Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 162
1.
Alzheimers Dement (Amst) ; 16(2): e12578, 2024.
Article En | MEDLINE | ID: mdl-38800122

Abstract: The utility of brain magnetic resonance imaging (MRI) for predicting dementia is debated. We evaluated the added value of repeated brain MRI, including atrophy and cerebral small vessel disease markers, for dementia prediction. We conducted a landmark competing risk analysis in 1716 participants of the French population-based Three-City Study to predict the 5-year risk of dementia using repeated measures of 41 predictors till year 4 of follow-up. Brain MRI markers improved significantly the individual prediction of dementia after accounting for demographics, health measures, and repeated measures of cognition and functional dependency (area under the ROC curve [95% CI] improved from 0.80 [0.79 to 0.82] to 0.83 [0.81 to 0.84]). Nonetheless, accounting for the change over time through repeated MRIs had little impact on predictive abilities. These results highlight the importance of multimodal analysis to evaluate the added predictive abilities of repeated brain MRI for dementia and offer new insights into the predictive performances of various MRI markers. Highlights: We evaluated whether repeated brain volumes and cSVD markers improve dementia prediction.The 5-year prediction of dementia is slightly improved when considering brain MRI markers.Measures of hippocampus volume are the main MRI predictors of dementia.Adjusted on cognition, repeated MRI has poor added value over single MRI for dementia prediction.We utilized a longitudinal analysis that considers error-and-missing-prone predictors, and competing death.

2.
JAMA Netw Open ; 7(5): e2412824, 2024 May 01.
Article En | MEDLINE | ID: mdl-38776079

Importance: Vascular disease is a treatable contributor to dementia risk, but the role of specific markers remains unclear, making prevention strategies uncertain. Objective: To investigate the causal association between white matter hyperintensity (WMH) burden, clinical stroke, blood pressure (BP), and dementia risk, while accounting for potential epidemiologic biases. Design, Setting, and Participants: This study first examined the association of genetically determined WMH burden, stroke, and BP levels with Alzheimer disease (AD) in a 2-sample mendelian randomization (2SMR) framework. Second, using population-based studies (1979-2018) with prospective dementia surveillance, the genetic association of WMH, stroke, and BP with incident all-cause dementia was examined. Data analysis was performed from July 26, 2020, through July 24, 2022. Exposures: Genetically determined WMH burden and BP levels, as well as genetic liability to stroke derived from genome-wide association studies (GWASs) in European ancestry populations. Main Outcomes and Measures: The association of genetic instruments for WMH, stroke, and BP with dementia was studied using GWASs of AD (defined clinically and additionally meta-analyzed including both clinically diagnosed AD and AD defined based on parental history [AD-meta]) for 2SMR and incident all-cause dementia for longitudinal analyses. Results: In 2SMR (summary statistics-based) analyses using AD GWASs with up to 75 024 AD cases (mean [SD] age at AD onset, 75.5 [4.4] years; 56.9% women), larger WMH burden showed evidence for a causal association with increased risk of AD (odds ratio [OR], 1.43; 95% CI, 1.10-1.86; P = .007, per unit increase in WMH risk alleles) and AD-meta (OR, 1.19; 95% CI, 1.06-1.34; P = .008), after accounting for pulse pressure for the former. Blood pressure traits showed evidence for a protective association with AD, with evidence for confounding by shared genetic instruments. In the longitudinal (individual-level data) analyses involving 10 699 incident all-cause dementia cases (mean [SD] age at dementia diagnosis, 74.4 [9.1] years; 55.4% women), no significant association was observed between larger WMH burden and incident all-cause dementia (hazard ratio [HR], 1.02; 95% CI, 1.00-1.04; P = .07). Although all exposures were associated with mortality, with the strongest association observed for systolic BP (HR, 1.04; 95% CI, 1.03-1.06; P = 1.9 × 10-14), there was no evidence for selective survival bias during follow-up using illness-death models. In secondary analyses using polygenic scores, the association of genetic liability to stroke, but not genetically determined WMH, with dementia outcomes was attenuated after adjusting for interim stroke. Conclusions: These findings suggest that WMH is a primary vascular factor associated with dementia risk, emphasizing its significance in preventive strategies for dementia. Future studies are warranted to examine whether this finding can be generalized to non-European populations.


Blood Pressure , Cerebral Small Vessel Diseases , Dementia , Humans , Cerebral Small Vessel Diseases/genetics , Cerebral Small Vessel Diseases/epidemiology , Female , Male , Aged , Dementia/genetics , Dementia/epidemiology , Blood Pressure/genetics , Genome-Wide Association Study , Mendelian Randomization Analysis , Alzheimer Disease/genetics , Alzheimer Disease/epidemiology , Stroke/genetics , Stroke/epidemiology , Risk Factors , Genetic Predisposition to Disease , Aged, 80 and over , Prospective Studies
3.
Neurology ; 102(9): e209307, 2024 May.
Article En | MEDLINE | ID: mdl-38626384

BACKGROUND AND OBJECTIVES: Elevated levels of Alzheimer disease (AD) blood-based biomarkers are associated with accelerated cognitive decline. However, their distinct relationships with specific cognitive and functional domains require further investigation. We aimed at estimating the associations between AD blood-based biomarkers and the trajectories of distinct cognitive and functional domains over a 5-year follow-up period. METHODS: We conducted a clinic-based prospective study using data from the MEMENTO study, a nationwide French cohort. We selected dementia-free individuals at baseline aged 60 years or older. Baseline measurements of ß-amyloid (Aß) 40 and 42, phosphorylated tau (p-tau181), and neurofilament light chain (NfL) concentrations were obtained using the Simoa HD-X analyzer. Mini-Mental State Examination (MMSE), Free and Cued Selective Reminding Test (FCSRT), animal fluency, Trail Making Tests A and B, Short Physical Performance Battery (SPPB), and Instrumental Activities of Daily Living were administered annually for up to 5 years. We used linear mixed models, adjusted for potential confounders, to model AD biomarkers' relation with cognitive and functional decline. RESULTS: A total of 1,938 participants were included in this study, with a mean (SD) baseline age of 72.8 (6.6) years, and 62% were women. Higher baseline p-tau181 and NfL were associated with significantly faster decline in most cognitive, physical, and functional outcomes (+1 SD p-tau181: ßMMSE = -0.055, 95% CI -0.067 to -0.043, ßFCSRT = -0.034, 95% CI -0.043 to -0.025, ßfluency = -0.029, 95% CI -0.038 to -0.020, ßSPPB = -0.040, 95% CI -0.057 to -0.022, and ß4IADL = -0.115, 95% CI 0.091-0.140. +1 SD NfL: ßMMSE = -0.039, 95% CI -0.053 to -0.025, ßFCSRT = -0.022, 95% CI -0.032 to -0.012, ßfluency = -0.014, 95% CI -0.024 to -0.004, and ß4IADL = 0.077, 95% CI 0.048-0.105). A multiplicative association of p-tau181 and NfL with worsening cognitive and functional trajectories was evidenced. Lower Aß42/40 ratio was only associated with slightly faster cognitive decline in FCSRT and semantic fluency (+1 SD: ß = 0.011, 95% CI 0.002-0.020, and ß = 0.011, 95% CI 0.003-0.020, respectively). These associations were not modified by APOE ε4, sex, nor education level. DISCUSSION: In a memory clinic sample, p-tau181 and NfL, both independently and jointly, are linked to more pronounced cognitive, physical and functional declines. Blood-based biomarker measurement in AD research may provide useful insights regarding biological processes underlying cognitive, physical, and functional declines in at-risk individuals.


Alzheimer Disease , Cognitive Dysfunction , Humans , Female , Male , tau Proteins , Prospective Studies , Activities of Daily Living , Alzheimer Disease/diagnosis , Alzheimer Disease/psychology , Amyloid beta-Peptides , Cognitive Dysfunction/diagnosis , Biomarkers , Cognition
4.
Neurology ; 102(8): e209219, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38527237

BACKGROUND AND OBJECTIVES: Patients' comorbidities can affect Alzheimer disease (AD) blood biomarker concentrations. Because a limited number of factors have been explored to date, our aim was to assess the proportion of the variance in fluid biomarker levels explained by the clinical features of AD and by a large number of non-AD-related factors. METHODS: MEMENTO enrolled 2,323 individuals with cognitive complaints or mild cognitive impairment in 26 French memory clinics. Baseline evaluation included clinical and neuropsychological assessments, brain MRI, amyloid-PET, CSF (optional), and blood sampling. Blood biomarker levels were determined using the Simoa-HDX analyzer. We performed linear regression analysis of the clinical features of AD (cognition, AD genetic risk score, and brain atrophy) to model biomarker concentrations. Next, we added covariates among routine biological tests, inflammatory markers, demographic and behavioral determinants, treatments, comorbidities, and preanalytical sample handling in final models using both stepwise selection processes and least absolute shrinkage and selection operator (LASSO). RESULTS: In total, 2,257 participants were included in the analysis (median age 71.7, 61.8% women, 55.2% with high educational levels). For blood biomarkers, the proportion of variance explained by clinical features of AD was 13.7% for neurofilaments (NfL), 11.4% for p181-tau, 3.0% for Aß-42/40, and 1.4% for total-tau. In final models accounting for non-AD-related factors, the variance was mainly explained by age, routine biological tests, inflammatory markers, and preanalytical sample handling. In CSF, the proportion of variance explained by clinical features of AD was 24.8% for NfL, 22.3% for Aß-42/40, 19.8% for total-tau, and 17.2% for p181-tau. In contrast to blood biomarkers, the largest proportion of variance was explained by cognition after adjustment for covariates. The covariates that explained the largest proportion of variance were also the most frequently selected with LASSO. The performance of blood biomarkers for predicting A+ and T+ status (PET or CSF) remained unchanged after controlling for drivers of variance. DISCUSSION: This comprehensive analysis demonstrated that the variance in AD blood biomarker concentrations was mainly explained by age, with minor contributions from cognition, brain atrophy, and genetics, conversely to CSF measures. These results challenge the use of blood biomarkers as isolated stand-alone biomarkers for AD.


Alzheimer Disease , Cognitive Dysfunction , Humans , Female , Male , Alzheimer Disease/genetics , tau Proteins , Amyloid beta-Peptides , Biomarkers , Cognitive Dysfunction/psychology , Atrophy , Peptide Fragments
5.
Neurology ; 102(6): e208054, 2024 Mar 26.
Article En | MEDLINE | ID: mdl-38412412

BACKGROUND AND OBJECTIVES: Global amyloid-PET is associated with cognition and cognitive decline, but most research on this association does not account for past cognitive information. We assessed the prognostic benefit of amyloid-PET measures for future cognition when prior cognitive assessments are available, evaluating the added value of amyloid measures beyond information on multiple past cognitive assessments. METHODS: The French MEMENTO cohort (a cohort of outpatients from French research memory centers to improve knowledge on Alzheimer disease and related disorders) includes older outpatients with incipient cognitive changes, but no dementia diagnosis at inclusion. Global amyloid burden was assessed using positron emission tomography (amyloid-PET) for a subset of participants; semiannual cognitive testing was subsequently performed. We predicted mini-mental state examination (MMSE) scores using demographic characteristics (age, sex, marital status, and education) alone or in combination with information on prior cognitive measures. The added value of amyloid burden as a predictor in these models was evaluated with percent reduction of the mean squared error (MSE). All models were conducted separately for evaluating the added value of dichotomous amyloid positivity status compared with a continuous amyloid-standardized uptake-value ratio. RESULTS: Our analytic sample comprised 510 individuals who underwent amyloid-PET scans with at least 4 MMSE assessments. The mean age at the PET scan was 71.6 (standard deviation 7.4) years; 60.7% were female. The median follow-up was 4.6 years (interquartile range: 0.9 years). Adding amyloid burden when adjusting for only demographic characteristics reduced the MSE of predictions by 5.08% (95% CI 0.97%-10.86%) and 12.64% (95% CI 3.35%-25.28%) for binary and continuous amyloid, respectively. If the model included 1 past MMSE measure, the MSE improvement was 3.51% (95% CI 1.01%-7.28%) when adding binary amyloid and 8.83% (95% CI 2.63%-16.37%) when adding continuous amyloid. Improvements in model fit were smaller with the addition of amyloid burden when more than 1 past cognitive assessment was included. For all models incorporating past cognitive assessments, differences in predictions amounted to a fraction of 1 MMSE point on average. DISCUSSION: In a clinical setting, global amyloid burden did not appreciably improve cognitive predictions when past cognitive assessments were available. TRIAL REGISTRATION INFORMATION: ClinicalTrials.gov Identifier: NCT02164643.


Alzheimer Disease , Cognitive Dysfunction , Humans , Female , Male , Amyloid beta-Peptides , Cognition , Amyloid , Cognitive Dysfunction/psychology , Positron-Emission Tomography , Alzheimer Disease/complications , Amyloidogenic Proteins
6.
Eur J Epidemiol ; 39(4): 409-417, 2024 Apr.
Article En | MEDLINE | ID: mdl-38190014

To explore to which extent neurodegeneration and cerebral small vessel disease (SVD) could mediate the association between type-2 diabetes and higher dementia risk. The analytical sample consisted in 2228 participants, out of the Three-City study, aged 65 and older, free of dementia at baseline who underwent brain MRI. Diabetes was defined by medication intake or fasting or non-fasting elevated glucose levels. Dementia status was assessed every 2 to 3 years, during up to 12 years of follow-up. Brain parenchymal fraction (BPF) and white matter hyperintensities volume (WMHV) were selected as markers of neurodegeneration and cerebral SVD respectively. We performed a mediation analysis of the effect of baseline BPF and WMHV (mediators) on the association between diabetes and dementia risk using linear and Cox models adjusted for age, sex, education level, hypertension, hypercholesterolemia, BMI, smoking and alcohol drinking status, APOE-ε4 status, and study site. At baseline, 8.8% of the participants had diabetes. Diabetes (yes vs. no) was associated with higher WMHV (ßdiab = 0.193, 95% CI 0.040; 0.346) and lower BPF (ßdiab = -0.342, 95% CI -0.474; -0.210), as well as with an increased risk of dementia over 12 years of follow-up (HRdiab = 1.65, 95% CI 1.04; 2.60). The association between diabetes status and dementia risk was statistically mediated by higher WMHV (HRdiab=1.05, 95% CI 1.01; 1.11, mediated part = 10.8%) and lower BPF (HRdiab = 1.12, 95% CI 1.05; 1.20, mediated part = 22.9%). This study showed that both neurodegeneration and cerebral SVD statistically explained almost 30% of the association between diabetes and dementia.


Cerebral Small Vessel Diseases , Dementia , Diabetes Mellitus, Type 2 , Magnetic Resonance Imaging , Mediation Analysis , Humans , Female , Male , Aged , Dementia/etiology , Dementia/epidemiology , Cerebral Small Vessel Diseases/diagnostic imaging , Cerebral Small Vessel Diseases/pathology , Diabetes Mellitus, Type 2/complications , Risk Factors , Brain/diagnostic imaging , Brain/pathology , Biomarkers/blood , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/epidemiology , Aged, 80 and over
7.
Alzheimers Dement ; 20(2): 809-818, 2024 Feb.
Article En | MEDLINE | ID: mdl-37779086

INTRODUCTION: Inferring the timeline from mild cognitive impairment (MCI) to severe dementia is pivotal for patients, clinicians, and researchers. Literature is sparse and often contains few patients. We aim to determine the time spent in MCI, mild-, moderate-, severe dementia, and institutionalization until death. METHODS: Multistate modeling with Cox regression was used to obtain the sojourn time. Covariates were age at baseline, sex, amyloid status, and Alzheimer's disease (AD) or other dementia diagnosis. The sample included a register (SveDem) and memory clinics (Amsterdam Dementia Cohort and Memento). RESULTS: Using 80,543 patients, the sojourn time from clinically identified MCI to death across all patient groups ranged from 6.20 (95% confidence interval [CI]: 5.57-6.98) to 10.08 (8.94-12.18) years. DISCUSSION: Generally, sojourn time was inversely associated with older age at baseline, males, and AD diagnosis. The results provide key estimates for researchers and clinicians to estimate prognosis.


Alzheimer Disease , Cognitive Dysfunction , Dementia , Male , Humans , Disease Progression , Alzheimer Disease/complications , Dementia/diagnosis , Dementia/complications , Cognitive Dysfunction/psychology , Institutionalization
8.
Alzheimers Res Ther ; 15(1): 205, 2023 11 22.
Article En | MEDLINE | ID: mdl-37993894

BACKGROUND: Cognitive complaints are often regarded as an early sign of Alzheimer's disease (AD) but may also occur in several other conditions and contexts. This study examines the correlates of cognitive complaint trajectories over a 5-year period in individuals who shared similar objective cognitive trajectories. METHODS: We analyzed a subsample (n = 1748) of the MEMENTO cohort, consisting of individuals with subjective cognitive decline or mild cognitive impairment at baseline. Participants were stratified based on their latent MMSE trajectory over a 5-year period: "high and increasing," "subtle decline," and "steep decline." Within each of the three strata, we used a latent-class longitudinal approach to identify distinct trajectories of cognitive complaints. We then used multiple logistic regressions to examine the association between these complaint trajectories and several factors, including AD biomarkers (blood pTau/Aß42 ratio, cortical thickness, APOE genotype), anxiety, depression, social relationships, a comorbidity-polypharmacy score, and demographic characteristics. RESULTS: Among participants with high and increasing MMSE scores, greater baseline comorbidity-polypharmacy scores (odds ratio (OR) = 1.30, adjusted p = 0.03) were associated with higher odds of moderate and increasing cognitive complaints (as opposed to mild and decreasing complaints). Baseline depression and social relationships also showed significant associations with the complaint pattern but did not survive correction for multiple comparisons. Among participants with subtle decline in MMSE scores, greater baseline depression (OR = 1.76, adjusted p = 0.02) was associated with higher odds of moderate and increasing cognitive complaints (versus mild and decreasing). Similarly, baseline comorbidity-polypharmacy scores and pTau/Aß42 ratio exhibited significant associations, but they did not survive correction. Among participants with a steep decline in MMSE scores, greater baseline comorbidity-polypharmacy scores increased the odds of moderate complaints (versus mild, OR = 1.38, unadjusted p = 0.03, adjusted p = 0.32), but this effect did not survive correction for multiple comparisons. CONCLUSIONS: Despite similar objective cognitive trajectory, there is heterogeneity in the subjective perception of these cognitive changes. This perception was explained by both AD-related and, more robustly, non-AD-related factors. These findings deepen our understanding of the multifaceted nature of subjective cognitive complaints in individuals at risk for dementia and underscore the importance of considering a range of factors when interpreting cognitive complaints.


Alzheimer Disease , Cognitive Dysfunction , Humans , Alzheimer Disease/diagnosis , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/epidemiology , Cognition
9.
medRxiv ; 2023 Aug 13.
Article En | MEDLINE | ID: mdl-37790435

Importance: There is increasing recognition that vascular disease, which can be treated, is a key contributor to dementia risk. However, the contribution of specific markers of vascular disease is unclear and, as a consequence, optimal prevention strategies remain unclear. Objective: To disentangle the causal relation of several key vascular traits to dementia risk: (i) white matter hyperintensity (WMH) burden, a highly prevalent imaging marker of covert cerebral small vessel disease (cSVD); (ii) clinical stroke; and (iii) blood pressure (BP), the leading risk factor for cSVD and stroke, for which efficient therapies exist. To account for potential epidemiological biases inherent to late-onset conditions like dementia. Design Setting and Participants: This study first explored the association of genetically determined WMH, BP levels and stroke risk with AD using summary-level data from large genome-wide association studies (GWASs) in a two-sample Mendelian randomization (MR) framework. Second, leveraging individual-level data from large longitudinal population-based cohorts and biobanks with prospective dementia surveillance, the association of weighted genetic risk scores (wGRSs) for WMH, BP, and stroke with incident all-cause-dementia was explored using Cox-proportional hazard and multi-state models. The data analysis was performed from July 26, 2020, through July 24, 2022. Exposures: Genetically determined levels of WMH volume and BP (systolic, diastolic and pulse blood pressures) and genetic liability to stroke. Main outcomes and measures: The summary-level MR analyses focused on the outcomes from GWAS of clinically diagnosed AD (n-cases=21,982) and GWAS additionally including self-reported parental history of dementia as a proxy for AD diagnosis (ADmeta, n-cases=53,042). For the longitudinal analyses, individual-level data of 157,698 participants with 10,699 incident all-cause-dementia were studied, exploring AD, vascular or mixed dementia in secondary analyses. Results: In the two-sample MR analyses, WMH showed strong evidence for a causal association with increased risk of ADmeta (OR, 1.16; 95%CI:1.05-1.28; P=.003) and AD (OR, 1.28; 95%CI:1.07-1.53; P=.008), after accounting for genetically determined pulse pressure for the latter. Genetically predicted BP traits showed evidence for a protective association with both clinically defined AD and ADmeta, with evidence for confounding by shared genetic instruments. In longitudinal analyses the wGRSs for WMH, but not BP or stroke, showed suggestive association with incident all-cause-dementia (HR, 1.02; 95%CI:1.00-1.04; P=.06). BP and stroke wGRSs were strongly associated with mortality but there was no evidence for selective survival bias during follow-up. In secondary analyses, polygenic scores with more liberal instrument definition showed association of both WMH and stroke with all-cause-dementia, AD, and vascular or mixed dementia; associations of stroke, but not WMH, with dementia outcomes were markedly attenuated after adjusting for interim stroke. Conclusion: These findings provide converging evidence that WMH is a leading vascular contributor to dementia risk, which may better capture the brain damage caused by BP (and other etiologies) than BP itself and should be targeted in priority for dementia prevention in the population.

10.
BMC Med Res Methodol ; 23(1): 199, 2023 09 05.
Article En | MEDLINE | ID: mdl-37670234

BACKGROUND: Alzheimer's disease and related dementia (ADRD) are characterized by multiple and progressive anatomo-clinical changes including accumulation of abnormal proteins in the brain, brain atrophy and severe cognitive impairment. Understanding the sequence and timing of these changes is of primary importance to gain insight into the disease natural history and ultimately allow earlier diagnosis. Yet, modeling changes over disease course from cohort data is challenging as the usual timescales (time since inclusion, chronological age) are inappropriate and time-to-clinical diagnosis is available on small subsamples of participants with short follow-up durations prior to diagnosis. One solution to circumvent this challenge is to define the disease time as a latent variable. METHODS: We developed a multivariate mixed model approach that realigns individual trajectories into the latent disease time to describe disease progression. In contrast with the existing literature, our methodology exploits the clinical diagnosis information as a partially observed and approximate reference to guide the estimation of the latent disease time. The model estimation was carried out in the Bayesian Framework using Stan. We applied the methodology to the MEMENTO study, a French multicentric clinic-based cohort of 2186 participants with 5-year intensive follow-up. Repeated measures of 12 ADRD markers stemmed from cerebrospinal fluid (CSF), brain imaging and cognitive tests were analyzed. RESULTS: The estimated latent disease time spanned over twenty years before the clinical diagnosis. Considering the profile of a woman aged 70 with a high level of education and APOE4 carrier (the main genetic risk factor for ADRD), CSF markers of tau proteins accumulation preceded markers of brain atrophy by 5 years and cognitive decline by 10 years. However we observed that individual characteristics could substantially modify the sequence and timing of these changes, in particular for CSF level of A[Formula: see text]. CONCLUSION: By leveraging the available clinical diagnosis timing information, our disease progression model does not only realign trajectories into the most homogeneous way. It accounts for the inherent residual inter-individual variability in dementia progression to describe the long-term anatomo-clinical degradations according to the years preceding clinical diagnosis, and to provide clinically meaningful information on the sequence of events. TRIAL REGISTRATION: clinicaltrials.gov, NCT01926249. Registered on 16 August 2013.


Alzheimer Disease , Cognitive Dysfunction , Female , Humans , Bayes Theorem , Educational Status , Disease Progression
11.
Proc Natl Acad Sci U S A ; 120(36): e2302720120, 2023 09 05.
Article En | MEDLINE | ID: mdl-37643212

Across multiancestry groups, we analyzed Human Leukocyte Antigen (HLA) associations in over 176,000 individuals with Parkinson's disease (PD) and Alzheimer's disease (AD) versus controls. We demonstrate that the two diseases share the same protective association at the HLA locus. HLA-specific fine-mapping showed that hierarchical protective effects of HLA-DRB1*04 subtypes best accounted for the association, strongest with HLA-DRB1*04:04 and HLA-DRB1*04:07, and intermediary with HLA-DRB1*04:01 and HLA-DRB1*04:03. The same signal was associated with decreased neurofibrillary tangles in postmortem brains and was associated with reduced tau levels in cerebrospinal fluid and to a lower extent with increased Aß42. Protective HLA-DRB1*04 subtypes strongly bound the aggregation-prone tau PHF6 sequence, however only when acetylated at a lysine (K311), a common posttranslational modification central to tau aggregation. An HLA-DRB1*04-mediated adaptive immune response decreases PD and AD risks, potentially by acting against tau, offering the possibility of therapeutic avenues.


Alzheimer Disease , HLA-DRB1 Chains , Parkinson Disease , Humans , Alzheimer Disease/genetics , Histocompatibility Antigens , HLA Antigens , HLA-DRB1 Chains/genetics , Parkinson Disease/genetics
12.
Alzheimers Dement ; 19(12): 5700-5718, 2023 Dec.
Article En | MEDLINE | ID: mdl-37422285

INTRODUCTION: This study aims to examine whether physical activity moderates the association between biomarkers of brain pathologies and dementia risk. METHODS: From the Memento cohort, we analyzed 1044 patients with mild cognitive impairment, aged 60 and older. Self-reported physical activity was assessed using the International Physical Activity Questionnaire. Biomarkers of brain pathologies comprised medial temporal lobe atrophy (MTA), white matter lesions, and plasma amyloid beta (Aß)42/40 and phosphorylated tau181. Association between physical activity and risk of developing dementia over 5 years of follow-up, and interactions with biomarkers of brain pathologies were tested. RESULTS: Physical activity moderated the association between MTA and plasma Aß42/40 level and increased dementia risk. Compared to participants with low physical activity, associations of both MTA and plasma Aß42/40 on dementia risk were attenuated in participants with high physical activity. DISCUSSION: Although reverse causality cannot be excluded, this work suggests that physical activity may contribute to cognitive reserve. HIGHLIGHTS: Physical activity is an interesting modifiable target for dementia prevention. Physical activity may moderate the impact of brain pathology on dementia risk. Medial temporal lobe atrophy and plasma amyloid beta 42/40 ratio were associated with increased dementia risk especially in those with low level of physical activity.


Alzheimer Disease , Cognitive Dysfunction , Dementia , Humans , Middle Aged , Aged , Dementia/complications , Amyloid beta-Peptides , Magnetic Resonance Imaging , Disease Progression , Cognitive Dysfunction/pathology , Biomarkers , Brain/pathology , Atrophy/pathology , Alzheimer Disease/pathology , tau Proteins
13.
Acta Neuropathol Commun ; 11(1): 68, 2023 04 26.
Article En | MEDLINE | ID: mdl-37101235

Amyloid PET imaging has been crucial for detecting the accumulation of amyloid beta (Aß) deposits in the brain and to study Alzheimer's disease (AD). We performed a genome-wide association study on the largest collection of amyloid imaging data (N = 13,409) to date, across multiple ethnicities from multicenter cohorts to identify variants associated with brain amyloidosis and AD risk. We found a strong APOE signal on chr19q.13.32 (top SNP: APOE ɛ4; rs429358; ß = 0.35, SE = 0.01, P = 6.2 × 10-311, MAF = 0.19), driven by APOE ɛ4, and five additional novel associations (APOE ε2/rs7412; rs73052335/rs5117, rs1081105, rs438811, and rs4420638) independent of APOE ɛ4. APOE ɛ4 and ε2 showed race specific effect with stronger association in Non-Hispanic Whites, with the lowest association in Asians. Besides the APOE, we also identified three other genome-wide loci: ABCA7 (rs12151021/chr19p.13.3; ß = 0.07, SE = 0.01, P = 9.2 × 10-09, MAF = 0.32), CR1 (rs6656401/chr1q.32.2; ß = 0.1, SE = 0.02, P = 2.4 × 10-10, MAF = 0.18) and FERMT2 locus (rs117834516/chr14q.22.1; ß = 0.16, SE = 0.03, P = 1.1 × 10-09, MAF = 0.06) that all colocalized with AD risk. Sex-stratified analyses identified two novel female-specific signals on chr5p.14.1 (rs529007143, ß = 0.79, SE = 0.14, P = 1.4 × 10-08, MAF = 0.006, sex-interaction P = 9.8 × 10-07) and chr11p.15.2 (rs192346166, ß = 0.94, SE = 0.17, P = 3.7 × 10-08, MAF = 0.004, sex-interaction P = 1.3 × 10-03). We also demonstrated that the overall genetic architecture of brain amyloidosis overlaps with that of AD, Frontotemporal Dementia, stroke, and brain structure-related complex human traits. Overall, our results have important implications when estimating the individual risk to a population level, as race and sex will needed to be taken into account. This may affect participant selection for future clinical trials and therapies.


Alzheimer Disease , Amyloidosis , Humans , Female , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/complications , Amyloid beta-Peptides/genetics , Genome-Wide Association Study , Amyloidosis/diagnostic imaging , Amyloidosis/genetics , Amyloid , Apolipoproteins E/genetics
14.
Eur J Epidemiol ; 38(6): 605-615, 2023 Jun.
Article En | MEDLINE | ID: mdl-37099244

Data discovery, the ability to find datasets relevant to an analysis, increases scientific opportunity, improves rigour and accelerates activity. Rapid growth in the depth, breadth, quantity and availability of data provides unprecedented opportunities and challenges for data discovery. A potential tool for increasing the efficiency of data discovery, particularly across multiple datasets is data harmonisation.A set of 124 variables, identified as being of broad interest to neurodegeneration, were harmonised using the C-Surv data model. Harmonisation strategies used were simple calibration, algorithmic transformation and standardisation to the Z-distribution. Widely used data conventions, optimised for inclusiveness rather than aetiological precision, were used as harmonisation rules. The harmonisation scheme was applied to data from four diverse population cohorts.Of the 120 variables that were found in the datasets, correspondence between the harmonised data schema and cohort-specific data models was complete or close for 111 (93%). For the remainder, harmonisation was possible with a marginal a loss of granularity.Although harmonisation is not an exact science, sufficient comparability across datasets was achieved to enable data discovery with relatively little loss of informativeness. This provides a basis for further work extending harmonisation to a larger variable list, applying the harmonisation to further datasets, and incentivising the development of data discovery tools.


Datasets as Topic , Knowledge Discovery , Humans , Reference Standards
15.
Nat Commun ; 14(1): 761, 2023 02 10.
Article En | MEDLINE | ID: mdl-36765056

The anticipation of progression of Alzheimer's disease (AD) is crucial for evaluations of secondary prevention measures thought to modify the disease trajectory. However, it is difficult to forecast the natural progression of AD, notably because several functions decline at different ages and different rates in different patients. We evaluate here AD Course Map, a statistical model predicting the progression of neuropsychological assessments and imaging biomarkers for a patient from current medical and radiological data at early disease stages. We tested the method on more than 96,000 cases, with a pool of more than 4,600 patients from four continents. We measured the accuracy of the method for selecting participants displaying a progression of clinical endpoints during a hypothetical trial. We show that enriching the population with the predicted progressors decreases the required sample size by 38% to 50%, depending on trial duration, outcome, and targeted disease stage, from asymptomatic individuals at risk of AD to subjects with early and mild AD. We show that the method introduces no biases regarding sex or geographic locations and is robust to missing data. It performs best at the earliest stages of disease and is therefore highly suitable for use in prevention trials.


Alzheimer Disease , Cognitive Dysfunction , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/psychology , Disease Progression , Neuroimaging/methods , Research Design , Biomarkers
16.
J Alzheimers Dis ; 92(1): 183-193, 2023.
Article En | MEDLINE | ID: mdl-36710672

BACKGROUND: The long-term effects of traumatic brain injury (TBI) with loss of consciousness (LOC) on magnetic resonance imaging (MRI) markers of brain health and on dementia risk are still debated. OBJECTIVE: To investigate the associations of history of TBI with LOC with incident dementia and neuroimaging markers of brain structure and small vessel disease lesions. METHODS: The analytical sample consisted in 4,144 participants aged 65 and older who were dementia-free at baseline from the Three City -Dijon study. History of TBI with LOC was self-reported at baseline. Clinical Dementia was assessed every two to three years, up to 12 years of follow-up. A subsample of 1,675 participants <80 years old underwent a brain MRI at baseline. We investigated the associations between history of TBI with LOC and 1) incident all cause and Alzheimer's disease (AD) dementia using illness-death models, and 2) neuroimaging markers at baseline. RESULTS: At baseline, 8.3% of the participants reported a history of TBI with LOC. In fully-adjusted models, participants with a history of TBI with LOC had no statistically significant differences in dementia risk (HR = 0.90, 95% CI = 0.60-1.36) or AD risk (HR = 1.03, 95% CI = 0.69-1.52), compared to participants without TBI history. History of TBI with LOC was associated with lower white matter volume (ß= -4.58, p = 0.048), but not with other brain volumes, white matter hyperintensities volume, nor covert brain infarct. CONCLUSION: This study did not find evidence of an association between history of TBI with LOC and dementia or AD dementia risks over 12-year follow-up, brain atrophy, or markers of small vessel disease.


Alzheimer Disease , Brain Injuries, Traumatic , Humans , Aged, 80 and over , Brain Injuries, Traumatic/complications , Brain/pathology , Alzheimer Disease/pathology , Unconsciousness/complications , Magnetic Resonance Imaging
17.
Alzheimers Dement ; 19(6): 2332-2342, 2023 06.
Article En | MEDLINE | ID: mdl-36464896

INTRODUCTION: Approximately 40% of dementia cases could be delayed or prevented acting on modifiable risk factors including hypertension. However, the mechanisms underlying the hypertension-dementia association are still poorly understood. METHODS: We conducted a cross-sectional analysis in 2048 patients from the MEMENTO cohort, a French multicenter clinic-based study of outpatients with either isolated cognitive complaints or mild cognitive impairment. Exposure to hypertension was defined as a combination of high blood pressure (BP) status and antihypertensive treatment intake. Pathway associations were examined through structural equation modeling integrating extensive collection of neuroimaging biomarkers and clinical data. RESULTS: Participants treated with high BP had significantly lower cognition compared to the others. This association was mediated by higher neurodegeneration and higher white matter hyperintensities load but not by Alzheimer's disease (AD) biomarkers. DISCUSSION: These results highlight the importance of controlling hypertension for prevention of cognitive decline and offer new insights on mechanisms underlying the hypertension-dementia association. HIGHLIGHTS: Paths of hypertension-cognition association were assessed by structural equation models. The hypertension-cognition association is not mediated by Alzheimer's disease biomarkers. The hypertension-cognition association is mediated by neurodegeneration and leukoaraiosis. Lower cognition was limited to participants treated with uncontrolled blood pressure. Blood pressure control could contribute to promote healthier brain aging.


Alzheimer Disease , Cognitive Dysfunction , Hypertension , Humans , Alzheimer Disease/metabolism , Cross-Sectional Studies , Positron-Emission Tomography , Magnetic Resonance Imaging , Cognition/physiology , Cognitive Dysfunction/metabolism , Biomarkers , Amyloid beta-Peptides/metabolism
18.
Neurology ; 100(5): e473-e484, 2023 01 31.
Article En | MEDLINE | ID: mdl-36261295

BACKGROUND AND OBJECTIVE: Blood biomarkers for Alzheimer disease (AD) have consistently proven to be associated with CSF or PET biomarkers and effectively discriminate AD from other neurodegenerative diseases. Our aim was to test their utility in clinical practice, from a multicentric unselected prospective cohort where patients presented with a large spectrum of cognitive deficits or complaints. METHODS: The MEMENTO cohort enrolled 2,323 outpatients with subjective cognitive complaint (SCC) or mild cognitive impairment (MCI) consulting in 26 French memory clinics. Participants had neuropsychological assessments, MRI, and blood sampling at baseline. CSF sampling and amyloid PET were optional. Baseline blood Aß42/40 ratio, total tau, p181-tau, and neurofilament light chain (NfL) were measured using a Simoa HD-X analyzer. An expert committee validated incident dementia cases during a 5-year follow-up period. RESULTS: Overall, 2,277 individuals had at least 1 baseline blood biomarker available (n = 357 for CSF subsample, n = 649 for PET subsample), among whom 257 were diagnosed with clinical AD/mixed dementia during follow-up. All blood biomarkers but total tau were mildly correlated with their equivalence in the CSF (r = 0.33 to 0.46, p < 0.0001) and were associated with amyloid-PET status (p < 0.0001). Blood p181-tau was the best blood biomarker to identify amyloid-PET positivity (area under the curve = 0.74 [95% CI = 0.69; 0.79]). Higher blood and CSF p181-tau and NfL concentrations were associated with accelerated time to AD dementia onset with similar incidence rates, whereas blood Aß42/40 was less efficient than CSF Aß42/40. Blood p181-tau alone was the best blood predictor of 5-year AD/mixed dementia risk (c-index = 0.73 [95% CI = 0.69; 0.77]); its accuracy was higher in patients with clinical dementia rating (CDR) = 0 (c-index = 0.83 [95% CI = 0.69; 0.97]) than in patients with CDR = 0.5 (c-index = 0.70 [95% CI = 0.66; 0.74]). A "clinical" reference model (combining demographics and neuropsychological assessment) predicted AD/mixed dementia risk with a c-index = 0.88 [95% CI = 0.86-0.91] and performance increased to 0.90 [95% CI = 0.88; 0.92] when adding blood p181-tau + Aß42/40. A "research" reference model (clinical model + apolipoprotein E genotype and AD signature on MRI) had a c-index = 0.91 [95% CI = 0.89-0.93] increasing to 0.92 [95% CI = 0.90; 0.93] when adding blood p181-tau + Aß42/40. Chronic kidney disease and vascular comorbidities did not affect predictive performances. DISCUSSION: In a clinic-based cohort of patients with SCC or MCI, blood biomarkers may be good hallmarks of underlying pathology but add little to 5-year dementia risk prediction models including traditional predictors.


Alzheimer Disease , Cognitive Dysfunction , Mixed Dementias , Humans , Alzheimer Disease/diagnostic imaging , Prospective Studies , Amyloid beta-Peptides , tau Proteins , Biomarkers , Cognitive Dysfunction/diagnostic imaging , Peptide Fragments
19.
Acta Neuropathol ; 144(5): 821-842, 2022 11.
Article En | MEDLINE | ID: mdl-36066633

Amyloid-beta 42 (Aß42) and phosphorylated tau (pTau) levels in cerebrospinal fluid (CSF) reflect core features of the pathogenesis of Alzheimer's disease (AD) more directly than clinical diagnosis. Initiated by the European Alzheimer & Dementia Biobank (EADB), the largest collaborative effort on genetics underlying CSF biomarkers was established, including 31 cohorts with a total of 13,116 individuals (discovery n = 8074; replication n = 5042 individuals). Besides the APOE locus, novel associations with two other well-established AD risk loci were observed; CR1 was shown a locus for Aß42 and BIN1 for pTau. GMNC and C16orf95 were further identified as loci for pTau, of which the latter is novel. Clustering methods exploring the influence of all known AD risk loci on the CSF protein levels, revealed 4 biological categories suggesting multiple Aß42 and pTau related biological pathways involved in the etiology of AD. In functional follow-up analyses, GMNC and C16orf95 both associated with lateral ventricular volume, implying an overlap in genetic etiology for tau levels and brain ventricular volume.


Alzheimer Disease , Alzheimer Disease/pathology , Amyloid beta-Peptides/cerebrospinal fluid , Apolipoproteins E/genetics , Biomarkers/cerebrospinal fluid , Cell Cycle Proteins , Humans , Peptide Fragments/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , tau Proteins/genetics
20.
Crit Care Explor ; 4(7): e0737, 2022 Jul.
Article En | MEDLINE | ID: mdl-35923591

Studies comparing outcomes of ICU patients admitted for either COVID-19 or seasonal influenza are limited. Our objective was to describe baseline clinical profiles, care procedures, and mortality outcomes by infection status (influenza vs COVID-19) of patients who received invasive mechanical ventilation in the ICU. DESIGN: Retrospective observational study. SETTING: Data were extracted from the Assistance Publique-Hopitaux de Paris database from September 1, 2016, to April 20, 2021. It includes data from the 39 university hospitals. PATIENTS: A total of 752 influenza adult patients and 3,465 COVID-19 adult patients received invasive mechanical ventilation in one of the ICUs of the Paris area university hospitals, France. INTERVENTION: The characteristics and outcome by infection status were compared. Factors associated with mortality were assessed using Cox proportional hazard models after controlling for potential confounders, including infection status. MEASUREMENTS AND MAIN RESULTS: The median age at admission to the ICU was 67 (interquartile range [IQR], 57-77) and 63 yr (IQR, 54-71 yr) for influenza and COVID-19 patients, respectively. At ICU admission, COVID-19 patients were more frequently obese, more frequently had diabetes mellitus or high blood pressure, and were less likely to have chronic heart failure, chronic respiratory disease, chronic kidney failure, or active cancer than influenza patients. The overall survival at 90 days was 57% for COVID-19 patients and 66% for influenza patients (p < 0.001). In a multivariable Cox model, higher age, organ transplant, severe acute respiratory syndrome coronavirus 2 infection, and chronic kidney failure were associated with shorter survival, whereas obesity and high blood pressure were associated with longer survival after invasive ventilation. CONCLUSIONS: COVID-19 and influenza patients requiring mechanical ventilation in the ICU differed by many characteristics. COVID-19 patients showed lower survival independently of potential confounders.

...