Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.896
1.
BMC Med Imaging ; 24(1): 103, 2024 May 03.
Article En | MEDLINE | ID: mdl-38702626

OBJECTIVE: This study aimed to identify features of white matter network attributes based on diffusion tensor imaging (DTI) that might lead to progression from mild cognitive impairment (MCI) and construct a comprehensive model based on these features for predicting the population at high risk of progression to Alzheimer's disease (AD) in MCI patients. METHODS: This study enrolled 121 MCI patients from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Among them, 36 progressed to AD after four years of follow-up. A brain network was constructed for each patient based on white matter fiber tracts, and network attribute features were extracted. White matter network features were downscaled, and white matter markers were constructed using an integrated downscaling approach, followed by forming an integrated model with clinical features and performance evaluation. RESULTS: APOE4 and ADAS scores were used as independent predictors and combined with white matter network markers to construct a comprehensive model. The diagnostic efficacy of the comprehensive model was 0.924 and 0.919, sensitivity was 0.864 and 0.900, and specificity was 0.871 and 0.815 in the training and test groups, respectively. The Delong test showed significant differences (P < 0.05) in the diagnostic efficacy of the combined model and APOE4 and ADAS scores, while there was no significant difference (P > 0.05) between the combined model and white matter network biomarkers. CONCLUSIONS: A comprehensive model constructed based on white matter network markers can identify MCI patients at high risk of progression to AD and provide an adjunct biomarker helpful in early AD detection.


Alzheimer Disease , Cognitive Dysfunction , Diffusion Tensor Imaging , Disease Progression , White Matter , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/pathology , White Matter/diagnostic imaging , White Matter/pathology , Diffusion Tensor Imaging/methods , Female , Male , Aged , Aged, 80 and over , Sensitivity and Specificity , Apolipoprotein E4/genetics
2.
Alzheimers Res Ther ; 16(1): 97, 2024 May 03.
Article En | MEDLINE | ID: mdl-38702802

BACKGROUND: The locus coeruleus (LC) and the nucleus basalis of Meynert (NBM) are altered in early stages of Alzheimer's disease (AD). Little is known about LC and NBM alteration in limbic-predominant age-related TDP-43 encephalopathy (LATE) and frontotemporal dementia (FTD). The aim of the present study is to investigate in vivo LC and NBM integrity in patients with suspected-LATE, early-amnestic AD and FTD in comparison with controls. METHODS: Seventy-two participants (23 early amnestic-AD patients, 17 suspected-LATE, 17 FTD patients, defined by a clinical-biological diagnosis reinforced by amyloid and tau PET imaging, and 15 controls) underwent neuropsychological assessment and 3T brain MRI. We analyzed the locus coeruleus signal intensity (LC-I) and the NBM volume as well as their relation with cognition and with medial temporal/cortical atrophy. RESULTS: We found significantly lower LC-I and NBM volume in amnestic-AD and suspected-LATE in comparison with controls. In FTD, we also observed lower NBM volume but a slightly less marked alteration of the LC-I, independently of the temporal or frontal phenotype. NBM volume was correlated with the global cognitive efficiency in AD patients. Strong correlations were found between NBM volume and that of medial temporal structures, particularly the amygdala in both AD and FTD patients. CONCLUSIONS: The alteration of LC and NBM in amnestic-AD, presumed-LATE and FTD suggests a common vulnerability of these structures to different proteinopathies. Targeting the noradrenergic and cholinergic systems could be effective therapeutic strategies in LATE and FTD.


Alzheimer Disease , Basal Nucleus of Meynert , Frontotemporal Dementia , Locus Coeruleus , Magnetic Resonance Imaging , Humans , Frontotemporal Dementia/diagnostic imaging , Frontotemporal Dementia/pathology , Male , Locus Coeruleus/diagnostic imaging , Locus Coeruleus/pathology , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Female , Aged , Magnetic Resonance Imaging/methods , Basal Nucleus of Meynert/diagnostic imaging , Basal Nucleus of Meynert/pathology , Middle Aged , Neuropsychological Tests , Amnesia/diagnostic imaging , Positron-Emission Tomography/methods
3.
Hum Brain Mapp ; 45(7): e26689, 2024 May.
Article En | MEDLINE | ID: mdl-38703095

Tau pathology and its spatial propagation in Alzheimer's disease (AD) play crucial roles in the neurodegenerative cascade leading to dementia. However, the underlying mechanisms linking tau spreading to glucose metabolism remain elusive. To address this, we aimed to examine the association between pathologic tau aggregation, functional connectivity, and cascading glucose metabolism and further explore the underlying interplay mechanisms. In this prospective cohort study, we enrolled 79 participants with 18F-Florzolotau positron emission tomography (PET), 18F-fluorodeoxyglucose PET, resting-state functional, and anatomical magnetic resonance imaging (MRI) images in the hospital-based Shanghai Memory Study. We employed generalized linear regression and correlation analyses to assess the associations between Florzolotau accumulation, functional connectivity, and glucose metabolism in whole-brain and network-specific manners. Causal mediation analysis was used to evaluate whether functional connectivity mediates the association between pathologic tau and cascading glucose metabolism. We examined 22 normal controls and 57 patients with AD. In the AD group, functional connectivity was associated with Florzolotau covariance (ß = .837, r = 0.472, p < .001) and glucose covariance (ß = 1.01, r = 0.499, p < .001). Brain regions with higher tau accumulation tend to be connected to other regions with high tau accumulation through functional connectivity or metabolic connectivity. Mediation analyses further suggest that functional connectivity partially modulates the influence of tau accumulation on downstream glucose metabolism (mediation proportion: 49.9%). Pathologic tau may affect functionally connected neurons directly, triggering downstream glucose metabolism changes. This study sheds light on the intricate relationship between tau pathology, functional connectivity, and downstream glucose metabolism, providing critical insights into AD pathophysiology and potential therapeutic targets.


Alzheimer Disease , Fluorodeoxyglucose F18 , Magnetic Resonance Imaging , Nerve Net , Positron-Emission Tomography , tau Proteins , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Male , Female , Aged , tau Proteins/metabolism , Middle Aged , Nerve Net/diagnostic imaging , Nerve Net/metabolism , Nerve Net/physiopathology , Glucose/metabolism , Connectome , Prospective Studies , Brain/diagnostic imaging , Brain/metabolism , Brain/physiopathology , Aged, 80 and over
4.
Alzheimers Res Ther ; 16(1): 99, 2024 May 04.
Article En | MEDLINE | ID: mdl-38704569

BACKGROUND: Patients with sporadic cerebral amyloid angiopathy (sCAA) frequently report cognitive or neuropsychiatric symptoms. The aim of this study is to investigate whether in patients with sCAA, cognitive impairment and neuropsychiatric symptoms are associated with a cerebrospinal fluid (CSF) biomarker profile associated with Alzheimer's disease (AD). METHODS: In this cross-sectional study, we included participants with sCAA and dementia- and stroke-free, age- and sex-matched controls, who underwent a lumbar puncture, brain MRI, cognitive assessments, and self-administered and informant-based-questionnaires on neuropsychiatric symptoms. CSF phosphorylated tau, total tau and Aß42 levels were used to divide sCAA patients in two groups: CAA with (CAA-AD+) or without a CSF biomarker profile associated with AD (CAA-AD-). Performance on global cognition, specific cognitive domains (episodic memory, working memory, processing speed, verbal fluency, visuoconstruction, and executive functioning), presence and severity of neuropsychiatric symptoms, were compared between groups. RESULTS: sCAA-AD+ (n=31; mean age: 72 ± 6; 42%, 61% female) and sCAA-AD- (n=23; 70 ± 5; 42% female) participants did not differ with respect to global cognition or type of affected cognitive domain(s). The number or severity of neuropsychiatric symptoms also did not differ between sCAA-AD+ and sCAA-AD- participants. These results did not change after exclusion of patients without prior ICH. CONCLUSIONS: In participants with sCAA, a CSF biomarker profile associated with AD does not impact global cognition or specific cognitive domains, or the presence of neuropsychiatric symptoms.


Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Cerebral Amyloid Angiopathy , Neuropsychological Tests , tau Proteins , Humans , Female , Male , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/complications , Alzheimer Disease/diagnostic imaging , Aged , Cross-Sectional Studies , Cerebral Amyloid Angiopathy/cerebrospinal fluid , Cerebral Amyloid Angiopathy/complications , Cerebral Amyloid Angiopathy/diagnostic imaging , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/etiology , Peptide Fragments/cerebrospinal fluid , Cognition/physiology , Middle Aged , Magnetic Resonance Imaging
5.
Brain Behav ; 14(5): e3533, 2024 May.
Article En | MEDLINE | ID: mdl-38715429

AIM: Although there exists substantial epidemiological evidence indicating an elevated risk of dementia in individuals with diabetes, our understanding of the neuropathological underpinnings of the association between Type-2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) remains unclear. This study aims to unveil the microstructural brain changes associated with T2DM in AD and identify the clinical variables contributing to these changes. METHODS: In this retrospective study involving 64 patients with AD, 31 individuals had concurrent T2DM. The study involved a comparative analysis of diffusion tensor imaging (DTI) images and clinical features between patients with and without T2DM. The FSL FMRIB software library was used for comprehensive preprocessing and tractography analysis of DTI data. After eddy current correction, the "bedpost" model was utilized to model diffusion parameters. Linear regression analysis with a stepwise method was used to predict the clinical variables that could lead to microstructural white matter changes. RESULTS: We observed a significant impairment in the left superior longitudinal fasciculus (SLF) among patients with AD who also had T2DM. This impairment in patients with AD and T2DM was associated with an elevation in creatine levels. CONCLUSION: The white matter microstructure in the left SLF appears to be sensitive to the impairment of kidney function associated with T2DM in patients with AD. The emergence of AD in association with T2DM may be driven by mechanisms distinct from the typical AD pathology. Compromised renal function in AD could potentially contribute to impaired white matter integrity.


Alzheimer Disease , Diabetes Mellitus, Type 2 , Diffusion Tensor Imaging , White Matter , Humans , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , White Matter/diagnostic imaging , White Matter/pathology , Male , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnostic imaging , Female , Aged , Retrospective Studies , Brain/diagnostic imaging , Brain/pathology , Middle Aged , Aged, 80 and over , Creatine/metabolism
6.
Sci Rep ; 14(1): 10083, 2024 05 02.
Article En | MEDLINE | ID: mdl-38698190

Differentiating clinical stages based solely on positive findings from amyloid PET is challenging. We aimed to investigate the neuroanatomical characteristics at the whole-brain level that differentiate prodromal Alzheimer's disease (AD) from cognitively unimpaired amyloid-positive individuals (CU A+) in relation to amyloid deposition and regional atrophy. We included 45 CU A+ participants and 135 participants with amyloid-positive prodromal AD matched 1:3 by age, sex, and education. All participants underwent 18F-florbetaben positron emission tomography and 3D structural T1-weighted magnetic resonance imaging. We compared the standardized uptake value ratios (SUVRs) and volumes in 80 regions of interest (ROIs) between CU A+ and prodromal AD groups using independent t-tests, and employed the least absolute selection and shrinkage operator (LASSO) logistic regression model to identify ROIs associated with prodromal AD in relation to amyloid deposition, regional atrophy, and their interaction. After applying False Discovery Rate correction at < 0.1, there were no differences in global and regional SUVR between CU A+ and prodromal AD groups. Regional volume differences between the two groups were observed in the amygdala, hippocampus, entorhinal cortex, insula, parahippocampal gyrus, and inferior temporal and parietal cortices. LASSO logistic regression model showed significant associations between prodromal AD and atrophy in the entorhinal cortex, inferior parietal cortex, both amygdalae, and left hippocampus. The mean SUVR in the right superior parietal cortex (beta coefficient = 0.0172) and its interaction with the regional volume (0.0672) were also selected in the LASSO model. The mean SUVR in the right superior parietal cortex was associated with an increased likelihood of prodromal AD (Odds ratio [OR] 1.602, p = 0.014), particularly in participants with lower regional volume (OR 3.389, p < 0.001). Only regional volume differences, not amyloid deposition, were observed between CU A+ and prodromal AD. The reduced volume in the superior parietal cortex may play a significant role in the progression to prodromal AD through its interaction with amyloid deposition in that region.


Alzheimer Disease , Aniline Compounds , Magnetic Resonance Imaging , Positron-Emission Tomography , Prodromal Symptoms , Stilbenes , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Male , Female , Aged , Magnetic Resonance Imaging/methods , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Middle Aged , Atrophy , Amyloid beta-Peptides/metabolism , Cognition , Aged, 80 and over , Amyloid/metabolism
7.
Transl Psychiatry ; 14(1): 215, 2024 May 28.
Article En | MEDLINE | ID: mdl-38806463

Previous observational investigations suggest that structural and diffusion imaging-derived phenotypes (IDPs) are associated with major neurodegenerative diseases; however, whether these associations are causal remains largely uncertain. Herein we conducted bidirectional two-sample Mendelian randomization analyses to infer the causal relationships between structural and diffusion IDPs and major neurodegenerative diseases using common genetic variants-single nucleotide polymorphism (SNPs) as instrumental variables. Summary statistics of genome-wide association study (GWAS) for structural and diffusion IDPs were obtained from 33,224 individuals in the UK Biobank cohort. Summary statistics of GWAS for seven major neurodegenerative diseases were obtained from the largest GWAS for each disease to date. The forward MR analyses identified significant or suggestively statistical causal effects of genetically predicted three structural IDPs on Alzheimer's disease (AD), frontotemporal dementia (FTD), and multiple sclerosis. For example, the reduction in the surface area of the left superior temporal gyrus was associated with a higher risk of AD. The reverse MR analyses identified significantly or suggestively statistical causal effects of genetically predicted AD, Lewy body dementia (LBD), and FTD on nine structural and diffusion IDPs. For example, LBD was associated with increased mean diffusivity in the right superior longitudinal fasciculus and AD was associated with decreased gray matter volume in the right ventral striatum. Our findings might contribute to shedding light on the prediction and therapeutic intervention for the major neurodegenerative diseases at the neuroimaging level.


Alzheimer Disease , Frontotemporal Dementia , Genome-Wide Association Study , Mendelian Randomization Analysis , Neurodegenerative Diseases , Phenotype , Polymorphism, Single Nucleotide , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/diagnostic imaging , Frontotemporal Dementia/genetics , Frontotemporal Dementia/diagnostic imaging , Frontotemporal Dementia/pathology , Male , Female , Diffusion Magnetic Resonance Imaging , Multiple Sclerosis/genetics , Multiple Sclerosis/diagnostic imaging , Brain/diagnostic imaging , Brain/pathology , Aged , Lewy Body Disease/genetics , Lewy Body Disease/diagnostic imaging , Middle Aged , Magnetic Resonance Imaging , United Kingdom
8.
Sci Rep ; 14(1): 12276, 2024 May 29.
Article En | MEDLINE | ID: mdl-38806509

Alzheimer's disease (AD) accounts for 60-70% of the population with dementia. Mild cognitive impairment (MCI) is a diagnostic entity defined as an intermediate stage between subjective cognitive decline and dementia, and about 10-15% of people annually convert to AD. We aimed to investigate the most robust model and modality combination by combining multi-modality image features based on demographic characteristics in six machine learning models. A total of 196 subjects were enrolled from four hospitals and the Alzheimer's Disease Neuroimaging Initiative dataset. During the four-year follow-up period, 47 (24%) patients progressed from MCI to AD. Volumes of the regions of interest, white matter hyperintensity, and regional Standardized Uptake Value Ratio (SUVR) were analyzed using T1, T2-weighted-Fluid-Attenuated Inversion Recovery (T2-FLAIR) MRIs, and amyloid PET (αPET), along with automatically provided hippocampal occupancy scores (HOC) and Fazekas scales. As a result of testing the robustness of the model, the GBM model was the most stable, and in modality combination, model performance was further improved in the absence of T2-FLAIR image features. Our study predicts the probability of AD conversion in MCI patients, which is expected to be useful information for clinician's early diagnosis and treatment plan design.


Alzheimer Disease , Cognitive Dysfunction , Disease Progression , Machine Learning , Magnetic Resonance Imaging , Positron-Emission Tomography , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/diagnosis , Female , Male , Aged , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/diagnosis , Magnetic Resonance Imaging/methods , Positron-Emission Tomography/methods , Aged, 80 and over , Neuroimaging/methods , Dementia/diagnostic imaging , Dementia/diagnosis
9.
BMC Med Inform Decis Mak ; 24(Suppl 1): 61, 2024 May 28.
Article En | MEDLINE | ID: mdl-38807132

BACKGROUND: Alzheimer's Disease (AD) is a progressive memory disorder that causes irreversible cognitive decline. Given that there is currently no cure, it is critical to detect AD in its early stage during the disease progression. Recently, many statistical learning methods have been presented to identify cognitive decline with temporal data, but few of these methods integrate heterogeneous phenotype and genetic information together to improve the accuracy of prediction. In addition, many of these models are often unable to handle incomplete temporal data; this often manifests itself in the removal of records to ensure consistency in the number of records across participants. RESULTS: To address these issues, in this work we propose a novel approach to integrate the genetic data and the longitudinal phenotype data to learn a fixed-length "enriched" biomarker representation derived from the temporal heterogeneous neuroimaging records. Armed with this enriched representation, as a fixed-length vector per participant, conventional machine learning models can be used to predict clinical outcomes associated with AD. CONCLUSION: The proposed method shows improved prediction performance when applied to data derived from Alzheimer's Disease Neruoimaging Initiative cohort. In addition, our approach can be easily interpreted to allow for the identification and validation of biomarkers associated with cognitive decline.


Alzheimer Disease , Cognitive Dysfunction , Neuroimaging , Humans , Cognitive Dysfunction/genetics , Cognitive Dysfunction/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/diagnostic imaging , Aged , Longitudinal Studies , Supervised Machine Learning , Female , Male , Machine Learning
10.
Proc Natl Acad Sci U S A ; 121(22): e2322617121, 2024 May 28.
Article En | MEDLINE | ID: mdl-38771873

Optimal decision-making balances exploration for new information against exploitation of known rewards, a process mediated by the locus coeruleus and its norepinephrine projections. We predicted that an exploitation-bias that emerges in older adulthood would be associated with lower microstructural integrity of the locus coeruleus. Leveraging in vivo histological methods from quantitative MRI-magnetic transfer saturation-we provide evidence that older age is associated with lower locus coeruleus integrity. Critically, we demonstrate that an exploitation bias in older adulthood, assessed with a foraging task, is sensitive and specific to lower locus coeruleus integrity. Because the locus coeruleus is uniquely vulnerable to Alzheimer's disease pathology, our findings suggest that aging, and a presymptomatic trajectory of Alzheimer's related decline, may fundamentally alter decision-making abilities in later life.


Aging , Decision Making , Locus Coeruleus , Magnetic Resonance Imaging , Locus Coeruleus/diagnostic imaging , Locus Coeruleus/physiology , Humans , Decision Making/physiology , Aged , Male , Female , Aging/physiology , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Middle Aged , Aged, 80 and over , Reward
11.
PLoS One ; 19(5): e0303278, 2024.
Article En | MEDLINE | ID: mdl-38771733

Currently, numerous studies focus on employing fMRI-based deep neural networks to diagnose neurological disorders such as Alzheimer's Disease (AD), yet only a handful have provided results regarding explainability. We address this gap by applying several prevalent explainability methods such as gradient-weighted class activation mapping (Grad-CAM) to an fMRI-based 3D-VGG16 network for AD diagnosis to improve the model's explainability. The aim is to explore the specific Region of Interest (ROI) of brain the model primarily focuses on when making predictions, as well as whether there are differences in these ROIs between AD and normal controls (NCs). First, we utilized multiple resting-state functional activity maps including ALFF, fALFF, ReHo, and VMHC to reduce the complexity of fMRI data, which differed from many studies that utilized raw fMRI data. Compared to methods utilizing raw fMRI data, this manual feature extraction approach may potentially alleviate the model's burden. Subsequently, 3D-VGG16 were employed for AD classification, where the final fully connected layers were replaced with a Global Average Pooling (GAP) layer, aimed at mitigating overfitting while preserving spatial information within the feature maps. The model achieved a maximum of 96.4% accuracy on the test set. Finally, several 3D CAM methods were employed to interpret the models. In the explainability results of the models with relatively high accuracy, the highlighted ROIs were primarily located in the precuneus and the hippocampus for AD subjects, while the models focused on the entire brain for NC. This supports current research on ROIs involved in AD. We believe that explaining deep learning models would not only provide support for existing research on brain disorders, but also offer important referential recommendations for the study of currently unknown etiologies.


Alzheimer Disease , Brain Mapping , Magnetic Resonance Imaging , Neural Networks, Computer , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/classification , Alzheimer Disease/physiopathology , Humans , Magnetic Resonance Imaging/methods , Female , Male , Brain Mapping/methods , Aged , Brain/diagnostic imaging , Brain/physiopathology
12.
Neurology ; 102(12): e209426, 2024 Jun 25.
Article En | MEDLINE | ID: mdl-38776513

BACKGROUND AND OBJECTIVES: With the aging US population and increasing incidence of Alzheimer disease (AD), understanding factors contributing to driving cessation among older adults is crucial for clinicians. Driving is integral for maintaining independence and functional mobility, but the risk factors for driving cessation, particularly in the context of normal aging and preclinical AD, are not well understood. We studied a well-characterized community cohort to examine factors associated with driving cessation. METHODS: This prospective, longitudinal observation study enrolled participants from the Knight Alzheimer Disease Research Center and The DRIVES Project. Participants were enrolled if they were aged 65 years or older, drove weekly, and were cognitively normal (Clinical Dementia Rating [CDR] = 0) at baseline. Participants underwent annual clinical, neurologic, and neuropsychological assessments, including ß-amyloid PET imaging and CSF (Aß42, total tau [t-Tau], and phosphorylated tau [p-Tau]) collection every 2-3 years. The primary outcome was time from baseline visit to driving cessation, accounting for death as a competing risk. The cumulative incidence function of driving cessation was estimated for each biomarker. The Fine and Gray subdistribution hazard model was used to examine the association between time to driving cessation and biomarkers adjusting for clinical and demographic covariates. RESULTS: Among the 283 participants included in this study, there was a mean follow-up of 5.62 years. Driving cessation (8%) was associated with older age, female sex, progression to symptomatic AD (CDR ≥0.5), and poorer performance on a preclinical Alzheimer cognitive composite (PACC) score. Aß PET imaging did not independently predict driving cessation, whereas CSF biomarkers, specifically t-Tau/Aß42 (hazard ratio [HR] 2.82, 95% CI 1.23-6.44, p = 0.014) and p-Tau/Aß42 (HR 2.91, 95% CI 1.28-6.59, p = 0.012) ratios, were independent predictors in the simple model adjusting for age, education, and sex. However, in the full model, progression to cognitive impairment based on the CDR and PACC score across each model was associated with a higher risk of driving cessation, whereas AD biomarkers were not statistically significant. DISCUSSION: Female sex, CDR progression, and neuropsychological measures of cognitive functioning obtained in the clinic were strongly associated with future driving cessation. The results emphasize the need for early planning and conversations about driving retirement in the context of cognitive decline and the immense value of clinical measures in determining functional outcomes.


Alzheimer Disease , Amyloid beta-Peptides , Automobile Driving , Biomarkers , tau Proteins , Humans , Female , Male , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/epidemiology , Alzheimer Disease/diagnosis , Aged , Biomarkers/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , tau Proteins/cerebrospinal fluid , Aged, 80 and over , Longitudinal Studies , Prospective Studies , Positron-Emission Tomography , Neuropsychological Tests , Cognition/physiology , Peptide Fragments/cerebrospinal fluid
13.
Alzheimers Res Ther ; 16(1): 115, 2024 May 23.
Article En | MEDLINE | ID: mdl-38778353

BACKGROUND: Maximizing the efficiency to screen amyloid-positive individuals in asymptomatic and non-demented aged population using blood-based biomarkers is essential for future success of clinical trials in the early stage of Alzheimer's disease (AD). In this study, we elucidate the utility of combination of plasma amyloid-ß (Aß)-related biomarkers and tau phosphorylated at threonine 217 (p-tau217) to predict abnormal Aß-positron emission tomography (PET) in the preclinical and prodromal AD. METHODS: We designed the cross-sectional study including two ethnically distinct cohorts, the Japanese trial-ready cohort for preclinica and prodromal AD (J-TRC) and the Swedish BioFINDER study. J-TRC included 474 non-demented individuals (CDR 0: 331, CDR 0.5: 143). Participants underwent plasma Aß and p-tau217 assessments, and Aß-PET imaging. Findings in J-TRC were replicated in the BioFINDER cohort including 177 participants (cognitively unimpaired: 114, mild cognitive impairment: 63). In both cohorts, plasma Aß(1-42) (Aß42) and Aß(1-40) (Aß40) were measured using immunoprecipitation-MALDI TOF mass spectrometry (Shimadzu), and p-tau217 was measured with an immunoassay on the Meso Scale Discovery platform (Eli Lilly). RESULTS: Aß-PET was abnormal in 81 participants from J-TRC and 71 participants from BioFINDER. Plasma Aß42/Aß40 ratio and p-tau217 individually showed moderate to high accuracies when detecting abnormal Aß-PET scans, which were improved by combining plasma biomarkers and by including age, sex and APOE genotype in the models. In J-TRC, the highest AUCs were observed for the models combining p-tau217/Aß42 ratio, APOE, age, sex in the whole cohort (AUC = 0.936), combining p-tau217, Aß42/Aß40 ratio, APOE, age, sex in the CDR 0 group (AUC = 0.948), and combining p-tau217/Aß42 ratio, APOE, age, sex in the CDR 0.5 group (AUC = 0.955), respectively. Each subgroup results were replicated in BioFINDER, where the highest AUCs were seen for models combining p-tau217, Aß42/40 ratio, APOE, age, sex in cognitively unimpaired (AUC = 0.938), and p-tau217/Aß42 ratio, APOE, age, sex in mild cognitive impairment (AUC = 0.914). CONCLUSIONS: Combination of plasma Aß-related biomarkers and p-tau217 exhibits high performance when predicting Aß-PET positivity. Adding basic clinical information (i.e., age, sex, APOE Îµ genotype) improved the prediction in preclinical AD, but not in prodromal AD. Combination of Aß-related biomarkers and p-tau217 could be highly useful for pre-screening of participants in clinical trials of preclinical and prodromal AD.


Amyloid beta-Peptides , Biomarkers , Brain , Positron-Emission Tomography , tau Proteins , Humans , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/metabolism , Female , Male , tau Proteins/blood , Aged , Positron-Emission Tomography/methods , Biomarkers/blood , Cross-Sectional Studies , Brain/diagnostic imaging , Brain/metabolism , Aged, 80 and over , Cohort Studies , Phosphorylation , Middle Aged , Alzheimer Disease/blood , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/diagnosis , Peptide Fragments/blood , Cognitive Dysfunction/blood , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/diagnosis
14.
PLoS One ; 19(5): e0303288, 2024.
Article En | MEDLINE | ID: mdl-38781243

BACKGROUND: Brain region segmentation and morphometry in humanized apolipoprotein E (APOE) mouse models with a human NOS2 background (HN) contribute to Alzheimer's disease (AD) research by demonstrating how various risk factors affect the brain. Photon-counting detector (PCD) micro-CT provides faster scan times than MRI, with superior contrast and spatial resolution to energy-integrating detector (EID) micro-CT. This paper presents a pipeline for mouse brain imaging, segmentation, and morphometry from PCD micro-CT. METHODS: We used brains of 26 mice from 3 genotypes (APOE22HN, APOE33HN, APOE44HN). The pipeline included PCD and EID micro-CT scanning, hybrid (PCD and EID) iterative reconstruction, and brain region segmentation using the Small Animal Multivariate Brain Analysis (SAMBA) tool. We applied SAMBA to transfer brain region labels from our new PCD CT atlas to individual PCD brains via diffeomorphic registration. Region-based and voxel-based analyses were used for comparisons by genotype and sex. RESULTS: Together, PCD and EID scanning take ~5 hours to produce images with a voxel size of 22 µm, which is faster than MRI protocols for mouse brain morphometry with voxel size above 40 µm. Hybrid iterative reconstruction generates PCD images with minimal artifacts and higher spatial resolution and contrast than EID images. Our PCD atlas is qualitatively and quantitatively similar to the prior MRI atlas and successfully transfers labels to PCD brains in SAMBA. Male and female mice had significant volume differences in 26 regions, including parts of the entorhinal cortex and cingulate cortex. APOE22HN brains were larger than APOE44HN brains in clusters from the hippocampus, a region where atrophy is associated with AD. CONCLUSIONS: This work establishes a pipeline for mouse brain analysis using PCD CT, from staining to imaging and labeling brain images. Our results validate the effectiveness of the approach, setting a foundation for research on AD mouse models while reducing scanning durations.


Brain , X-Ray Microtomography , Animals , Brain/diagnostic imaging , Mice , X-Ray Microtomography/methods , Female , Male , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Image Processing, Computer-Assisted/methods , Apolipoproteins E/genetics , Mice, Transgenic
15.
Alzheimers Res Ther ; 16(1): 100, 2024 May 06.
Article En | MEDLINE | ID: mdl-38711107

BACKGROUND: Retinal microvascular signs are accessible measures of early alterations in microvascular dysregulation and have been associated with dementia; it is unclear if they are associated with AD (Alzheimer's disease) pathogenesis as a potential mechanistic link. This study aimed to test the association of retinal microvascular abnormalities in mid and late life and late life cerebral amyloid. METHODS: Participants from the ARIC-PET (Atherosclerosis Risk in Communities-Positron Emission Tomography) study with a valid retinal measure (N = 285) were included. The associations of mid- and late-life retinal signs with late-life amyloid-ß (Aß) by florbetapir PET were tested. Two different measures of Aß burden were included: (1) elevated amyloid (SUVR > 1.2) and (2) continuous amyloid SUVR. The retinal measures' association with Aß burden was assessed using logistic and robust linear regression models. A newly created retinal score, incorporating multiple markers of retinal abnormalities, was also evaluated in association with greater Aß burden. RESULTS: Retinopathy in midlife (OR (95% CI) = 0.36 (0.08, 1.40)) was not significantly associated with elevated amyloid burden. In late life, retinopathy was associated with increased continuous amyloid standardized value uptake ratio (SUVR) (ß (95%CI) = 0.16 (0.02, 0.32)) but not elevated amyloid burden (OR (95%CI) = 2.37 (0.66, 9.88)) when accounting for demographic, genetic and clinical risk factors. A high retinal score in late life, indicating a higher burden of retinal abnormalities, was also significantly associated with increased continuous amyloid SUVR (ß (95% CI) = 0.16 (0.04, 0.32)) independent of vascular risk factors. CONCLUSIONS: Retinopathy in late life may be an easily obtainable marker to help evaluate the mechanistic vascular pathway between retinal measures and dementia, perhaps acting via AD pathogenesis. Well-powered future studies with a greater number of retinal features and other microvascular signs are needed to test these findings.


Amyloid beta-Peptides , Aniline Compounds , Brain , Positron-Emission Tomography , Retinal Vessels , Humans , Female , Male , Amyloid beta-Peptides/metabolism , Positron-Emission Tomography/methods , Aged , Middle Aged , Brain/diagnostic imaging , Brain/metabolism , Retinal Vessels/diagnostic imaging , Retinal Diseases/diagnostic imaging , Retinal Diseases/metabolism , Microvessels/diagnostic imaging , Microvessels/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Ethylene Glycols
16.
Front Neural Circuits ; 18: 1345692, 2024.
Article En | MEDLINE | ID: mdl-38694272

Novel brain clearing methods revolutionize imaging by increasing visualization throughout the brain at high resolution. However, combining the standard tool of immunostaining targets of interest with clearing methods has lagged behind. We integrate whole-mount immunostaining with PEGASOS tissue clearing, referred to as iPEGASOS (immunostaining-compatible PEGASOS), to address the challenge of signal quenching during clearing processes. iPEGASOS effectively enhances molecular-genetically targeted fluorescent signals that are otherwise compromised during conventional clearing procedures. Additionally, we demonstrate the utility of iPEGASOS for visualizing neurochemical markers or viral labels to augment visualization that transgenic mouse lines cannot provide. Our study encompasses three distinct applications, each showcasing the versatility and efficacy of this approach. We employ whole-mount immunostaining to enhance molecular signals in transgenic reporter mouse lines to visualize the whole-brain spatial distribution of specific cellular populations. We also significantly improve the visualization of neural circuit connections by enhancing signals from viral tracers injected into the brain. Last, we show immunostaining without genetic markers to selectively label beta-amyloid deposits in a mouse model of Alzheimer's disease, facilitating the comprehensive whole-brain study of pathological features.


Alzheimer Disease , Brain , Mice, Transgenic , Animals , Brain/metabolism , Brain/diagnostic imaging , Mice , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Immunohistochemistry , Neuroimaging/methods , Amyloid beta-Peptides/metabolism , Mice, Inbred C57BL
17.
Zh Nevrol Psikhiatr Im S S Korsakova ; 124(4. Vyp. 2): 33-40, 2024.
Article Ru | MEDLINE | ID: mdl-38696149

OBJECTIVE: To study the severity and localization of dilated perivascular spaces (DPVS), the levels of protein markers of amyloidosis and neurodegeneration in the cerebrospinal fluid (CSF) at different daily blood pressure (BP) profiles in patients with Alzheimer's disease (AD) and other types of cognitive impairment. MATERIAL AND METHODS: A total of 119 people, aged 53 to 92 years, including 55 patients with AD, 27 patients with vascular cognitive disorders (VCD), 19 patients with frontotemporal degeneration (FTD). All patients underwent BP monitoring for 24 hours using a standard oscillometric measurement method, lumbar puncture to assess Aß-42 and Aß-40 amyloid protein, total and phosphorylated tau protein in the CSF, magnetic resonance imaging tomography of the brain with subsequent assessment of the severity of expansion and localization of DPVS according to the G.M. Potter scale. RESULTS: In 58.3% of patients with AD, there is no adequate reduction in BP at night in comparison with patients with VCD (p<0.05). A significant degree of expansion of the DPVS turned out to be most typical for patients with AD: grade 3 was detected in 45.7% of patients, and the maximum, grade 4, was detected in 13.4%. At the same time, DPVSs were significantly more often detected in the group of subjects with insufficient reduction in diastolic BP (DBP) at night. A strong inverse correlation was established between the level of Aß-42 in the CSF and the variability of DBP at night (r= -0.92; p<0.05). The decrease in the level of Aß-42 in AD, especially at the prodromal stage, is directly related to the low variability of DBP at night, which is more characteristic of an insufficient decrease or increase in BP during night sleep. CONCLUSION: Patients with AD were characterized by an insufficient decrease in BP at night, which is associated with the severity and degree of maximum expansion of the DPVS. A decrease in the level of Aß-42 amyloid protein in the CSF strongly correlates with the variability of DBP at night.


Alzheimer Disease , Amyloid beta-Peptides , Hypertension , tau Proteins , Humans , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnostic imaging , Aged , Female , Male , Middle Aged , Amyloid beta-Peptides/cerebrospinal fluid , Hypertension/complications , Hypertension/cerebrospinal fluid , Aged, 80 and over , tau Proteins/cerebrospinal fluid , Magnetic Resonance Imaging , Glymphatic System/diagnostic imaging , Blood Pressure/physiology , Peptide Fragments/cerebrospinal fluid , Dementia, Vascular/cerebrospinal fluid , Dementia, Vascular/diagnostic imaging , Biomarkers/cerebrospinal fluid , Brain/diagnostic imaging , Brain/pathology
18.
Zh Nevrol Psikhiatr Im S S Korsakova ; 124(4. Vyp. 2): 17-24, 2024.
Article Ru | MEDLINE | ID: mdl-38696147

OBJECTIVE: To investigate the pattern and connections of neuropsychological and metabolic indices in patients with cognitive disorders of Alzheimer's and vascular (subcortical-cortical) types of different severity. MATERIAL AND METHODS: A total of 177 patients were examined, including 85 patients with Alzheimer's disease (AD) and 92 patients with vascular cognitive impairment (VCI). All patients underwent complex neuropsychological examination; 18F-FDG PET was performed in 17 patients with AD and 15 patients with VCI. RESULTS: The greatest changes in patients with AD were noted in the mnestic sphere, and the indicators significantly differed from the results of the study of patients with VCI already at the pre-dementia stage. Neurodynamic and dysregulatory disorders prevailed in patients with VCI. Patients with AD showed bilateral symmetrical reduction of metabolic activity in the cortex of parietal and temporal lobes, often in combination with marked hypometabolism in the hippocampal region. In patients with VCI, there were areas of decreased brain tissue metabolism of different localization and size, mainly in the projection of the basal ganglia and in the prefrontal and parietal cortex, as well as in the cingulate gyrus, which indirectly confirms the mechanism of disconnection of subcortical and cortical structures. In AD, impaired metabolic activity in the hippocampal region correlated with impaired temporal and spatial orientation (ρ=-0.54, p<0.05), memory impairment (ρ=-0.71, p<0.005). Hypometabolism of the parietal lobe cortex was associated with total MMSE score (ρ=-0.8, p<0.001), 10-word test (ρ=-0.89, p<0.001 and ρ=-0.82, p<0.001), visual-spatial impairment (ρ=-0.64, p<0.01), categorical association test (ρ=-0.73, p<0.005). In patients with VCI, dysregulatory disorders correlated with hypometabolism in the thalamic projection (ρ=-0.56, p<0.05), prefrontal cortex (ρ=-0.64, p<0.05) and in the cingulate gyrus (anterior regions) (ρ=-0.53, p<0.05). CONCLUSION: The results indicate the presence of differences in cognitive impairment and cerebral metabolism in patients with AD and VCI.


Alzheimer Disease , Cognitive Dysfunction , Fluorodeoxyglucose F18 , Neuropsychological Tests , Positron-Emission Tomography , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Male , Female , Aged , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/etiology , Cognitive Dysfunction/diagnostic imaging , Dementia, Vascular/diagnostic imaging , Dementia, Vascular/metabolism , Dementia, Vascular/physiopathology , Middle Aged , Brain/metabolism , Brain/diagnostic imaging , Aged, 80 and over
19.
Zh Nevrol Psikhiatr Im S S Korsakova ; 124(4. Vyp. 2): 56-63, 2024.
Article Ru | MEDLINE | ID: mdl-38696152

The most common cause of severe cognitive impairment in adults is Alzheimer's disease (AD). Depending on the age of onset, AD is divided into early (<65 years) and late (≥65 years) forms. Early-onset AD (EOAD) is significantly less common than later-onset AD (LOAD) and accounts for only about 5-10% of cases. However, its medical and social significance, as a disease leading to loss of ability to work and legal capacity, as well as premature death in patients aged 40-64 years, is extremely high. Patients with EOAD compared with LOAD have a greater number of atypical clinical variants - 25% and 6-12.5%, respectively, which complicates the differential diagnosis of EOAD with other neurodegenerative diseases. However, the typical classical amnestic variant predominates in both EOAD and LOAD. Also, patients with EOAD have peculiarities according to neuroimaging data: when performing MRI of the brain, patients with EOAD often have more pronounced parietal atrophy and less pronounced hippocampal atrophy compared to patients with LOAD. The article pays attention to the features of the clinical and neuroimaging data in patients with EOAD; a case of a patient with EOAD is presented.


Age of Onset , Alzheimer Disease , Magnetic Resonance Imaging , Neuroimaging , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/diagnosis , Neuroimaging/methods , Middle Aged , Atrophy/diagnostic imaging , Diagnosis, Differential , Male , Brain/diagnostic imaging , Brain/pathology , Female , Hippocampus/diagnostic imaging , Hippocampus/pathology
20.
Hum Brain Mapp ; 45(7): e26709, 2024 May.
Article En | MEDLINE | ID: mdl-38746977

The high prevalence of conversion from amnestic mild cognitive impairment (aMCI) to Alzheimer's disease (AD) makes early prevention of AD extremely critical. Neuroticism, a heritable personality trait associated with mental health, has been considered a risk factor for conversion from aMCI to AD. However, whether the neuroticism genetic risk could predict the conversion of aMCI and its underlying neural mechanisms is unclear. Neuroticism polygenic risk score (N-PRS) was calculated in 278 aMCI patients with qualified genomic and neuroimaging data from ADNI. After 1-year follow-up, N-PRS in patients of aMCI-converted group was significantly greater than those in aMCI-stable group. Logistic and Cox survival regression revealed that N-PRS could significantly predict the early-stage conversion risk from aMCI to AD. These results were well replicated in an internal dataset and an independent external dataset of 933 aMCI patients from the UK Biobank. One sample Mendelian randomization analyses confirmed a potentially causal association from higher N-PRS to lower inferior parietal surface area to higher conversion risk of aMCI patients. These analyses indicated that neuroticism genetic risk may increase the conversion risk from aMCI to AD by impairing the inferior parietal structure.


Alzheimer Disease , Cognitive Dysfunction , Disease Progression , Magnetic Resonance Imaging , Multifactorial Inheritance , Neuroticism , Parietal Lobe , Humans , Alzheimer Disease/genetics , Alzheimer Disease/diagnostic imaging , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/genetics , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Male , Female , Aged , Parietal Lobe/diagnostic imaging , Parietal Lobe/pathology , Aged, 80 and over , Mendelian Randomization Analysis , Middle Aged , Genetic Predisposition to Disease
...