Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Am Med Dir Assoc ; 24(5): 638-644.e1, 2023 05.
Article in English | MEDLINE | ID: mdl-33239240

ABSTRACT

OBJECTIVE: In cognitively normal adults, nutritional parameters are related to cognitive decline and incidence of dementia. Studies on the role of nutrition in predementia stages subjective cognitive decline and mild cognitive impairment, and mild stages of Alzheimer's disease (AD) dementia in a clinical setting are lacking. In the absence of a curative treatment, this evidence is important for targeting nutritional factors to potentially prevent or delay further cognitive decline. Our aim is to investigate associations of nutritional parameters with clinical progression in patients ranging from those who are cognitively normal to those who have AD dementia. DESIGN: Longitudinal. SETTING AND PARTICIPANTS: Memory clinic, 551 patients (219 with subjective cognitive decline, 135 with mild cognitive impairment, and 197 with AD dementia), mean age 64 ± 8 years. MEASUREMENTS: We assessed body mass index, fat-free mass, Mini-Nutritional Assessment, and dietary intake with the Dutch Healthy Diet food frequency questionnaire and the 238-item healthy life in an urban setting (HELIUS) food frequency questionnaire at baseline. Cox proportional hazard models were used to evaluate associations of nutritional parameters with clinical progression. Additional analyses were restricted to patients who were amyloid positive. RESULTS: We observed clinical progression in 170 patients (31%) over 2.2 ± 0.9 years. Poorer Mini-Nutritional Assessment score [hazard ratio (95% confidence interval) 1.39 (1.18-1.64)], lower body mass index [1.15 (0.96-1.38)], lower fat-free mass [1.40 (0.93-2.10)], and a less healthy dietary pattern [1.22 (1.01-1.48)] were associated with a higher risk of clinical progression. Similar effect sizes were found in patients who were amyloid positive. CONCLUSIONS AND IMPLICATIONS: Poorer nutritional status and a less healthy dietary pattern are associated with a higher risk of clinical progression. This study provides support for investigating whether improving nutritional status can alter the clinical trajectory of AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Middle Aged , Aged , Alzheimer Disease/epidemiology , Nutritional Status , Cognitive Dysfunction/epidemiology , Diet , Disease Progression
2.
Alzheimers Dement ; 17(2): 205-214, 2021 02.
Article in English | MEDLINE | ID: mdl-32886448

ABSTRACT

INTRODUCTION: Our aim was to study whether systemic metabolites are associated with magnetic resonance imaging (MRI) measures of brain and hippocampal atrophy and white matter hyperintensities (WMH). METHODS: We studied associations of 143 plasma-based metabolites with MRI measures of brain and hippocampal atrophy and WMH in three independent cohorts (n = 3962). We meta-analyzed the results of linear regression analyses to determine the association of metabolites with MRI measures. RESULTS: Higher glucose levels and lower levels of three small high density lipoprotein (HDL) particles were associated with brain atrophy. Higher glucose levels were associated with WMH. DISCUSSION: Glucose levels were associated with brain atrophy and WMH, and small HDL particle levels were associated with brain atrophy. Circulating metabolites may aid in developing future intervention trials.


Subject(s)
Alzheimer Disease , Atrophy/pathology , Blood Glucose/metabolism , Brain/pathology , White Matter/pathology , Aged , Alzheimer Disease/blood , Alzheimer Disease/pathology , Female , Hippocampus/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged
3.
Front Immunol ; 12: 794519, 2021.
Article in English | MEDLINE | ID: mdl-35173707

ABSTRACT

Introduction: Several studies have reported alterations in gut microbiota composition of Alzheimer's disease (AD) patients. However, the observed differences are not consistent across studies. We aimed to investigate associations between gut microbiota composition and AD biomarkers using machine learning models in patients with AD dementia, mild cognitive impairment (MCI) and subjective cognitive decline (SCD). Materials and Methods: We included 170 patients from the Amsterdam Dementia Cohort, comprising 33 with AD dementia (66 ± 8 years, 46%F, mini-mental state examination (MMSE) 21[19-24]), 21 with MCI (64 ± 8 years, 43%F, MMSE 27[25-29]) and 116 with SCD (62 ± 8 years, 44%F, MMSE 29[28-30]). Fecal samples were collected and gut microbiome composition was determined using 16S rRNA sequencing. Biomarkers of AD included cerebrospinal fluid (CSF) amyloid-beta 1-42 (amyloid) and phosphorylated tau (p-tau), and MRI visual scores (medial temporal atrophy, global cortical atrophy, white matter hyperintensities). Associations between gut microbiota composition and dichotomized AD biomarkers were assessed with machine learning classification models. The two models with the highest area under the curve (AUC) were selected for logistic regression, to assess associations between the 20 best predicting microbes and the outcome measures from these machine learning models while adjusting for age, sex, BMI, diabetes, medication use, and MMSE. Results: The machine learning prediction for amyloid and p-tau from microbiota composition performed best with AUCs of 0.64 and 0.63. Highest ranked microbes included several short chain fatty acid (SCFA)-producing species. Higher abundance of [Clostridium] leptum and lower abundance of [Eubacterium] ventriosum group spp., Lachnospiraceae spp., Marvinbryantia spp., Monoglobus spp., [Ruminococcus] torques group spp., Roseburia hominis, and Christensenellaceae R-7 spp., was associated with higher odds of amyloid positivity. We found associations between lower abundance of Lachnospiraceae spp., Lachnoclostridium spp., Roseburia hominis and Bilophila wadsworthia and higher odds of positive p-tau status. Conclusions: Gut microbiota composition was associated with amyloid and p-tau status. We extend on recent studies that observed associations between SCFA levels and AD CSF biomarkers by showing that lower abundances of SCFA-producing microbes were associated with higher odds of positive amyloid and p-tau status.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Brain-Gut Axis , Disease Susceptibility , Gastrointestinal Microbiome , Aged , Alzheimer Disease/diagnosis , Biomarkers , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/etiology , Comorbidity , Dementia/diagnosis , Dementia/etiology , Female , Humans , Machine Learning , Male , Middle Aged , Risk Factors
4.
Alzheimers Dement (N Y) ; 6(1): e12021, 2020.
Article in English | MEDLINE | ID: mdl-32864412

ABSTRACT

INTRODUCTION: Higher brain tocopherol levels have been associated with lower levels of Alzheimer's disease (AD) neuropathology; however, the underlying mechanisms are unclear. METHODS: We studied the relations of α- and γ-tocopherol brain levels to microglia density in 113 deceased participants from the Memory and Aging Project. We used linear regression analyses to examine associations between tocopherol levels and microglia densities in a basic model adjusted for age, sex, education, apolipoprotein E (APOE)ε4 genotype (any ε4 allele vs. none) , and post-mortem time interval, and a second model additionally adjusted for total amyloid load and neurofibrillary tangle severity. RESULTS: Higher α- and γ-tocopherol levels were associated with lower total and activated microglia density in cortical but not in subcortical brain regions. The association between cortical α-tocopherol and total microglia density remained statistically significant after adjusting for AD neuropathology. DISCUSSION: These results suggest that the relation between tocopherols and AD might be partly explained by the alleviating effects of tocopherols on microglia activation.

5.
Alzheimers Res Ther ; 12(1): 116, 2020 09 26.
Article in English | MEDLINE | ID: mdl-32979927

ABSTRACT

BACKGROUND: Malnutrition is common in patients with Alzheimer's disease (AD) dementia and mild cognitive impairment (MCI) and is associated with institutionalization and increased mortality. Malnutrition is the result of a negative energy balance, which could be due to reduced dietary intake and/or higher energy expenditure. To study underlying mechanisms for malnutrition, we investigated dietary intake and resting energy expenditure (REE) of patients with AD dementia, MCI, and controls. In addition, we studied associations of global cognition (Mini-Mental State Examination (MMSE)) and AD biomarkers with dietary intake and REE. METHODS: We included 219 participants from the NUDAD project, 71 patients with AD dementia (age 68 ± 8 years, 58% female, MMSE 24 ± 3), 52 with MCI (67 ± 8 years, 42% female, MMSE 26 ± 2), and 96 controls (62 ± 7 years, 52% female, MMSE 28 ± 2). We used a 238-item food frequency questionnaire to assess dietary intake (energy, protein, carbohydrate, and fat). In a subgroup of 92 participants (30 patients with AD dementia, 22 with MCI, and 40 controls) we measured REE with indirect calorimetry. Between-group differences in dietary intake and REE were tested with ANOVAs. In the total sample, linear regression analyses were used to explore potential associations of MMSE score and AD biomarkers with dietary intake and REE. All analyses were adjusted for age, sex, education, and body mass index or fat-free mass. RESULTS: Patients with AD dementia and MCI did not differ from controls in total energy intake (1991 ± 71 and 2172 ± 80 vs 2022 ± 61 kcal/day, p > 0.05) nor in protein, carbohydrate, or fat intake. Patients with AD dementia and MCI had a higher REE than controls (1704 ± 41 and 1754 ± 47 vs 1569 ± 34 kcal/day, p < 0.05). We did not find any association of MMSE score or AD biomarkers with dietary intake or REE. CONCLUSIONS: We found a higher REE, despite similar energy intake in patients with AD and MCI compared to controls. These findings suggest that elevated metabolism rather than reduced energy intake explains malnutrition in AD. These results could be useful to optimize dietary advice for patients with AD dementia and MCI.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Aged , Alzheimer Disease/complications , Cognition , Energy Intake , Female , Health Expenditures , Humans , Male , Middle Aged
6.
Alzheimers Dement (Amst) ; 12(1): e12063, 2020.
Article in English | MEDLINE | ID: mdl-32793798

ABSTRACT

INTRODUCTION: Weight loss is associated with higher mortality and progression of cognitive decline, but its associations with magnetic resonance imaging (MRI) changes related to Alzheimer's disease (AD) are unknown. METHODS: We included 412 patients from the NUDAD project, comprising 129 with AD dementia, 107 with mild cognitive impairment (MCI), and 176 controls. Associations between nutritional status and MRI measures were analyzed using linear regression, adjusted for age, sex, education, cognitive functioning, and cardiovascular risk factors. RESULTS: Lower body mass index (BMI), fat mass (FM), and fat free mass index were associated with higher medial temporal atrophy (MTA) scores. Lower BMI, FM, and waist circumference were associated with more microbleeds. Stratification by diagnosis showed that the observed associations with microbleeds were only significant in MCI. DISCUSSION: Lower indicators of nutritional status were associated with more MTA and microbleeds, with largest effect sizes in MCI.

7.
J Alzheimers Dis ; 77(2): 619-627, 2020.
Article in English | MEDLINE | ID: mdl-32741813

ABSTRACT

BACKGROUND: Higher vitamin E intake has been widely related to lower risks of cognitive decline and dementia. Animal models suggest that this relationship might be (partially) explained by the protection of vitamin E against presynaptic protein oxidation. OBJECTIVE: In this cross-sectional study, we aimed to examine the associations between brain tocopherols and presynaptic protein levels in elderly humans. METHODS: We examined associations of α- and γ-tocopherol brain levels with presynaptic protein levels in 113 deceased participants (age 88.5±6.0 years, 45 (40%) female) from the prospective Memory and Aging project. Three distinct presynaptic proteins, a SNARE protein composite, a synaptotagmin synaptophysin composite and the protein-protein interaction between synaptosomal-associated protein 25 (SNAP-25), and syntaxin were measured in two cortical brain regions. Linear regression models assessed associations of brain tocopherols with presynaptic protein levels. RESULTS: Higher brain γ-tocopherol levels were associated with higher levels of the SNARE protein composite, complexin-I, complexin-II, the synaptotagmin synaptophysin composite, and septin-5 in the midfrontal cortex (B(SE) = 0.272 to 0.412 (0.084 to 0.091), p < 0.001 to 0.003). When additionally adjusted for global Alzheimer's disease pathology, cerebral infarcts, and Lewy body disease pathology, these associations remained largely similar. No associations were found between α-tocopherol and presynaptic protein levels. CONCLUSION: In this cross-sectional study, we found higher brain γ-tocopherol levels were associated with presynaptic protein levels in the midfrontal cortex. These results are consistent with a proposed role of vitamin E to maintain presynaptic protein levels.


Subject(s)
Frontal Lobe/metabolism , Membrane Proteins/metabolism , Presynaptic Terminals/metabolism , gamma-Tocopherol/administration & dosage , gamma-Tocopherol/metabolism , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Frontal Lobe/drug effects , Frontal Lobe/pathology , Humans , Male , Presynaptic Terminals/drug effects , Presynaptic Terminals/pathology , Single-Blind Method , Surveys and Questionnaires
9.
Nutrients ; 12(3)2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32155876

ABSTRACT

Nutrition is one of the modifiable risk factors for cognitive decline and Alzheimer's disease (AD) dementia, and is therefore highly relevant in the context of prevention. However, knowledge of dietary quality in clinical populations on the spectrum of AD dementia is lacking, therefore we studied the association between dietary quality and cognitive impairment in Alzheimer's disease (AD) dementia, mild cognitive impairment (MCI) and controls. We included 357 participants from the NUDAD project (134 AD dementia, 90 MCI, 133 controls). We assessed adherence to dietary guidelines (components: vegetables, fruit, fibers, fish, saturated fat, trans-fat, salt, and alcohol), and cognitive performance (domains: memory, language, visuospatial functioning, attention, and executive functioning). In the total population, linear regression analyses showed a lower vegetable intake is associated with poorer global cognition, visuospatial functioning, attention and executive functioning. In AD dementia, lower total adherence to dietary guidelines and higher alcohol intake were associated with poorer memory, a lower vegetable intake with poorer global cognition and executive functioning, and a higher trans-fat intake with poorer executive functioning. In conclusion, a suboptimal diet is associated with more severely impaired cognition-this association is mostly attributable to a lower vegetable intake and is most pronounced in AD dementia.


Subject(s)
Alzheimer Disease/etiology , Cognitive Dysfunction/etiology , Diet, Healthy , Eating , Elder Nutritional Physiological Phenomena , Feeding Behavior , Malnutrition/complications , Recommended Dietary Allowances , Vegetables , Alcohol Drinking/adverse effects , Alzheimer Disease/psychology , Cognition , Cognitive Dysfunction/psychology , Dietary Fats/adverse effects , Executive Function , Female , Humans , Male , Patient Compliance , Risk Factors , Trans Fatty Acids/adverse effects
10.
J Am Med Dir Assoc ; 21(10): 1513.e1-1513.e17, 2020 10.
Article in English | MEDLINE | ID: mdl-32001171

ABSTRACT

OBJECTIVES: Nutritional insufficiencies have been associated with cognitive impairment. Understanding whether nutritional biomarker levels are associated with clinical progression could help to design dietary intervention trials. This longitudinal study examined a panel of nutritional biomarkers in relation to clinical progression in patients with subjective cognitive decline (SCD) or mild cognitive impairment (MCI). DESIGN, SETTING AND PARTICIPANTS: We included 299 patients without dementia (n = 149 SCD; age 61 ± 10 years, female 44%, n = 150 MCI; age 66 ± 8 years, female 38%). Median (interquartile range) follow-up was 3 (2-5) years. METHODS: We measured 28 nutritional biomarkers in blood and 5 in cerebrospinal fluid (CSF), associated with 3 Alzheimer's disease pathologic processes: vascular change (lipids), synaptic dysfunction (homocysteine-related metabolites), and oxidative stress (minerals and vitamins). Nutritional biomarker associations with clinical progression to MCI/dementia and cognitive decline based on the Mini-Mental State Examination score were evaluated using Cox proportional hazard models and linear mixed models. We used partial least squares Cox models (PLS-Cox) to examine nutritional biomarker profiles associated with clinical progression. RESULTS: In the total group, high high-density lipoprotein (HDL) levels were associated with clinical progression and cognitive decline. In SCD, high folate and low bilirubin levels were associated with cognitive decline. In MCI, low CSF S-adenosylmethionine (SAM) and high theobromine were associated with clinical progression to dementia and high HDL, cholesterol, iron, and 1,25(OH)2 vitamin D were associated with cognitive decline. PLS-Cox showed 1 profile for SCD, characterized by high betaine and folate and low zinc associated with clinical progression. In MCI, a profile with high theobromine and HDL and low triglycerides and a second profile with high plasma SAM and low cholesterol were associated with risk of dementia. CONCLUSION AND IMPLICATIONS: High HDL was most consistently associated with clinical progression. Moreover, different nutritional biomarker profiles for SCD and MCI showed promising associations with clinical progression. Future dietary (intervention) studies could use nutritional biomarker profiles to select patients, taking into account the disease stage.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Aged , Biomarkers , Cognitive Dysfunction/diagnosis , Disease Progression , Female , Humans , Longitudinal Studies , Middle Aged , Neuropsychological Tests
11.
Alzheimers Dement (Amst) ; 12(1): e12120, 2020.
Article in English | MEDLINE | ID: mdl-33392381

ABSTRACT

INTRODUCTION: We examined associations between nutritional biomarkers and clinical progression in individuals with subjective cognitive decline (SCD), mild cognitive impairment (MCI), and Alzheimer's disease (AD)-type dementia. METHODS: We included 528 individuals (64 ± 8 years, 46% F, follow-up 2.1 ± 0.87 years) with SCD (n = 204), MCI (n = 130), and AD (n = 194). Baseline levels of cholesterol, triglycerides, glucose, homocysteine, folate, vitamin A, B12, E and uridine were measured in blood and S-adenosylmethionine and S-adenosylhomocysteine in cerebrospinal fluid. We determined associations between nutritional biomarkers and clinical progression using Cox proportional hazard models. RESULTS: Twenty-two (11%) patients with SCD, 45 (35%) patients with MCI, and 100 (52%) patients with AD showed clinical progression. In SCD, higher levels of low-density lipoprotein (LDL) cholesterol were associated with progression (hazard ratio [HR] [95% confidence interval (CI)] 1.88 [1.04 to 3.41]). In AD, lower uridine levels were associated with progression (0.79 [0.63 to 0.99]). DISCUSSION: Our findings suggest that LDL cholesterol and uridine play a-stage-dependent-role in the clinical progression of AD.

12.
Nutrients ; 11(5)2019 May 11.
Article in English | MEDLINE | ID: mdl-31083522

ABSTRACT

As nutrition is one of the modifiable risk factors for cognitive decline, we studied the relationship between dietary quality and clinical characteristics in cognitively normal individuals with subjective cognitive decline (SCD). We included 165 SCD subjects (age: 64 ± 8 years; 45% female) from the SCIENCe project, a prospective memory clinic based cohort study on SCD. The Dutch Healthy Diet Food Frequency Questionnaire (DHD-FFQ) was used to assess adherence to Dutch guidelines on vegetable, fruit, fibers, fish, saturated fat, trans fatty acids, salt and alcohol intake (item score 0-10, higher score indicating better adherence). We measured global cognition (Mini Mental State Examination), cognitive complaints (Cognitive Change Index self-report; CCI) and depressive symptoms (Center for Epidemiologic Studies Depression Scale; CES-D). Using principal component analysis, we identified dietary components and investigated their relation to clinical characteristics using linear regression models adjusted for age, sex and education. We identified three dietary patterns: (i) "low-Fat-low-Salt", (ii) "high-Veggy", and (iii) "low-Alcohol-low-Fish". Individuals with lower adherence on "low-Fat-low-Salt" had more depressive symptoms (ß -0.18 (-2.27--0.16)). Higher adherence to "high-Veggy" was associated with higher MMSE scores (ß 0.30 (0.21-0.64)). No associations were found with the low-Alcohol-low-Fish component. We showed that in SCD subjects, dietary quality was related to clinically relevant outcomes. These findings could be useful to identify individuals that might benefit most from nutritional prevention strategies to optimize brain health.


Subject(s)
Cognition , Cognitive Dysfunction/prevention & control , Diagnostic Self Evaluation , Diet/classification , Feeding Behavior , Memory , Aged , Brain , Cognitive Dysfunction/complications , Cross-Sectional Studies , Dementia/prevention & control , Depression/complications , Depression/prevention & control , Diet Surveys , Female , Humans , Male , Middle Aged , Netherlands , Principal Component Analysis , Prospective Studies , Vegetables
13.
Nutrients ; 11(5)2019 May 23.
Article in English | MEDLINE | ID: mdl-31126170

ABSTRACT

As malnutrition is common in patients with Alzheimer's disease (AD), we evaluated nutritional status and body composition of patients with AD, mild cognitive impairment (MCI) and controls, and studied associations of AD biomarkers and cognitive performance with nutritional status and body composition. We included 552 participants, of which 198 patients had AD, 135 patients had MCI and 219 controls. We assessed nutritional status (mini nutritional assessment (MNA)) and body composition (body mass index (BMI), fat-free mass (FFM) and waist circumference). Linear regression analyses (adjusted for age, gender and education where appropriate) were applied to test associations of AD biomarkers and cognitive performance on five domains with nutritional parameters (dependent). Patients with MCI and AD had a lower BMI and MNA score than controls. Worse performance in all cognitive domains was associated with lower MNA score, but not with body composition. AD biomarkers were associated with MNA score, BMI and waist circumference, and associations with MNA score remained after adjustment for cognitive performance. Both AD biomarkers and cognitive performance were associated with nutritional status, associations with AD biomarkers remained after adjustment for cognition. Our data suggest that malnutrition is not only related to impaired cognition but also to AD pathology.


Subject(s)
Alzheimer Disease/psychology , Body Composition , Cognition , Cognitive Dysfunction/psychology , Malnutrition/physiopathology , Nutritional Status , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/cerebrospinal fluid , Apolipoprotein E4/genetics , Biomarkers/cerebrospinal fluid , Body Mass Index , Case-Control Studies , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Netherlands , Peptide Fragments/cerebrospinal fluid , Phosphorylation , Prospective Studies , Risk Factors , Waist Circumference , tau Proteins/cerebrospinal fluid
14.
Alzheimers Dement ; 14(6): 707-722, 2018 06.
Article in English | MEDLINE | ID: mdl-29316447

ABSTRACT

INTRODUCTION: Identifying circulating metabolites that are associated with cognition and dementia may improve our understanding of the pathogenesis of dementia and provide crucial readouts for preventive and therapeutic interventions. METHODS: We studied 299 metabolites in relation to cognition (general cognitive ability) in two discovery cohorts (N total = 5658). Metabolites significantly associated with cognition after adjusting for multiple testing were replicated in four independent cohorts (N total = 6652), and the associations with dementia and Alzheimer's disease (N = 25,872) and lifestyle factors (N = 5168) were examined. RESULTS: We discovered and replicated 15 metabolites associated with cognition including subfractions of high-density lipoprotein, docosahexaenoic acid, ornithine, glutamine, and glycoprotein acetyls. These associations were independent of classical risk factors including high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides, glucose, and apolipoprotein E (APOE) genotypes. Six of the cognition-associated metabolites were related to the risk of dementia and lifestyle factors. DISCUSSION: Circulating metabolites were consistently associated with cognition, dementia, and lifestyle factors, opening new avenues for prevention of cognitive decline and dementia.


Subject(s)
Biomarkers/metabolism , Cognitive Dysfunction/metabolism , Dementia/metabolism , Adult , Aged , Alzheimer Disease/metabolism , Cohort Studies , Female , Humans , Life Style , Male , Middle Aged , Reproducibility of Results , Risk Factors
15.
Alzheimers Dement (Amst) ; 8: 196-207, 2017.
Article in English | MEDLINE | ID: mdl-28951883

ABSTRACT

INTRODUCTION: Identification of blood-based metabolic changes might provide early and easy-to-obtain biomarkers. METHODS: We included 127 Alzheimer's disease (AD) patients and 121 control subjects with cerebrospinal fluid biomarker-confirmed diagnosis (cutoff tau/amyloid ß peptide 42: 0.52). Mass spectrometry platforms determined the concentrations of 53 amine compounds, 22 organic acid compounds, 120 lipid compounds, and 40 oxidative stress compounds. Multiple signatures were assessed: differential expression (nested linear models), classification (logistic regression), and regulatory (network extraction). RESULTS: Twenty-six metabolites were differentially expressed. Metabolites improved the classification performance of clinical variables from 74% to 79%. Network models identified five hubs of metabolic dysregulation: tyrosine, glycylglycine, glutamine, lysophosphatic acid C18:2, and platelet-activating factor C16:0. The metabolite network for apolipoprotein E (APOE) ε4 negative AD patients was less cohesive compared with the network for APOE ε4 positive AD patients. DISCUSSION: Multiple signatures point to various promising peripheral markers for further validation. The network differences in AD patients according to APOE genotype may reflect different pathways to AD.

SELECTION OF CITATIONS
SEARCH DETAIL
...