by atrophy of corpus callosum splenium) may occur simultaneously with allocortical neurodegeneration in mild AD. Moreover, ROC analysis may provide a statistical framework to determine atrophy patterns of different brain structures in neurodegenerative diseases in vivo.


Subject(s)
Alzheimer Disease/pathology , Corpus Callosum/pathology , Hippocampus/pathology , Nerve Degeneration/physiopathology , Aged , Aged, 80 and over , Amygdala/pathology , Analysis of Variance , Area Under Curve , Atrophy/pathology , Case-Control Studies , Disease Progression , Female , Humans , Magnetic Resonance Imaging , Male , Matched-Pair Analysis , Middle Aged , Neocortex/pathology , Nerve Degeneration/pathology , Neuropsychological Tests , ROC Curve , Reference Values , Severity of Illness Index
5.
J Neural Transm (Vienna) ; 109(5-6): 837-55, 2002 May.
Article in English | MEDLINE | ID: mdl-12111472

ABSTRACT

Neuropathological studies in Alzheimer's disease (AD) indicate specific loss of layer III and V large pyramidal neurons in association cortex. These neurons give rise to long cortico-cortical connections, projecting through the corpus callosum, in an anterior-posterior topology. Based on these findings we hypothesized that regional corpus callosum atrophy may be a potential in vivo marker for neocortical neuronal loss in AD. To evaluate this hypothesis, we developed a method to measure cross-sectional area of the corpus callosum and of five corpus callosum subregions on midsagittal magnetic resonance imaging scans (MRI). In a subsequent series of six experimental studies using MRI, (18)FDG-PET and EEG, we investigated the relation of white matter hyperintensities (WMH) to corpus callosum size and correlated regional pattern of corpus callosum atrophy with regional cortical metabolic decline as well as intracortical coherencies. Mean total corpus callosum area was reduced significantly in AD patients compared to healthy age-matched controls, with the greatest changes in the rostrum and the splenium and relative sparing of the truncus. The regional pattern of corpus callosum atrophy was independent of WMH load and correlated significantly with pattern of regional metabolic decline measured with (18)FDG-PET, the degree of cognitive impairment and regional decline of bilateral intracortical-coherency in EEG in AD patients. We further found that hippocampus atrophy, as a marker of early allocortical degeneration, was more pronounced than total corpus callosum atrophy in mild stages of AD. Regional corpus callosum atrophy in mild disease, however, suggested early neocortical degeneration in AD. In a longitudinal study, AD patients showed significantly greater rates of corpus callosum atrophy than controls. Rates of atrophy correlated with progression of clinical dementia severity in AD. Our results indicate that regional corpus callosum atrophy in AD patients represents the loss of callosal efferent neurons in corresponding regions of the neocortex. As these neurons are a subset of cortico-cortical projecting neurons, region-specific corpus callosum atrophy may serve as a marker of progressive neocortical disconnection in AD. In combination with measurement of hippocampal atrophy, assessment of corpus callosum atrophy over time in individual patients is useful to evaluate effects on brain structure of currently developed drugs, thought to slow or modify AD progression.


Subject(s)
Alzheimer Disease/diagnosis , Neocortex/diagnostic imaging , Neocortex/pathology , Nerve Degeneration/diagnosis , Alzheimer Disease/psychology , Alzheimer Disease/therapy , Atrophy , Corpus Callosum/diagnostic imaging , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Disease Progression , Electroencephalography , Fluorodeoxyglucose F18 , Hippocampus/pathology , Humans , Longitudinal Studies , Magnetic Resonance Imaging , Neocortex/physiopathology , Neurons/pathology , Radiography , Radiopharmaceuticals , Severity of Illness Index , Tomography, Emission-Computed
6.
JAMA ; 286(17): 2120-7, 2001 Nov 07.
Article in English | MEDLINE | ID: mdl-11694153

ABSTRACT

CONTEXT: Deficits in cerebral glucose utilization have been identified in patients with cognitive dysfunction attributed to various disease processes, but their prognostic and diagnostic value remains to be defined. OBJECTIVE: To assess the sensitivity and specificity with which cerebral metabolic patterns at a single point in time forecast subsequent documentation of progressive dementia. DESIGN, SETTING, AND PATIENTS: Positron emission tomography (PET) studies of [(18)F]fluorodeoxyglucose in 146 patients undergoing evaluation for dementia with at least 2 years' follow-up for disease progression at the University of California, Los Angeles, from 1991 to 2000, and PET studies in 138 patients undergoing evaluation for dementia at an international consortium of facilities, with histopathological diagnoses an average of 2.9 years later, conducted from 1984 to 2000. MAIN OUTCOME MEASURES: Regional distribution of [(18)F]fluorodeoxyglucose in each patient, classified by criteria established a priori as positive or negative for presence of a progressive neurodegenerative disease in general and of Alzheimer disease (AD) specifically, compared with results of longitudinal or neuropathologic analyses. RESULTS: Progressive dementia was detected by PET with a sensitivity of 93% (191/206) and a specificity of 76% (59/78). Among patients with neuropathologically based diagnoses, PET identified patients with AD and patients with any neurodegenerative disease with a sensitivity of 94% and specificities of 73% and 78%, respectively. The negative likelihood ratio of experiencing a progressive vs nonprogressive course over the several years following a single negative brain PET scan was 0.10 (95% confidence interval, 0.06-0.16), and the initial pattern of cerebral metabolism was significantly associated with the subsequent course of progression overall (P<.001). CONCLUSION: In patients presenting with cognitive symptoms of dementia, regional brain metabolism was a sensitive indicator of AD and of neurodegenerative disease in general. A negative PET scan indicated that pathologic progression of cognitive impairment during the mean 3-year follow-up was unlikely to occur.


Subject(s)
Brain/metabolism , Dementia/diagnostic imaging , Glucose/metabolism , Tomography, Emission-Computed , Aged , Brain/diagnostic imaging , Dementia/diagnosis , Dementia/physiopathology , Disease Progression , Female , Fluorodeoxyglucose F18 , Humans , Likelihood Functions , Male , Middle Aged , Predictive Value of Tests , Prognosis , Radiopharmaceuticals , Sensitivity and Specificity
7.
J Neurol Sci ; 189(1-2): 93-8, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11535238

ABSTRACT

In a previous cross-sectional study of 100 asymptomatic individuals aged 49-69, we reported age-related decline in immediate and delayed memory that was steeper in apolipoprotein E (apoE)-e4/4 homozygotes than in members of other genetic subgroups. These findings were preliminarily based upon the statistical problem of multiple comparisons. We therefore sought to replicate these findings in a new cohort. From 1998 to 2000, 80 asymptomatic residents of Maricopa County, AZ were recruited through newspaper ads. 20 apoE-e4/4 homozygotes, 20 e3/4 heterozygotes, and 40 e4 noncarriers were matched (1:1:2) by age, gender, and years of education. All had normal neurologic and psychiatric examinations, including Folstein minimental status exam (MMSE) and Hamilton depression scale, and underwent a battery of neuropsychological tests identical to those in our previous study. The groups were well-matched for age (55.9+/-5.9 years), gender (60% women), and education (15.9+/-2.2 years), and were demographically similar to our previous cohort. Complex figure test recall was lower in e3/4 heterozygotes than noncarriers, but there was no significant difference between e4/4 homozygotes and noncarriers. There were no other significant differences in mean test scores between groups, but Wechsler adult intelligence scale-revised (WAIS-R) digit span showed a significant negative correlation with age in the e4/4 homozygote group relative to e4 noncarriers (p=0.008) as we had found in our previous study. In conclusion, we found a significant negative correlation of WAIS-R digit span with age in apoE-e4/4 homozygotes relative to e4 noncarriers in two separate cohorts, possibly reflecting an age-related effect on frontal lobe function in this genetic subgroup.


Subject(s)
Apolipoproteins E/genetics , Cognition Disorders/genetics , Age of Onset , Aged , Alzheimer Disease/epidemiology , Alzheimer Disease/genetics , Apolipoprotein E4 , Cohort Studies , Depression , Female , Genetic Predisposition to Disease , Heterozygote , Homozygote , Humans , Male , Memory , Middle Aged , Neuropsychological Tests
8.
Neurology ; 57(4): 626-32, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11524470

ABSTRACT

OBJECTIVE: (1)H-MRS studies have shown abnormalities in brain levels of myo-inositol (mI) and N-acetyl aspartate (NAA) in AD, but the relation of these abnormalities with dementia severity was not examined. The authors sought to determine whether altered brain levels of mI and other metabolites occur in mild AD and whether they change as dementia severity worsens. METHODS: The authors used (1)H-MRS with external standards to measure absolute brain concentrations of mI, NAA, total creatine (Cr), and choline (Cho)-containing compounds in 21 subjects with AD and 17 age- and sex-matched controls in occipital and left and right parietal regions. RESULTS: Concentrations of NAA were significantly decreased, whereas mI and Cr concentrations were significantly increased in all three brain regions in subjects with AD compared with controls. Higher concentrations of mI and Cr occurred even in mild AD. A discriminant analysis of the (1)H-MRS data combined with CSF volume measurements distinguished subjects with AD, ranging from mild to severe dementia, from controls with 100% correct classification. NAA concentration, though not other metabolites, was positively correlated with Mini-Mental State Examination score. CONCLUSION: The measurements with (1)H-MRS of absolute metabolite concentrations in the neocortex showed abnormal concentrations of brain metabolites in AD; these metabolite concentrations do not necessarily correlate with disease severity. Although changes in myo-inositol and creatine occur in the early stages of AD, abnormalities of N-acetyl aspartate do not occur in mild AD but progressively change with dementia severity. Further, subjects with mild AD can be differentiated from controls with (1)H-MRS.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/psychology , Analysis of Variance , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Choline/metabolism , Creatine/metabolism , Dementia/metabolism , Female , Humans , Hydrogen , Inositol/metabolism , Magnetic Resonance Imaging , Male , Middle Aged , Regression Analysis
9.
J Neurosci ; 21(8): 2919-28, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11306644

ABSTRACT

Reaching movements performed without vision of the moving limb are continuously monitored, during their execution, by feedback loops (designated nonvisual). In this study, we investigated the functional anatomy of these nonvisual loops using positron emission tomography (PET). Seven subjects had to "look at" (eye) or "look and point to" (eye-arm) visual targets whose location either remained stationary or changed undetectably during the ocular saccade (when vision is suppressed). Slightly changing the target location during gaze shift causes an increase in the amount of correction to be generated. Functional anatomy of nonvisual feedback loops was identified by comparing the reaching condition involving large corrections (jump) with the reaching condition involving small corrections (stationary), after subtracting the activations associated with saccadic movements and hand movement planning [(eye-arm-jumping minus eye-jumping) minus (eye-arm-stationary minus eye-stationary)]. Behavioral data confirmed that the subjects were both accurate at reaching to the stationary targets and able to update their movement smoothly and early in response to the target jump. PET difference images showed that these corrections were mediated by a restricted network involving the left posterior parietal cortex, the right anterior intermediate cerebellum, and the left primary motor cortex. These results are consistent with our knowledge of the functional properties of these areas and more generally with models emphasizing parietal-cerebellar circuits for processing a dynamic motor error signal.


Subject(s)
Arm/physiology , Brain/physiology , Feedback/physiology , Nerve Net/physiology , Psychomotor Performance/physiology , Adult , Behavior/physiology , Brain/anatomy & histology , Brain/diagnostic imaging , Cerebellum/anatomy & histology , Cerebellum/diagnostic imaging , Cerebellum/physiology , Cerebral Cortex/anatomy & histology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/physiology , Female , Fixation, Ocular/physiology , Hand/physiology , Humans , Male , Nerve Net/anatomy & histology , Nerve Net/diagnostic imaging , Parietal Lobe/anatomy & histology , Parietal Lobe/diagnostic imaging , Parietal Lobe/physiology , Reaction Time/physiology , Saccades/physiology , Tomography, Emission-Computed
10.
Arch Neurol ; 58(3): 480-6, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11255453

ABSTRACT

BACKGROUND: Brain glucose metabolic rates measured by positron emission tomography can be more affected by partial volume effects in Alzheimer disease (AD) than in healthy aging because of disease-associated brain atrophy. OBJECTIVE: To determine whether the distinct distribution of cerebral metabolic lesions in patients with the visual variant of AD (AD + VS) represents a true index of neuronal/synaptic dysfunction or is the consequence of brain atrophy. SETTING: Government research hospital. DESIGN: Resting cerebral metabolic rate for glucose was measured with positron emission tomography in a cross-sectional study of AD and AD + VS groups and in healthy control subjects. Segmented magnetic resonance images were used to correct for brain atrophy. PATIENTS: Patients with AD + VS had prominent visual and visuospatial symptoms. There were 15 patients with AD, 10 with AD + VS, and 37 age-matched control subjects. MAIN OUTCOME MEASURE: Measurement of the rate of cerebral glucose metabolism. RESULTS: Before atrophy correction, the AD + VS group, compared with the control subjects, showed hypometabolism in primary and extrastriate visual areas and in parietal and superior temporal cortical areas. Compared with the AD group, the AD + VS group showed hypometabolism in visual association areas. After atrophy correction, hypometabolism remained significantly different between patients and controls and between the 2 AD groups. CONCLUSIONS: The reductions in cerebral hypometabolism represent a true loss of functional activity and are not simply an artifact caused by brain atrophy. The different patterns of hypometabolism indicate the differential development of the lesions between the AD and AD + VS groups.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Energy Metabolism , Visual Cortex/metabolism , Visual Cortex/pathology , Aged , Alzheimer Disease/diagnostic imaging , Atrophy , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Glucose/metabolism , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Parietal Lobe/metabolism , Parietal Lobe/pathology , Temporal Lobe/metabolism , Temporal Lobe/pathology , Tomography, Emission-Computed
11.
Proc Natl Acad Sci U S A ; 98(6): 3334-9, 2001 Mar 13.
Article in English | MEDLINE | ID: mdl-11248079

ABSTRACT

Cross-sectional positron emission tomography (PET) studies find that cognitively normal carriers of the apolipoprotein E (APOE) epsilon4 allele, a common Alzheimer's susceptibility gene, have abnormally low measurements of the cerebral metabolic rate for glucose (CMRgl) in the same regions as patients with Alzheimer's dementia. In this article, we characterize longitudinal CMRgl declines in cognitively normal epsilon4 heterozygotes, estimate the power of PET to test the efficacy of treatments to attenuate these declines in 2 years, and consider how this paradigm could be used to efficiently test the potential of candidate therapies for the prevention of Alzheimer's disease. We studied 10 cognitively normal epsilon4 heterozygotes and 15 epsilon4 noncarriers 50-63 years of age with a reported family history of Alzheimer's dementia before and after an interval of approximately 2 years. The epsilon4 heterozygotes had significant CMRgl declines in the vicinity of temporal, posterior cingulate, and prefrontal cortex, basal forebrain, parahippocampal gyrus, and thalamus, and these declines were significantly greater than those in the epsilon4 noncarriers. In testing candidate primary prevention therapies, we estimate that between 50 and 115 cognitively normal epsilon4 heterozygotes are needed per active and placebo treatment group to detect a 25% attenuation in these CMRgl declines with 80% power and P = 0.005 in 2 years. Assuming these CMRgl declines are related to the predisposition to Alzheimer's dementia, this study provides a paradigm for testing the potential of treatments to prevent the disorder without having to study thousands of research subjects or wait many years to determine whether or when treated individuals develop symptoms.


Subject(s)
Alzheimer Disease/genetics , Apolipoproteins E/genetics , Brain/physiopathology , Cognition/physiology , Heterozygote , Tomography, Emission-Computed/methods , Aged , Alzheimer Disease/metabolism , Apolipoprotein E4 , Brain/metabolism , Female , Glucose/metabolism , Humans , Male , Middle Aged
12.
Neurobiol Aging ; 21(4): 577-84, 2000.
Article in English | MEDLINE | ID: mdl-10924775

ABSTRACT

To assess age-related differences in cortical activation during form perception, two classes of visual textures were shown to young and older subjects undergoing positron emission tomography (PET). Subjects viewed even textures that were rich in rectangular blocks and extended contours and random textures that lacked these organized form elements. Within-group significant increases in regional cerebral blood flow (rCBF) during even stimulation relative to random stimulation in young subjects were seen in occipital, inferior and medial temporal regions, and cerebellum, and in older subjects, in posterior occipital and frontal regions. Group by texture type interactions revealed significantly smaller rCBF increases in older subjects relative to young in occipital and medial temporal regions. These results indicate that young subjects activate the occipitotemporal pathway during form perception, whereas older subjects activate occipital and frontal regions. The between-group differences suggest that age-related reorganization of cortical activation occur during early visual processes in humans.


Subject(s)
Aging/physiology , Tomography, Emission-Computed , Visual Perception/physiology , Adult , Aged , Female , Humans , Male , Middle Aged , Parietal Lobe/physiology , Photic Stimulation , Visual Cortex/physiology , Visual Pathways/physiology
13.
Pharmacol Biochem Behav ; 66(3): 475-81, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10899358

ABSTRACT

In imaging studies of brain functions using pharmacological probes, identification of the time point at which central effects of intravenously infused drugs become stable is crucial to separate the effects of experimental variables from the concomitant changes in drug effects over time. We evaluated the time courses of the pharmacokinetics and pharmacodynamics, including butyrylcholinesterase inhibition and central neural responses, of physostigmine in healthy young subjects. Ten positron emission tomography (PET) scans that alternated between a rest condition (eyes open, ears unplugged) and a working memory for faces (WM) task were acquired in healthy subjects. Subjects in the drug group received a saline infusion for the first two scans, providing a baseline measure, then received an infusion of physostigmine for all subsequent scans. Subjects in the control group received a placebo infusion of saline for all scans. Physostigmine plasma levels and percent butyrylcholinesterase inhibition increased over time (p < 0. 0001), and both became stable by 40 min. Physostigmine decreased reaction time (RT) (p = 0.0005), and this effect was detected after 20 min of infusion and stable thereafter. Physostigmine also decreased regional cerebral blood flow (rCBF) in right prefrontal cortex during task (p = 0.0002), and this effect was detected after 40 min of infusion and stable thereafter. No change in RT or rCBF was observed in the control group. These results indicate that a 40-min infusion of physostigmine was necessary to obtain stable central effects. More generally, we have demonstrated that experimental effects can vary with time, especially during the initial phases of a drug infusion, indicating that it is critical that these changes are controlled.


Subject(s)
Brain/metabolism , Physostigmine/pharmacokinetics , Adult , Analysis of Variance , Brain/diagnostic imaging , Brain/drug effects , Cholinesterase Inhibitors/pharmacokinetics , Cholinesterase Inhibitors/pharmacology , Cognition/drug effects , Female , Humans , Male , Middle Aged , Physostigmine/pharmacology , Psychomotor Performance/drug effects , Time Factors , Tomography, Emission-Computed
14.
Int J Psychophysiol ; 37(1): 87-98, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10828377

ABSTRACT

Alzheimer's disease, the most common form of dementia in the elderly, is characterized by the progressive, global and irreversible deterioration of cognitive abilities. The development of positron emission tomography (PET) methodologies has made it possible to study the in vivo brain metabolic correlates of human cognitive and behavioral functions. Moreover, as PET scan examinations can be repeated, the progression of the neuropathological process and its relation to cognitive dysfunction can be followed over time. In an effort to understand the changes in neural function that precede and accompany onset of dementia and their relation to clinical manifestations, in the last several years, we have conducted clinical, neuropsychological and brain metabolic studies in groups of Alzheimer's disease patients at different stages of dementia severity or with distinct clinical pictures and in populations at risk for developing the disease. Here, we discuss the main findings and implications obtained from these studies.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Brain/metabolism , Cognition Disorders/metabolism , Tomography, Emission-Computed , Alzheimer Disease/diagnostic imaging , Cognition Disorders/diagnostic imaging , Dementia/psychology , Glucose/metabolism , Humans
15.
Exp Brain Res ; 132(2): 243-59, 2000 May.
Article in English | MEDLINE | ID: mdl-10853949

ABSTRACT

It is known that the saccadic system shows adaptive changes when the command sent to the extraocular muscles is inappropriate. Despite an abundance of supportive psychophysical investigations, the neurophysiological substrate of this process is still debated. The present study addresses this issue using H2(15)O positron emission tomography (PET). We contrasted three conditions in which healthy human subjects were required to perform saccadic eye movements toward peripheral visual targets. Two conditions involved a modification of the target location during the course of the initial saccade, when there is suppression of visual perception. In the RAND condition, intra-saccadic target displacement was random from trial-to-trial, precluding any systematic modification of the primary saccade amplitude. In the ADAPT condition, intra-saccadic target displacement was uniform, causing adaptive modification of the primary saccade amplitude. In the third condition (stationary, STAT), the target remained at the same location during the entire trial. Difference images reflecting regional cerebral-blood-flow changes attributable to the process of saccadic adaptation (ADAPT minus RAND; ADAPT minus STAT) showed a selective activation in the oculomotor cerebellar vermis (OCV; lobules VI and VII). This finding is consistent with neurophysiological studies in monkeys. Additional analyses indicated that the cerebellar activation was not related to kinematic factors, and that the absence of significant activation within the frontal eye fields (FEF) or the superior colliculus (SC) did not represent a false negative inference. Besides the contribution of the OCV to saccadic adaptation, we also observed, in the RAND condition, that the saccade amplitude was significantly larger when the previous trial involved a forward jump than when the previous trial involved a backward jump. This observation indicates that saccade accuracy is constantly monitored on a trial-to-trial basis. Behavioral measurements and PET observations (RAND minus STAT) suggest that this single-trial control of saccade amplitude may be functionally distinct from the process of saccadic adaptation.


Subject(s)
Adaptation, Physiological/physiology , Saccades/physiology , Tomography, Emission-Computed , Adult , Biomechanical Phenomena , Cerebellum/diagnostic imaging , Cerebellum/physiology , Female , Humans , Male , Middle Aged , Superior Colliculi/diagnostic imaging , Superior Colliculi/physiology
16.
Neuropsychologia ; 38(8): 1126-35, 2000.
Article in English | MEDLINE | ID: mdl-10838147

ABSTRACT

A cued visual search task was used to examine the dynamic range over which spatial attention affects target identification during visual search. Precues varied in validity (valid, invalid, or neutral) and in precision (cue size) of target localization. Participants were "young-old" (65-74 years) and "old-old" (75-85 years) elderly adults and individuals in the mild stage of dementia of the Alzheimer type (DAT). For all participants, search was speeded as the precision with which a precue surrounding the location of a subsequently appearing target increased (precue size decreased). The cue size effect was evident in both feature and conjunction search, but was greatly reduced in both old-old and DAT groups compared to the young-old. However, whereas all non-demented adults showed a progressive modulation of search efficiency over the entire range of cue sizes, the dynamic range of spatial attention was restricted to the most precise cue in the DAT group. The restriction in the dynamic range of spatial attention may represent an underlying component of the impairment in perceptual and memory functioning found in early-stage DAT.


Subject(s)
Alzheimer Disease/diagnosis , Attention , Orientation , Pattern Recognition, Visual , Adult , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Attention/physiology , Color Perception/physiology , Female , Humans , Male , Orientation/physiology , Parietal Lobe/physiopathology , Pattern Recognition, Visual/physiology , Reaction Time/physiology , Reference Values , Size Perception/physiology
18.
J Nucl Med ; 41(4): 575-83, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10768555

ABSTRACT

UNLABELLED: Alzheimer's disease is associated with reductions in resting-state brain metabolism, as measured by PET, progressing with dementia severity. The purpose of this study was to see to what extent brain regions with reduced resting-state metabolic rates in Alzheimer patients could be activated by a passive audiovisual stimulation test and to compare the result with activation in age-matched healthy volunteers. The extent of activation in Alzheimer's disease is considered to reflect the integrity of synaptic function, or inherent viability, and the potential responsiveness of the Alzheimer brain to drug therapy. METHODS: Regional cerebral metabolic rates for glucose (rCMRglc, in mg/ 100 g tissue/min) were measured in the resting state (eyes and ears covered) and during passive audiovisual stimulation (watching a movie) in 15 otherwise healthy Alzheimer patients of differing dementia severity (Mattis Dementia Rating Scale score, 23-128) and in 14 age-matched healthy volunteers (score, 141 +/- 3) using PET with 2 sequential injections of FDG. RESULTS: In the volunteers, audiovisual stimulation caused significant rCMRglc increases in visual and auditory cortical areas but significant decreases in frontal areas. In the mildly demented patients, rCMRglc responses were within 2 SDs of the mean in volunteers. However, the magnitude of the rCMRglc responses during stimulation declined significantly with dementia severity in the right occipitotemporal, right and left occipital association, and left calcarine cortical regions. CONCLUSION: Functional brain responsiveness, evaluated by a passive audiovisual stimulation paradigm with PET, is within normal limits in mildly demented Alzheimer patients but fails with worsening dementia severity. Declining responsiveness may account for the limited success of neurotransmitter replacement therapy in Alzheimer patients with moderate-to-severe dementia.


Subject(s)
Alzheimer Disease/diagnostic imaging , Brain/metabolism , Tomography, Emission-Computed , Acoustic Stimulation , Aged , Alzheimer Disease/physiopathology , Brain/diagnostic imaging , Case-Control Studies , Female , Fluorine Radioisotopes , Fluorodeoxyglucose F18 , Glucose/metabolism , Humans , Male , Photic Stimulation , Radiopharmaceuticals
19.
Brain Res Bull ; 51(3): 213-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10718513

ABSTRACT

Previously, we have shown that physostigmine, an acetylcholinesterase inhibitor, improved performance on a working memory for faces task, as reflected by reduced reaction time (RT), and reduced task-specific regional cerebral blood flow (rCBF) in right prefrontal cortex and, further, that these reductions in RT and right frontal rCBF were significantly correlated. Here we investigated the relation between the effects of physostigmine on task performance and task-specific functional brain response throughout the cortex by examining correlations between physostigmine-related changes in rCBF in all brain areas and changes in RT. In subjects who received an infusion of physostigmine, reduced RT correlated (p<0.001) positively with reduced rCBF in right frontal cortex, left temporal cortex, anterior cingulate, and left hippocampus; and correlated with increased rCBF in medial occipital visual cortex. In subjects who received a placebo infusion of saline, no significant correlations between changes in RT and cortical rCBF were observed. The results show that cholinergically induced improvements in working memory performance are related to alterations in neural activity in multiple cortical regions, including increased neural activity in regions associated with early perceptual processing and decreased neural activity in regions associated with attention, memory encoding, and memory maintenance.


Subject(s)
Brain/diagnostic imaging , Brain/physiology , Cerebrovascular Circulation/drug effects , Cholinesterase Inhibitors/pharmacology , Memory/drug effects , Physostigmine/pharmacology , Tomography, Emission-Computed , Adult , Aged , Brain/drug effects , Brain Mapping , Female , Humans , Male , Middle Aged , Reaction Time/drug effects , Reference Values
20.
Am J Psychiatry ; 156(12): 1879-86, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10588400

ABSTRACT

OBJECTIVE: An extra portion of chromosome 21 in Down's syndrome leads to a dementia in later life that is phenotypically similar to Alzheimer's disease. Down's syndrome therefore represents a model for studying preclinical stages of Alzheimer's disease. Markers that have been investigated in symptomatic Alzheimer's disease are myoinositol and N-acetyl-aspartate. The authors investigated whether abnormal brain levels of myo-inositol and other metabolites occur in the preclinical stages of Alzheimer's disease associated with Down's syndrome. METHOD: The authors used 1H magnetic resonance spectroscopy (MRS) with external standards to measure absolute brain metabolite concentrations in 19 nondemented adults with Down's syndrome and 17 age- and sex-matched healthy comparison subjects. RESULTS: Concentrations of myoinositol and choline-containing compounds were significantly higher in the occipital and parietal regions of the adults with Down's syndrome than in the comparison subjects. Within the Down's syndrome group, older subjects (42-62 years, N = 11) had higher myo-inositol levels than younger subjects (28-39 years, N = 8). Older subjects in both groups had lower N-acetylaspartate levels than the respective younger subjects, although this old-young difference was not greater in the Down's syndrome group. CONCLUSIONS: The approximately 50% higher level of myo-inositol in Down's syndrome suggests a gene dose effect of the extra chromosome 21, where the human osmoregulatory sodium/myo-inositol cotransporter gene is located. The even higher myoinositol level in older adults with Down's syndrome extends to the predementia phase earlier findings of high myoinositol levels in symptomatic Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Brain Chemistry , Brain/metabolism , Down Syndrome/metabolism , Inositol/analysis , Magnetic Resonance Spectroscopy , Adult , Age Factors , Alzheimer Disease/diagnosis , Biomarkers , Diagnosis, Differential , Down Syndrome/diagnosis , Female , Humans , Inositol/metabolism , Magnetic Resonance Spectroscopy/statistics & numerical data , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...