1
|
Abdullah L, Zhou Z, Hall J, Petersen M, Zhang F, O'Bryant S. Association of Alzheimer's disease biomarkers with low premorbid intellectual functioning in a multi-ethnic community-dwelling cohort: A cross-sectional study of HABS-HD. J Alzheimers Dis 2025; 104:1201-1211. [PMID: 40116640 DOI: 10.1177/13872877251322966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Background: Individuals with intellectual disability (ID) may have a five-fold increased risk for developing Alzheimer's disease (AD). However, studies investigating brain aging among individuals with ID without Down syndrome (DS) are lacking. To begin addressing this gap, our study utilized word reading, a widely recognized indicator of an individual's premorbid intellectual ability (pIQ), to examine the effects of ID without DS on plasma AD biomarker outcomes. Objective: To investigate the relationship between premorbid intellectual ability (pIQ) and plasma AD biomarkers in individuals with ID without DS, while considering ethnic differences in these associations. Methods: Participants from the Health & Aging Brain Study - Health Disparities (HABS-HD) were categorized into low (z ≤ -2.00) or average (z = 0.00 ± 1.00) pIQ groups based on word reading scores. Plasma biomarkers including Aβ40, Aβ42, Aβ42/40, phosphorylated tau 181 (p-Tau181), neurofilament light chain (NfL), and total tau (t-tau) were assayed using Simoa technology. Results: Individuals with low pIQ exhibited significantly higher levels of p-Tau181 (p < 0.05), NfL (p < 0.05), and t-tau (p < 0.05) compared to those with average pIQ. Stratified analysis by ethnicity revealed differential associations, with Hispanic and non-Hispanic White (NHW) participants showing distinct biomarker profiles relative to non-Hispanic Black (NHB) individuals. Conclusions: The findings demonstrate that low pIQ is a reliable factor associated with plasma AD biomarker outcomes. Ethnicity appears to modulate these associations, suggesting complex interactions between factors driving AD susceptibility across diverse populations. This study highlights the importance of considering both pIQ and ethnicity in neurodegenerative processes, particularly in individuals with non-DS intellectual developmental disability.
Collapse
Affiliation(s)
- Lubnaa Abdullah
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Zhengyang Zhou
- Department of Public Health, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - James Hall
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Melissa Petersen
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Fan Zhang
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Sid O'Bryant
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
2
|
Jeong S, Lin L, Leone AP, Hsu YH. Type 2 diabetes and late-onset Alzheimer's disease and related dementia: A longitudinal cohort study integrating polygenic risk score. J Alzheimers Dis 2025:13872877251326107. [PMID: 40129417 DOI: 10.1177/13872877251326107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
BackgroundThe inherent genetic effects were not established between type 2 diabetes (T2DM) and Alzheimer's disease and related dementia (ADRD).ObjectiveWe aimed to investigate the association between T2DM and ADRD by integrating T2DM polygenic risk score (PRS) and applying matching in every subgroup.MethodsWe utilized UK Biobank First-occurrences datasets. T2DM were 1:1 matched to non-T2DM using propensity scores generated by 8 covariates; age at diagnosis, sex, cerebrovascular disease, ischemic heart disease, hypertensive disorders, lipid disorders, obesity, and mood disorders. T2DM PRS was additionally matched in T2DM PRS matched analysis. Subgroup analyses by age at diagnosis, sex, and APOE4 genotype were performed with the same matching criteria within each subgroup. Cox proportional hazard and Fine & Gray competing risk model were utilized.ResultsIn T2DM PRS unmatched cohort, 24,583 T2DM were 1:1 matched to non-T2DM. The mean age at diagnosis was around 62 years old, with females constituting around 40%. Up to 25-year follow-up, ADRD rate/1000 person-years was 0.88 versus 1.52 (Non-T2DM versus T2DM); PRS unmatched (cHR: 1.72, 95% CI: 1.46-2.03) and matched (cHR:1.75, 95% CI: 1.47-2.09). Except for older age onset (≥75 years), the other subgroups demonstrated significantly increased ADRD risks in T2DM. T2DM PRS was higher in non-ADRD group across all subgroups. Contrarily, T2DM PRS was higher in ADRD in younger onset group (<55 years).ConclusionsT2DM is one of the strong risk factors of ADRD but genetic T2DM effect does not contribute to ADRD risk. However, a genetic link might be present in younger age onset group.
Collapse
Affiliation(s)
- Sohyun Jeong
- Department of Pharmacy Practice, College of Pharmacy, Massachusetts College of Pharmacy and Health Science, Boston, MA, USA
| | - Lisha Lin
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
| | - Alvaro-Pascual Leone
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, MA, USA
| | - Yi-Hsiang Hsu
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| |
Collapse
|
3
|
Liu M, Ma LY, Li QY, Huang LY, Hu HY, Tan L, Hu H. Associations of varicose veins with cerebrospinal fluid biomarkers of Alzheimer's disease pathologies in adults without dementia: the CABLE study. Front Aging Neurosci 2025; 17:1502154. [PMID: 40103929 PMCID: PMC11913850 DOI: 10.3389/fnagi.2025.1502154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/14/2025] [Indexed: 03/20/2025] Open
Abstract
Background Previous studies have found a correlation between varicose veins (VVs) and cognitive decline, and individuals with VVs have a higher prevalence of Alzheimer's disease (AD). However, the associations between VVs and the core pathologies of AD have not yet been investigated. The research was designed to analyze the relationships between VVs and cerebrospinal fluid (CSF) biomarkers of AD pathologies. Methods We included 1,298 participants from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) database without dementia. Multiple linear regression (MLR) model was applied to assess the relationships between the VVs and CSF AD biomarkers. Then, we conducted subgroup analyses according to age, gender, education levels and apolipoprotein E genotype ε4 (APOE-ε4) carrier status. Additionally, mediation effects were assessed using causal mediation analyses with 10,000 bootstrapped iterations. Results In total subjects, VVs had negative correlations with CSF Aβ42 (β = -0.157, p = 0.038) and CSF Aβ42/Aβ40 ratio (β = -0.272, p < 0.001), as well as positive correlations with CSF Aβ40 (β = 0.170, p = 0.024), CSF p-tau (β = 0.192, p = 0.008), CSF t-tau/Aβ42 ratio (β = 0.190, p = 0.011), and CSF p-tau/Aβ42 ratio (β = 0.248, p = 0.001), after adjusting for age, sex, education levels and APOE-ε4 carrier status. Subgroup analyses demonstrated that the relations between VVs and CSF AD biomarkers were more significant in female, mid-life adults (40-65 years), less-educated individuals and APOE-ε4 non-carriers. Moreover, CSF Aβ42/Aβ40 ratio might be a partial mediator of the association between VVs and p-tau pathology. Conclusion Our study found correlations between VVs and CSF AD biomarkers, suggesting that VVs may be a potential risk factor for the development of AD.
Collapse
Affiliation(s)
- Min Liu
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Li-Yun Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Qiong-Yao Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Han X, Li Y, Wang J, Liu X, Zhang Y, Dong Q, Song Y, Cong L, Zhang Q, Tang S, Song L, Hou T, Wang Y, Du Y, Qiu C. Associations between lifelong cognitive reserve, Alzheimer's disease-related plasma biomarkers, and cognitive function in dementia-free older adults: A population-based study. J Alzheimers Dis 2025; 103:821-832. [PMID: 39791378 DOI: 10.1177/13872877241306448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
BACKGROUND Cognitive reserve (CR), typically measured through socio-behavioral proxies, can partially explain better cognitive performance despite underlying brain aging or neuropathology. OBJECTIVE To examine the associations of CR with mild cognitive impairment (MCI) and cognitive function while considering Alzheimer's disease (AD)-related plasma biomarkers. METHODS This population-based cross-sectional study included 4706 dementia-free individuals from MIND-China. Data on AD-related plasma biomarkers were available for 1204 individuals. A composite CR score was generated by integrating education, occupational complexity, mental activity, and social support, using structural equation modeling. A neuropsychological test battery was used to assess the function of episodic memory, executive function, attention, and verbal fluency. MCI and subtypes of MCI were defined according to the Petersen's criteria. Data were analyzed using general linear and logistic regression models. RESULTS Controlling for AD-related plasma biomarkers, higher educational attainment was associated with better performance in all four examined cognitive domains (p < 0.001) and with lower likelihoods of MCI, amnestic MCI (aMCI), and non-aMCI (p < 0.05); late-life mental activity was significantly related to lower likelihoods of MCI and aMCI (p < 0.05). Midlife occupation and late-life social support were not significantly associated with MCI or subtypes (p > 0.05). Higher composite CR scores were related to better performance in all the examined cognitive domains as well as lower likelihoods of MCI, aMCI, and non-aMCI (p < 0.05). CONCLUSIONS Greater composite CR, derived from the CR indicators across different stages of the lifespan, is associated with better cognitive function independent of AD-related plasma biomarkers, driven mainly by early-life educational attainment.
Collapse
Affiliation(s)
- Xiaojuan Han
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yuanjing Li
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institute-Stockholm University, Stockholm, Sweden
- The Swedish School of Sport and Health Science, GIH, Stockholm, Sweden
| | - Jiafeng Wang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Xinyu Liu
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yu Zhang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Qiwei Dong
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Yiming Song
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Lin Cong
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Qinghua Zhang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Shi Tang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Lin Song
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Tingting Hou
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yongxiang Wang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institute-Stockholm University, Stockholm, Sweden
- Institute of Brain Science and Brain-Inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
- Institute of Brain Science and Brain-Inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Chengxuan Qiu
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institute-Stockholm University, Stockholm, Sweden
- Institute of Brain Science and Brain-Inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
5
|
Yang Q, Yu T. Study on the age-period-cohort effects of cognitive abilities among older Chinese adults based on the cognitive reserve hypothesis. BMC Geriatr 2024; 24:992. [PMID: 39633278 PMCID: PMC11616311 DOI: 10.1186/s12877-024-05576-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Cognitive abilities serves as a critical indicator of healthy aging. As China progresses into a stage of advanced population aging, there has been a significant increase in the number of elderly individuals experiencing age-related cognitive decline. Despite this demographic shift, there is a paucity of longitudinal research examining cognitive abilities among older Chinese adults over extended time periods. This study aims to investigate changes in cognitive abilities and explore group differences among older Chinese adults aged 65 to 110 years, employing a multidimensional temporal approach that encompasses age, period, and birth cohort effects. METHODS This study utilizes data from eight waves of the Chinese Longitudinal Healthy Longevity Survey (CLHLS), spanning from 1998 to 2018. The dataset comprises 94,116 observations from 36,157 unique participants. Cognitive abilities are assessed using Mini-Mental State Examination (MMSE) scores as a proxy measure. To address the issue of perfect collinearity in the temporal dimension, the study employs the Hierarchical Age-Period-Cohort Cross-Classified Random Effects Model (HAPC-CCREM). This model allows for the examination of age effects, period effects, and cohort effects on cognitive abilities among older Chinese adults. In the model specification, age is treated as a fixed effect, while period and birth cohort are incorporated as random effects. Drawing upon the cognitive reserve hypothesis, the study investigates significant factors influencing cognitive abilities in this population. RESULTS At the fixed-effect level, demographic factors, health behaviors, self-rated health, subjective well-being, and childhood adversity significantly influence cognitive abilities among older Chinese adults. The age effects are significant, with cognitive abilities exhibiting an inverted U-shaped curve across the lifespan. At the random-effect level, period effects are significant, revealing a gradual annual increase in overall cognitive levels among older Chinese adults since 2008. Cohort effects are also significant, demonstrating an increasing trend in overall cognitive levels for the earlier-born cohorts in the first six groups. Conversely, later-born cohorts in the latter five groups show a declining trend in overall cognitive levels. Notably, period effects significantly enhance cohort effects. CONCLUSIONS The cognitive reserve hypothesis supports the significance of the majority of identified influencing factors. Cognitive abilities demonstrate an accelerating decline with increasing age, following an evolutionary trajectory consistent with physiological principles among older Chinese adults. Since 2008, cognitive abilities have shown a monotonic increasing trend annually, further validating the Flynn effect in this population. The cognitive abilities of the six earlier-born cohorts exhibit an increasing trend, supporting the compression of morbidity hypothesis. Conversely, the cognitive abilities of the five later-born cohorts show a declining trend, supporting the expansion of morbidity hypothesis. These findings collectively contribute to our understanding of cognitive aging patterns and their underlying mechanisms among older Chinese adults.
Collapse
Affiliation(s)
- Qian Yang
- Department of Neurology, The First Affiliated Hospital of Wannan Medical College, NO.2 Zheshan West Road,Jinghu District, Wuhu, Anhui, 241001, China
| | - Tong Yu
- School of Humanities and Management, Wannan Medical College, NO.22 Wenchang West Road,Yijiang District, Wuhu, Anhui, 241002, China.
| |
Collapse
|
6
|
Seifar F, Fox EJ, Shantaraman A, Liu Y, Dammer EB, Modeste E, Duong DM, Yin L, Trautwig AN, Guo Q, Xu K, Ping L, Reddy JS, Allen M, Quicksall Z, Heath L, Scanlan J, Wang E, Wang M, Linden AV, Poehlman W, Chen X, Baheti S, Ho C, Nguyen T, Yepez G, Mitchell AO, Oatman SR, Wang X, Carrasquillo MM, Runnels A, Beach T, Serrano GE, Dickson DW, Lee EB, Golde TE, Prokop S, Barnes LL, Zhang B, Haroutunian V, Gearing M, Lah JJ, De Jager P, Bennett DA, Greenwood A, Ertekin‐Taner N, Levey AI, Wingo A, Wingo T, Seyfried NT. Large-scale deep proteomic analysis in Alzheimer's disease brain regions across race and ethnicity. Alzheimers Dement 2024; 20:8878-8897. [PMID: 39535480 PMCID: PMC11667503 DOI: 10.1002/alz.14360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/09/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most prevalent neurodegenerative disease, yet our comprehension predominantly relies on studies within non-Hispanic White (NHW) populations. Here we provide an extensive survey of the proteomic landscape of AD across diverse racial/ethnic groups. METHODS Two cortical regions, from multiple centers, were harmonized by uniform neuropathological diagnosis. Among 998 unique donors, 273 donors self-identified as African American, 229 as Latino American, and 434 as NHW. RESULTS While amyloid precursor protein and the microtubule-associated protein tau demonstrated higher abundance in AD brains, no significant race-related differences were observed. Further proteome-wide and focused analyses (specific amyloid beta [Aβ] species and the tau domains) supported the absence of racial differences in these AD pathologies within the brain proteome. DISCUSSION Our findings indicate that the racial differences in AD risk and clinical presentation are not underpinned by dramatically divergent patterns in the brain proteome, suggesting that other determinants account for these clinical disparities. HIGHLIGHTS We present a large-scale proteome (∼10,000 proteins) of DLPFC (998) and STG (244) across AD cases. About 50% of samples were from racially and ethnically diverse brain donors. Key AD proteins (amyloid and tau) correlated with CERAD and Braak stages. No significant race-related differences in amyloid and tau protein levels were observed in AD brains. AD-associated protein changes showed a strong correlation between the brain proteomes of African American and White individuals. This dataset advances understanding of ethnoracial-specific AD pathways and potential therapies.
Collapse
|
7
|
Seto M, Hohman TJ, Mormino EC, Papp KV, Amariglio RE, Rentz DM, Johnson KA, Schultz AP, Sperling RA, Buckley RF, Yang HS. Parental History of Memory Impairment and β-Amyloid in Cognitively Unimpaired Older Adults. JAMA Neurol 2024; 81:798-804. [PMID: 38884955 PMCID: PMC11184498 DOI: 10.1001/jamaneurol.2024.1763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/19/2024] [Indexed: 06/18/2024]
Abstract
Importance Studies have suggested that maternal history of late-onset Alzheimer disease, but not paternal, predisposes individuals to higher brain β-amyloid (Aβ) burden, reduced brain metabolism, and lower gray matter volumes. Objective To characterize maternal vs paternal history of memory impairment in terms of brain Aβ-positron emission tomography (Aβ-PET) and baseline cognition among a large sample of cognitively unimpaired older adults. Design, Setting, and Participants This cross-sectional study leveraged data from 4413 individuals who were screened for the Anti-Amyloid Treatment in Asymptomatic Alzheimer (A4) study, a randomized clinical trial conducted across 67 sites in the US, Australia, Canada, and Japan aimed at Alzheimer disease prevention. Data were collected between April 2014 and December 2017 and analyzed from December 2022 to June 2023. Participants were cognitively unimpaired adults (Clinical Dementia Rating = 0 and/or Mini-Mental State Examination score ≥25) between the ages of 65 and 85 years who underwent PET imaging to assess cortical Aβ levels for trial eligibility. A total of 4492 participants were screened, and 79 missing data were excluded. Main Outcomes and Measures Demographic characteristics (eg, age, sex, education), apolipoprotein E genotyping, participant-reported parental history of memory impairment and parental age at symptom onset were collected as variables. Parental history was assessed in terms of continuous neocortical 18F-florbetapir Aβ-PET and the Preclinical Alzheimer Cognitive Composite. Results Of 4413 individuals (mean [SD] age, 71.27 [4.66] years, 2617 women [59.3%]), mean Aβ-PET was elevated in individuals with history of memory impairment in both parents (n = 455; mean [SD] standardized uptake value ratio [SUVR] = 1.12 [0.19]; Wilcoxon P = 1.1 × 10-5) and in those with only maternal history (n = 1772; mean [SD] SUVR = 1.10 [0.19]; Wilcoxon P = 2.70 × 10-5) compared with those with only paternal history (n = 632; mean [SD] SUVR = 1.08 [0.18]; Wilcoxon P = 1.1 × 10-5) or no family history (n = 1554; mean [SD] SUVR = 1.08 [0.19]; Wilcoxon P = 1.1 × 10-5). Paternal history of early-onset memory impairment (age <65 years) but not late-onset (age ≥65 years) was associated with elevated participant Aβ-PET (mean [SD] SUVR = 1.19 [0.21]; P = 3.00 × 10-6) in comparison with no paternal history (mean [SD] SUVR = 1.09 [0.19]) whereas maternal history was associated with elevated Aβ in both early-onset and late-onset groups. There was no association with cognition. Conclusions and Relevance In this study, maternal history (at any age) and paternal history of early-onset memory impairment were associated with Aβ burden among asymptomatic older individuals. Sex-specific parental history may help inform clinicians on likelihood of Aβ burden in offspring and help identify high-risk individuals at the earliest stages of disease for prevention.
Collapse
Affiliation(s)
- Mabel Seto
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Elizabeth C. Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California
| | - Kathryn V. Papp
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Rebecca E. Amariglio
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Dorene M. Rentz
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Keith A. Johnson
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Radiology, Massachusetts General Hospital, Boston
| | - Aaron P. Schultz
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown
| | - Reisa A. Sperling
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Rachel F. Buckley
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Hyun-Sik Yang
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
8
|
Seifar F, Fox EJ, Shantaraman A, Liu Y, Dammer EB, Modeste E, Duong DM, Yin L, Trautwig AN, Guo Q, Xu K, Ping L, Reddy JS, Allen M, Quicksall Z, Heath L, Scanlan J, Wang E, Wang M, Linden AV, Poehlman W, Chen X, Baheti S, Ho C, Nguyen T, Yepez G, Mitchell AO, Oatman SR, Wang X, Carrasquillo MM, Runnels A, Beach T, Serrano GE, Dickson DW, Lee EB, Golde TE, Prokop S, Barnes LL, Zhang B, Haroutunian V, Gearing M, Lah JJ, Jager PD, Bennett DA, Greenwood A, Ertekin-Taner N, Levey AI, Wingo A, Wingo T, Seyfried NT. Large-scale Deep Proteomic Analysis in Alzheimer's Disease Brain Regions Across Race and Ethnicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590547. [PMID: 38712030 PMCID: PMC11071432 DOI: 10.1101/2024.04.22.590547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Introduction Alzheimer's disease (AD) is the most prevalent neurodegenerative disease, yet our comprehension predominantly relies on studies within the non-Hispanic White (NHW) population. Here we aimed to provide comprehensive insights into the proteomic landscape of AD across diverse racial and ethnic groups. Methods Dorsolateral prefrontal cortex (DLPFC) and superior temporal gyrus (STG) brain tissues were donated from multiple centers (Mayo Clinic, Emory University, Rush University, Mt. Sinai School of Medicine) and were harmonized through neuropathological evaluation, specifically adhering to the Braak staging and CERAD criteria. Among 1105 DLPFC tissue samples (998 unique individuals), 333 were from African American donors, 223 from Latino Americans, 529 from NHW donors, and the rest were from a mixed or unknown racial background. Among 280 STG tissue samples (244 unique individuals), 86 were African American, 76 Latino American, 116 NHW and the rest were mixed or unknown ethnicity. All tissues were uniformly homogenized and analyzed by tandem mass tag mass spectrometry (TMT-MS). Results As a Quality control (QC) measure, proteins with more than 50% missing values were removed and iterative principal component analysis was conducted to remove outliers within brain regions. After QC, 9,180 and 9,734 proteins remained in the DLPC and STG proteome, respectively, of which approximately 9,000 proteins were shared between regions. Protein levels of microtubule-associated protein tau (MAPT) and amyloid-precursor protein (APP) demonstrated AD-related elevations in DLPFC tissues with a strong association with CERAD and Braak across racial groups. APOE4 protein levels in brain were highly concordant with APOE genotype of the individuals. Discussion This comprehensive region resolved large-scale proteomic dataset provides a resource for the understanding of ethnoracial-specific protein differences in AD brain.
Collapse
Affiliation(s)
| | - Edward J Fox
- Emory University School of Medicine, Atlanta, GA USA
| | | | - Yue Liu
- Emory University School of Medicine, Atlanta, GA USA
| | - Eric B Dammer
- Emory University School of Medicine, Atlanta, GA USA
| | - Erica Modeste
- Emory University School of Medicine, Atlanta, GA USA
| | - Duc M Duong
- Emory University School of Medicine, Atlanta, GA USA
| | - Luming Yin
- Emory University School of Medicine, Atlanta, GA USA
| | | | - Qi Guo
- Emory University School of Medicine, Atlanta, GA USA
| | - Kaiming Xu
- Emory University School of Medicine, Atlanta, GA USA
| | - Lingyan Ping
- Emory University School of Medicine, Atlanta, GA USA
| | - Joseph S Reddy
- Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL USA
| | - Mariet Allen
- Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL USA
| | - Zachary Quicksall
- Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL USA
| | | | | | - Erming Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | | | | | - Xianfeng Chen
- Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL USA
| | - Saurabh Baheti
- Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL USA
| | - Charlotte Ho
- Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL USA
| | - Thuy Nguyen
- Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL USA
| | - Geovanna Yepez
- Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL USA
| | | | | | - Xue Wang
- Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL USA
| | | | | | - Thomas Beach
- Banner Sun Health Research Institute, Sun City, AR USA
| | | | - Dennis W Dickson
- Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL USA
| | - Edward B Lee
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelpha, PA, USA
| | - Todd E Golde
- Emory University School of Medicine, Atlanta, GA USA
| | | | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Varham Haroutunian
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Marla Gearing
- Emory University School of Medicine, Atlanta, GA USA
| | - James J Lah
- Emory University School of Medicine, Atlanta, GA USA
| | | | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL USA
| | | | - Nilüfer Ertekin-Taner
- Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL USA
- Mayo Clinic Florida, Department of Neurology, Jacksonville, FL USA
| | - Allan I Levey
- Emory University School of Medicine, Atlanta, GA USA
| | - Aliza Wingo
- Emory University School of Medicine, Atlanta, GA USA
| | - Thomas Wingo
- Emory University School of Medicine, Atlanta, GA USA
| | | |
Collapse
|
9
|
Castillo-Ordoñez WO, Cajas-Salazar N, Velasco-Reyes MA. Genetic and epigenetic targets of natural dietary compounds as anti-Alzheimer's agents. Neural Regen Res 2024; 19:846-854. [PMID: 37843220 PMCID: PMC10664119 DOI: 10.4103/1673-5374.382232] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/15/2023] [Accepted: 07/18/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder and the most common cause of dementia that principally affects older adults. Pathogenic factors, such as oxidative stress, an increase in acetylcholinesterase activity, mitochondrial dysfunction, genotoxicity, and neuroinflammation are present in this syndrome, which leads to neurodegeneration. Neurodegenerative pathologies such as Alzheimer's disease are considered late-onset diseases caused by the complex combination of genetic, epigenetic, and environmental factors. There are two main types of Alzheimer's disease, known as familial Alzheimer's disease (onset < 65 years) and late-onset or sporadic Alzheimer's disease (onset ≥ 65 years). Patients with familial Alzheimer's disease inherit the disease due to rare mutations on the amyloid precursor protein (APP), presenilin 1 and 2 (PSEN1 and PSEN2) genes in an autosomal-dominantly fashion with closely 100% penetrance. In contrast, a different picture seems to emerge for sporadic Alzheimer's disease, which exhibits numerous non-Mendelian anomalies suggesting an epigenetic component in its etiology. Importantly, the fundamental pathophysiological mechanisms driving Alzheimer's disease are interfaced with epigenetic dysregulation. However, the dynamic nature of epigenetics seems to open up new avenues and hope in regenerative neurogenesis to improve brain repair in Alzheimer's disease or following injury or stroke in humans. In recent years, there has been an increase in interest in using natural products for the treatment of neurodegenerative illnesses such as Alzheimer's disease. Through epigenetic mechanisms, such as DNA methylation, non-coding RNAs, histone modification, and chromatin conformation regulation, natural compounds appear to exert neuroprotective effects. While we do not purport to cover every in this work, we do attempt to illustrate how various phytochemical compounds regulate the epigenetic effects of a few Alzheimer's disease-related genes.
Collapse
Affiliation(s)
- Willian Orlando Castillo-Ordoñez
- Facultad de Ciencias Naturales-Exactas y de la Educación, Departamento de Biología. Universidad del Cauca, Popayán-Cauca, Colombia
- Departamento de Estudios Psicológicos, Universidad Icesi, Cali, Colombia
| | - Nohelia Cajas-Salazar
- Facultad de Ciencias Naturales-Exactas y de la Educación, Departamento de Biología. Universidad del Cauca, Popayán-Cauca, Colombia
| | - Mayra Alejandra Velasco-Reyes
- Facultad de Ciencias Naturales-Exactas y de la Educación, Departamento de Biología. Universidad del Cauca, Popayán-Cauca, Colombia
| |
Collapse
|
10
|
Weaver DF. Thirty Risk Factors for Alzheimer's Disease Unified by a Common Neuroimmune-Neuroinflammation Mechanism. Brain Sci 2023; 14:41. [PMID: 38248256 PMCID: PMC10813027 DOI: 10.3390/brainsci14010041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/27/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024] Open
Abstract
One of the major obstacles confronting the formulation of a mechanistic understanding for Alzheimer's disease (AD) is its immense complexity-a complexity that traverses the full structural and phenomenological spectrum, including molecular, macromolecular, cellular, neurological and behavioural processes. This complexity is reflected by the equally complex diversity of risk factors associated with AD. However, more than merely mirroring disease complexity, risk factors also provide fundamental insights into the aetiology and pathogenesis of AD as a neurodegenerative disorder since they are central to disease initiation and subsequent propagation. Based on a systematic literature assessment, this review identified 30 risk factors for AD and then extended the analysis to further identify neuroinflammation as a unifying mechanism present in all 30 risk factors. Although other mechanisms (e.g., vasculopathy, proteopathy) were present in multiple risk factors, dysfunction of the neuroimmune-neuroinflammation axis was uniquely central to all 30 identified risk factors. Though the nature of the neuroinflammatory involvement varied, the activation of microglia and the release of pro-inflammatory cytokines were a common pathway shared by all risk factors. This observation provides further evidence for the importance of immunopathic mechanisms in the aetiopathogenesis of AD.
Collapse
Affiliation(s)
- Donald F Weaver
- Krembil Research Institute, University Health Network, Departments of Medicine, Chemistry, Pharmaceutical Sciences, University of Toronto, Toronto, ON M5T 0S8, Canada
| |
Collapse
|
11
|
Ehret F, Pelz MS, Senko AN, Soto KEG, Liu H, Kempermann G. Presymptomatic Reduction of Individuality in the App NL-F Knockin Model of Alzheimer's Disease. Biol Psychiatry 2023; 94:721-731. [PMID: 37076091 DOI: 10.1016/j.biopsych.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND One-third of the risk for Alzheimer's disease is explained by environment and lifestyle, but Alzheimer's disease pathology might also affect lifestyle and thereby impair the individual potential for health behavior and prevention. METHODS We examined in mice how the AppNL-F/NL-F (NL-F) knockin mutation affects the presymptomatic response to environmental enrichment (ENR) as an experimental paradigm addressing nongenetic factors. We assessed the emergence of interindividual phenotypic variation under the condition that both the genetic background and the shared environment were held constant, thereby isolating the contribution of individual behavior (nonshared environment). RESULTS After 4 months of ENR, the mean and variability of plasma ApoE were increased in NL-F mice, suggesting a presymptomatic variation in pathogenic processes. Roaming entropy as a measure of behavioral activity was continuously assessed with radiofrequency identification (RFID) technology and revealed reduced habituation and variance in NL-F mice compared with control animals, which do not carry a Beyreuther/Iberian mutation. Intraindividual variation decreased, while behavioral stability was reduced in NL-F mice. Seven months after discontinuation of ENR, we found no difference in plaque size and number, but ENR increased variance in hippocampal plaque counts in NL-F mice. A reactive increase in adult hippocampal neurogenesis in NL-F mice, known from other models, was normalized by ENR. CONCLUSIONS Our data suggest that while NL-F has early effects on individual behavioral patterns in response to ENR, there are lasting effects on cellular plasticity even after the discontinuation of ENR. Hence, early behavior matters for maintaining individual behavioral trajectories and brain plasticity even under maximally constrained conditions.
Collapse
Affiliation(s)
- Fanny Ehret
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany; Institute of Anatomy, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Meike S Pelz
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany
| | - Anna N Senko
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany
| | - Karla E G Soto
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany
| | - Hang Liu
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany
| | - Gerd Kempermann
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany.
| |
Collapse
|
12
|
Lepinay E, Cicchetti F. Tau: a biomarker of Huntington's disease. Mol Psychiatry 2023; 28:4070-4083. [PMID: 37749233 DOI: 10.1038/s41380-023-02230-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 09/27/2023]
Abstract
Developing effective treatments for patients with Huntington's disease (HD)-a neurodegenerative disorder characterized by severe cognitive, motor and psychiatric impairments-is proving extremely challenging. While the monogenic nature of this condition enables to identify individuals at risk, robust biomarkers would still be extremely valuable to help diagnose disease onset and progression, and especially to confirm treatment efficacy. If measurements of cerebrospinal fluid neurofilament levels, for example, have demonstrated use in recent clinical trials, other proteins may prove equal, if not greater, relevance as biomarkers. In fact, proteins such as tau could specifically be used to detect/predict cognitive affectations. We have herein reviewed the literature pertaining to the association between tau levels and cognitive states, zooming in on Alzheimer's disease, Parkinson's disease and traumatic brain injury in which imaging, cerebrospinal fluid, and blood samples have been interrogated or used to unveil a strong association between tau and cognition. Collectively, these areas of research have accrued compelling evidence to suggest tau-related measurements as both diagnostic and prognostic tools for clinical practice. The abundance of information retrieved in this niche of study has laid the groundwork for further understanding whether tau-related biomarkers may be applied to HD and guide future investigations to better understand and treat this disease.
Collapse
Affiliation(s)
- Eva Lepinay
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada.
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada.
| |
Collapse
|
13
|
Giaquinto F, Tosi G, Abbatantuono C, Pepe I, Iaia M, Macchitella L, Rizzi E, De Caro MF, Romano D, Taurisano P, Angelelli P. The indirect effect of cognitive reserve on the relationship between age and cognition in pathological ageing: A cross-sectional retrospective study in an unselected and consecutively enrolled sample. J Neuropsychol 2023; 17:477-490. [PMID: 37184066 DOI: 10.1111/jnp.12323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
Cognitive reserve (CR) allows individuals to maintain cognitive functionality even in the presence of pathologies. The compensation hypothesis suggests that CR plays an indirect role between age and cognitive decline, contrasting the negative effect of ageing on cognition. We test this hypothesis in an unselected and consecutively enrolled sample of memory clinic attendees (n = 134) who completed the CR Index questionnaire and three neuropsychological tests assessing global cognition (MMSE, FAB, CDT). Participants were divided into two groups based on standard diagnostic criteria (DSM-5): those who were cognitively impaired (n = 92) and those who were preserved (n = 42). A principal component analysis was used to extract a composite measure of global cognitive functioning from the three neuropsychological tests, and mediation analysis was used to examine the relationship between CR, age and global cognitive functioning in the two groups. Results revealed that: (i) age had a significant direct negative effect on the global cognitive score in both groups; (ii) the three socio-behavioural proxies of CR together suppress the direct negative relationship between age and global cognitive score in cognitively impaired patients but not in cognitively preserved participants. This study confirms the association between CR, age and cognition and allows us to validate its role in a population with cognitive impairment and extend findings to a low-to-middle educated population. These results hold important implications for public health and wellness promotion, emphasising the beneficial role of maintaining healthy and active physical, cognitive and social lifestyles.
Collapse
Affiliation(s)
- Francesco Giaquinto
- Laboratory of Applied Psychology and Intervention, Department of Human and Social Sciences, University of Salento, Lecce, Italy
| | - Giorgia Tosi
- Laboratory of Applied Psychology and Intervention, Department of Human and Social Sciences, University of Salento, Lecce, Italy
- Psychology Department, University of Milano-Bicocca, Milan, Italy
| | - Chiara Abbatantuono
- Department of Translational Biomedicine and Neuroscience "DiBraiN", University of Bari "Aldo Moro", Bari, Italy
| | - Ilaria Pepe
- Department of Translational Biomedicine and Neuroscience "DiBraiN", University of Bari "Aldo Moro", Bari, Italy
| | - Marika Iaia
- Laboratory of Applied Psychology and Intervention, Department of Human and Social Sciences, University of Salento, Lecce, Italy
| | - Luigi Macchitella
- Laboratory of Applied Psychology and Intervention, Department of Human and Social Sciences, University of Salento, Lecce, Italy
- Scientific Institute I.R.C.C.S. "E. Medea"- Unit for Severe Disabilities in Developmental Age and Young Adults (Developmental Neurology and Neurorehabilitation), Brindisi, Italy
| | - Ezia Rizzi
- Laboratory of Applied Psychology and Intervention, Department of Human and Social Sciences, University of Salento, Lecce, Italy
- Psychology Department, University of Milano-Bicocca, Milan, Italy
| | - Maria Fara De Caro
- Department of Translational Biomedicine and Neuroscience "DiBraiN", University of Bari "Aldo Moro", Bari, Italy
| | - Daniele Romano
- Psychology Department, University of Milano-Bicocca, Milan, Italy
| | - Paolo Taurisano
- Department of Translational Biomedicine and Neuroscience "DiBraiN", University of Bari "Aldo Moro", Bari, Italy
| | - Paola Angelelli
- Laboratory of Applied Psychology and Intervention, Department of Human and Social Sciences, University of Salento, Lecce, Italy
| |
Collapse
|
14
|
Weigand AJ, Ortiz G, Walker KS, Galasko DR, Bondi MW, Thomas KR. APOE differentially moderates cerebrospinal fluid and plasma phosphorylated tau181 associations with multi-domain cognition. Neurobiol Aging 2023; 125:1-8. [PMID: 36780762 DOI: 10.1016/j.neurobiolaging.2022.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 01/19/2023]
Abstract
Biofluid markers of phosphorylated tau181 (p-tau181) are increasingly popular for the detection of early Alzheimer's pathologic changes. However, the differential dynamics of cerebrospinal fluid (CSF) and plasma p-tau181 remain under investigation. We studied 727 participants from the Alzheimer's Disease Neuroimaging Initiative with plasma and CSF p-tau181 data, apolipoprotein (APOE) ε4 carrier status, amyloid positron emission tomography (PET) imaging, and neuropsychological data. Higher levels of plasma and CSF p-tau181 were observed among APOE ε4 carriers. CSF and plasma p-tau181 were significantly associated with memory, and this effect was greater in APOE ε4 carriers. However, whereas CSF p-tau181 was not significantly associated with language or attention/executive function among ε4 carriers or non-carriers, APOE ε4 status moderated the association of plasma p-tau181 with both language and attention/executive function. These findings lend support to the notion that p-tau181 biofluid markers are useful in measuring AD pathologic changes but also suggest that CSF and plasma p-tau181 have unique properties and dynamics that should be considered when using these markers in research and clinical practice.
Collapse
Affiliation(s)
- Alexandra J Weigand
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Gema Ortiz
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Kayla S Walker
- San Diego State University, Department of Psychology, San Diego, CA, USA
| | - Douglas R Galasko
- VA San Diego Healthcare System, San Diego, CA, USA; University of California San Diego, Department of Neurosciences, La Jolla, CA, USA
| | - Mark W Bondi
- VA San Diego Healthcare System, San Diego, CA, USA; University of California San Diego, Department of Psychiatry, La Jolla, CA, USA
| | - Kelsey R Thomas
- VA San Diego Healthcare System, San Diego, CA, USA; University of California San Diego, Department of Psychiatry, La Jolla, CA, USA.
| | | |
Collapse
|
15
|
Chen R, Yi Y, Xiao W, Zhong B, Zhang L, Zeng Y. Urinary protein biomarkers based on LC-MS/MS analysis to discriminate vascular dementia from Alzheimer's disease in Han Chinese population. Front Aging Neurosci 2023; 15:1070854. [PMID: 36761180 PMCID: PMC9905227 DOI: 10.3389/fnagi.2023.1070854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023] Open
Abstract
Objective This study aimed to identify the potential urine biomarkers of vascular dementia (VD) and unravel the disease-associated mechanisms by applying Liquid chromatography tandem-mass spectrometry (LC-MS/MS). Methods LC-MS/MS proteomic analysis was applied to urine samples from 3 groups, including 14 patients with VD, 9 patients with AD, and 21 normal controls (NC). By searching the MS data by Proteome Discoverer software, analyzing the protein abundances qualitatively and quantitatively, comparing between groups, combining bioinformatics analysis using Gene Ontology (GO) and pathway crosstalk analysis using Kyoto Encyclopedia of Genes and Genomes (KEGG), and literature searching, the differentially expressed proteins (DEPs) of VD can be comprehensively determined at last and were further quantified by receiver operating characteristic (ROC) curve methods. Results The proteomic findings showed quantitative changes in patients with VD compared to patients with NC and AD groups; among 4,699 identified urine proteins, 939 and 1,147 proteins displayed quantitative changes unique to VD vs. NC and AD, respectively, including 484 overlapped common DEPs. Then, 10 unique proteins named in KEGG database (including PLOD3, SDCBP, SRC, GPRC5B, TSG101/STP22/VPS23, THY1/CD90, PLCD, CDH16, NARS/asnS, AGRN) were confirmed by a ROC curve method. Conclusion Our results suggested that urine proteins enable detection of VD from AD and VC, which may provide an opportunity for intervention.
Collapse
Affiliation(s)
- Ruijuan Chen
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China,Department of Emergency, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan, China
| | - Yuanjing Yi
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenbiao Xiao
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bowen Zhong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Le Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Zeng
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China,*Correspondence: Yi Zeng,
| |
Collapse
|
16
|
Amofa-Ho PA, Stickel AM, Chen R, Kobayashi LC, Glymour MM, Eng CW. The Mediating Roles of Neurobiomarkers in the Relationship Between Education and Late-Life Cognition. J Alzheimers Dis 2023; 95:1405-1416. [PMID: 37694365 PMCID: PMC10578223 DOI: 10.3233/jad-230244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND The mediating roles of neuropathologies and neurovascular damage in the relationship between early-life education and later-life cognitive function are unknown. OBJECTIVE To examine whether Alzheimer's and neurovascular biomarkers mediate the relationships between education and cognitive functions. METHODS Data were from 537 adults aged 55-94 in the Alzheimer's Disease Neuroimaging Initiative 3. We tested whether the relationships between education (continuous, years) and cognitive function (memory, executive functioning, and language composites) were mediated by neuroimaging biomarkers (hippocampal volumes, cortical gray matter volumes, meta-temporal tau PET standard uptake value ratio, and white matter hyperintensity volumes). Models were adjusted for age, race, sex/gender, cardiovascular history, body mass index, depression, and Apolipoprotein E-ɛ4 status. RESULTS Hippocampal volumes and white matter hyperintensities partially mediated the relationships between education and cognitive function across all domains (6.43% to 15.72% mediated). The direct effects of education on each cognitive domain were strong and statistically significant. CONCLUSIONS Commonly measured neurobiomarkers only partially mediate the relationships between education and multi-domain cognitive function.
Collapse
Affiliation(s)
- Priscilla A. Amofa-Ho
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
| | - Ariana M. Stickel
- Department of Psychology, San Diego State University, San Diego, CA, USA
| | - Ruijia Chen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Lindsay C. Kobayashi
- Center for Social Epidemiology and Population Health, Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - M. Maria Glymour
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Chloe W. Eng
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
- Department of Epidemiology, Stanford University, Palo Alto, CA, USA
| | | |
Collapse
|
17
|
Wolff L, Quan Y, Perry G, Forde Thompson W. Music Engagement as a Source of Cognitive Reserve. Am J Alzheimers Dis Other Demen 2023; 38:15333175231214833. [PMID: 37993973 PMCID: PMC10666690 DOI: 10.1177/15333175231214833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Music engagement is a ubiquitous activity that is thought to have cognitive benefits for the rapidly aging population. In the absence of robust treatment approaches for many age-related and neuropathological health issues, interest has emerged surrounding lifestyle-enriching activities, like exercise and music engagement, to build cognitive reserve across the lifespan and preserve neurocognitive function in older adults. The present review evaluates evidence of neurocognitive preservation arising from lifelong music engagement with respect to the cognitive reserve hypothesis. We collated a body of neuroimaging, behavioral and epidemiological evidence to adjudicate the benefits of music engagement for cognitive reserve. The findings suggest that music engagement should be considered in tandem with other well-established cognitive reserve proxies as a contributor to differential clinical outcomes in older populations at risk of age-related and neuropathological cognitive decline.
Collapse
Affiliation(s)
- Lee Wolff
- Department of Psychology, Bond University, Robina, QLD, Australia
| | - Yixue Quan
- Department of Psychology, Macquarie University, Sydney, NSW, Australia
| | - Gemma Perry
- Department of Psychology, Bond University, Robina, QLD, Australia
| | - William Forde Thompson
- Department of Psychology, Bond University, Robina, QLD, Australia
- Department of Psychology, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
18
|
Sidenkova A, Calabrese V, Tomasello M, Fritsch T. Subjective cognitive decline and cerebral-cognitive reserve in late age. TRANSLATIONAL MEDICINE OF AGING 2023; 7:137-147. [DOI: 10.1016/j.tma.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2024] Open
|
19
|
Grasset L, Proust-Lima C, Mangin JF, Habert MO, Dubois B, Paquet C, Hanon O, Gabelle A, Ceccaldi M, Annweiler C, David R, Jonveaux T, Belin C, Julian A, Rouch-Leroyer I, Pariente J, Locatelli M, Chupin M, Chêne G, Dufouil C. Explaining the association between social and lifestyle factors and cognitive functions: a pathway analysis in the Memento cohort. Alzheimers Res Ther 2022; 14:68. [PMID: 35585559 PMCID: PMC9115948 DOI: 10.1186/s13195-022-01013-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/27/2022] [Indexed: 11/10/2022]
Abstract
Abstract
Background
This work aimed to investigate the potential pathways involved in the association between social and lifestyle factors, biomarkers of Alzheimer’s disease and related dementia (ADRD), and cognition.
Methods
The authors studied 2323 participants from the Memento study, a French nationwide clinical cohort. Social and lifestyle factors were education level, current household incomes, physical activity, leisure activities, and social network from which two continuous latent variables were computed: an early to midlife (EML) and a latelife (LL) indicator. Brain magnetic resonance imaging (MRI), lumbar puncture, and amyloid-positron emission tomography (PET) were used to define three latent variables: neurodegeneration, small vessel disease (SVD), and AD pathology. Cognitive function was defined as the underlying factor of a latent variable with four cognitive tests. Structural equation models were used to evaluate cross-sectional pathways between social and lifestyle factors and cognition.
Results
Participants’ mean age was 70.9 years old, 62% were women, 28% were apolipoprotein-ε4 carriers, and 59% had a Clinical Dementia Rating (CDR) score of 0.5. Higher early to midlife social indicator was only directly associated with better cognitive function (direct β = 0.364 (0.322; 0.405), with no indirect pathway through ADRD biomarkers (total β = 0.392 (0.351; 0.429)). In addition to a direct effect on cognition (direct β = 0.076 (0.033; 0.118)), the association between latelife lifestyle indicator and cognition was also mostly mediated by an indirect effect through lower neurodegeneration (indirect β = 0.066 (0.042; 0.090) and direct β = − 0.116 (− 0.153; − 0.079)), but not through AD pathology nor SVD.
Conclusions
Early to midlife social factors are directly associated with higher cognitive functions. Latelife lifestyle factors may help preserve cognitive functions through lower neurodegeneration.
Collapse
|
20
|
Cognitive performance protects against Alzheimer's disease independently of educational attainment and intelligence. Mol Psychiatry 2022; 27:4297-4306. [PMID: 35840796 DOI: 10.1038/s41380-022-01695-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/21/2022] [Accepted: 06/30/2022] [Indexed: 02/07/2023]
Abstract
Mendelian-randomization (MR) studies using large-scale genome-wide association studies (GWAS) have identified causal association between educational attainment and Alzheimer's disease (AD). However, the underlying mechanisms are still required to be explored. Here, we conduct univariable and multivariable MR analyses using large-scale educational attainment, cognitive performance, intelligence and AD GWAS datasets. In stage 1, we found significant causal effects of educational attainment on cognitive performance (beta = 0.907, 95% confidence interval (CI): 0.884-0.930, P < 1.145E-299), and vice versa (beta = 0.571, 95% CI: 0.557-0.585, P < 1.145E-299). In stage 2, we found that both increase in educational attainment (odds ratio (OR) = 0.72, 95% CI: 0.66-0.78, P = 1.39E-14) and cognitive performance (OR = 0.69, 95% CI: 0.64-0.75, P = 1.78E-20) could reduce the risk of AD. In stage 3, we found that educational attainment may protect against AD dependently of cognitive performance (OR = 1.07, 95% CI: 0.90-1.28, P = 4.48E-01), and cognitive performance may protect against AD independently of educational attainment (OR = 0.69, 95% CI: 0.53-0.89, P = 5.00E-03). In stage 4, we found significant causal effects of cognitive performance on intelligence (beta = 0.907, 95% CI: 0.877-0.938, P < 1.145E-299), and vice versa (beta = 0.957, 95% CI: 0.937-0.978, P < 1.145E-299). In stage 5, we identified that cognitive performance may protect against AD independently of intelligence (OR = 0.74, 95% CI: 0.61-0.90, P = 2.00E-03), and intelligence may protect against AD dependently of cognitive performance (OR = 1.17, 95% CI: 0.40-3.43, P = 4.48E-01). Collectively, our univariable and multivariable MR analyses highlight the protective role of cognitive performance in AD independently of educational attainment and intelligence. In addition to the intelligence, we extend the mechanisms underlying the associations of educational attainment with AD.
Collapse
|
21
|
Amelianchik A, Sweetland-Martin L, Norris EH. The effect of dietary fat consumption on Alzheimer's disease pathogenesis in mouse models. Transl Psychiatry 2022; 12:293. [PMID: 35869065 PMCID: PMC9307654 DOI: 10.1038/s41398-022-02067-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) is a fatal cognitive disorder with proteinaceous brain deposits, neuroinflammation, cerebrovascular dysfunction, and extensive neuronal loss over time. AD is a multifactorial disease, and lifestyle factors, including diet, are likely associated with the development of AD pathology. Since obesity and diabetes are recognized as risk factors for AD, it might be predicted that a high-fat diet (HFD) would worsen AD pathology. However, modeling HFD-induced obesity in AD animal models has yielded inconclusive results. Some studies report a deleterious effect of HFD on Aβ accumulation, neuroinflammation, and cognitive function, while others report that HFD worsens memory without affecting AD brain pathology. Moreover, several studies report no major effect of HFD on AD-related phenotypes in mice, while other studies show that HFD might, in fact, be protective. The lack of a clear association between dietary fat consumption and AD-related pathology and cognitive function in AD mouse models might be explained by experimental variations, including AD mouse model, sex and age of the animals, composition of the HFD, and timeline of HFD consumption. In this review, we summarize recent studies that aimed at elucidating the effect of HFD-induced obesity on AD-related pathology in mice and provide an overview of the factors that may have contributed to the results reported in these studies. Based on the heterogeneity of these animal model studies and given that the human population itself is quite disparate, it is likely that people will benefit most from individualized nutritional plans based on their medical history and clinical profiles.
Collapse
Affiliation(s)
- Anna Amelianchik
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA
| | - Lauren Sweetland-Martin
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA.
| |
Collapse
|
22
|
Jeong S, Huang LK, Tsai MJ, Liao YT, Lin YS, Hu CJ, Hsu YH. Cognitive Function Associated with Gut Microbial Abundance in Sucrose and S-Adenosyl-L-Methionine (SAMe) Metabolic Pathways. J Alzheimers Dis 2022; 87:1115-1130. [DOI: 10.3233/jad-215090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background: Differential abundance of gut microbiota has found to be associated with Alzheimer’s disease (AD). However, the relative abundance of gut microbiota between dementia and mild cognitive impairment (MCI) in AD is not well studied. Objective: We attempted to identify differentially enriched gut microbes and their metabolic pathways in AD patients with dementia comparing to AD patients with MCI. Methods: Fecal samples were collected at Shuang Ho Hospital, Taipei Medical University, Taiwan and analyzed by whole metagenomic sequencing technique. For normal controls without AD (NC), 16S rRNA sequencing was obtained from the Taiwan Microbiome Database. A total of 48 AD (38 dementia and 10 MCI defined by cognitive function scores) and 50 NC were included. Microbiome alpha and beta diversities were estimated. Differentially enriched microbes were identified with HAllA, MaAsLin, DESeq2, and LEfSe statistical modeling approaches. Results: We found significantly increased abundance of Firmicutes but decreased abundance of Bacteroidetes at phylum level in AD compared to NC. In AD patients, cognitive function scores were negatively associated with abundance of Blautia hydrogenotrophica (Firmicutes), Anaerotruncus colihominis (Firmicutes), and Gordonibacter pamelaeae (Actinobacteria). In addition, microbial abundance in the sucrose and S-Adenosyl-L-methionine (SAMe) metabolic pathways were more enriched in MCI AD than dementia AD; and significantly associated with higher cognitive function scores. Conclusion: Gut microbe community diversity was similar in AD patients regardless of MCI or dementia status. However, differential analyses probed in lower-level taxa and metabolic pathways suggested that specific gut microbes in Firmicutes and Actinobacteria might involve in cognitive decline.
Collapse
Affiliation(s)
- Sohyun Jeong
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Li-Kai Huang
- Dementia Center and Department of Neurology, Shuang Ho Hospital, School of Medicine, College of Medicine, Taipei Medical University, New Taipei City, Taiwan
| | - Ming-Ju Tsai
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Yi-Tyng Liao
- Development Center for Biotechnology, Taipei, Taiwan
| | - Yow-Sien Lin
- Development Center for Biotechnology, Taipei, Taiwan
| | - Chaur-Jong Hu
- Dementia Center and Department of Neurology, Shuang Ho Hospital, School of Medicine, College of Medicine, Taipei Medical University, New Taipei City, Taiwan
| | - Yi-Hsiang Hsu
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
23
|
Cognitive reserve proxies, Alzheimer pathologies, and cognition. Neurobiol Aging 2022; 110:88-95. [PMID: 34879329 PMCID: PMC9234822 DOI: 10.1016/j.neurobiolaging.2021.10.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 10/09/2021] [Accepted: 10/10/2021] [Indexed: 02/03/2023]
Abstract
This study aimed to explore the moderating effects of the frequently used cognitive reserve (CR) proxies [i.e., education, premorbid intelligence quotient (pIQ), occupational complexity (OC), and lifetime cognitive activity (LCA)] on the relationships between various in vivo Alzheimer's disease (AD) pathologies and cognition. In total, 351 [268 cognitively unimpaired (CU), 83 cognitive impaired (CI)] older adults underwent multi-modal brain imaging to measure AD pathologies and cognitive assessments, and information on CR proxies was obtained. For overall participants, only education moderated the relationship between Aβ deposition and cognition. Education, pIQ, and LCA, but not OC, showed moderating effect on the relationship between AD-signature cerebral hypometabolism and cognition. In contrast, only OC had a moderating effect on the relationship between cortical atrophy of the AD-signature regions and cognition. Such moderation effects of the CR proxies were similarly observed in CI individuals, but most of them were not in CU individuals. The findings suggest that the proposed CR proxies have different moderating effects on the relationships between specific AD pathologies and cognition.
Collapse
|
24
|
Berkes M, Bialystok E. Bilingualism as a Contributor to Cognitive Reserve: What it Can do and What it Cannot do. Am J Alzheimers Dis Other Demen 2022; 37:15333175221091417. [PMID: 35470704 PMCID: PMC10581104 DOI: 10.1177/15333175221091417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In the absence of effective pharmacological interventions for the prevention of dementia, attention has turned to lifestyle factors that contribute to cognitive reserve. Although cognitive reserve cannot prevent the occurrence of disease, the trajectory is different for high reserve and low reserve patients, giving more time for independent living to high reserve individuals. We argue that lifelong bilingual experience meets the criteria for an experience that confers cognitive reserve, although neural reserve, a related concept, is more difficult to validate. Bilingual patients show symptoms at a later stage of disease and decline more rapidly than comparable monolingual patients. These patterns are considered in terms of evidence from behavioural, imaging and epidemiological studies. Finally, the role of bilingualism in protecting against symptoms of some forms of dementia are discussed in the context of other protective factors and the limits of this reserve approach in dealing with the consequences of dementia.
Collapse
|
25
|
Requena-Ocaña N, Araos P, Flores M, García-Marchena N, Silva-Peña D, Aranda J, Rivera P, Ruiz JJ, Serrano A, Pavón FJ, Suárez J, Rodríguez de Fonseca F. Evaluation of neurotrophic factors and education level as predictors of cognitive decline in alcohol use disorder. Sci Rep 2021; 11:15583. [PMID: 34341419 PMCID: PMC8328971 DOI: 10.1038/s41598-021-95131-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023] Open
Abstract
Cognitive reserve (CR) is the capability of an individual to cope with a brain pathology through compensatory mechanisms developed through cognitive stimulation by mental and physical activity. Recently, it has been suggested that CR has a protective role against the initiation of substance use, substance consumption patterns and cognitive decline and can improve responses to treatment. However, CR has never been linked to cognitive function and neurotrophic factors in the context of alcohol consumption. The present cross-sectional study aims to evaluate the association between CR (evaluated by educational level), cognitive impairment (assessed using a frontal and memory loss assessment battery) and circulating levels of brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) in patients with alcohol use disorder (AUD). Our results indicated that lower educational levels were accompanied by earlier onset of alcohol consumption and earlier development of alcohol dependence, as well as impaired frontal cognitive function. They also suggest that CR, NT-3 and BDNF may act as compensatory mechanisms for cognitive decline in the early stages of AUD, but not in later phases. These parameters allow the identification of patients with AUD who are at risk of cognitive deterioration and the implementation of personalized interventions to preserve cognitive function.
Collapse
Affiliation(s)
- Nerea Requena-Ocaña
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain.
- School of Psychology, Complutense University of Madrid, Madrid, Spain.
- Laboratorio de Investigación, IBIMA, Hospital Universitario Regional de Málaga, Avenida Carlos Haya 82, 29010, Málaga, Spain.
| | - Pedro Araos
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain
- Department of Psychobiology and Methodology of Behavioral Sciences, School of Psychology, University of Málaga, 29010, Málaga, Spain
| | - María Flores
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain
| | - Nuria García-Marchena
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain
| | - Daniel Silva-Peña
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain
| | - Jesús Aranda
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain
- School of Medicine, University of Málaga, 29071, Málaga, Spain
| | - Patricia Rivera
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain
| | - Juan Jesús Ruiz
- Provincial Drug Addiction Center of Málaga, Provincial Council of Málaga, Málaga, Spain
| | - Antonia Serrano
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain
| | - Francisco Javier Pavón
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain
- Cardiac Clinical Management Unit, IBIMA, University Hospital Virgen de la Victoria, 29010, Málaga, Spain
| | - Juan Suárez
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain.
- Department of Human Anatomy, Legal Medicine and History of Science, IBIMA, Facultad de Medicina, University of Málaga, Bulevar Louis Pausteur, 29071, Málaga, Spain.
| | - Fernando Rodríguez de Fonseca
- Mental Health Clinical Management Unit, Institute of Biomedical Research of Malaga-IBIMA, Regional University Hospital of Málaga, 29010, Málaga, Spain.
| |
Collapse
|
26
|
Kang SH, Kim ME, Jang H, Kwon H, Lee H, Kim HJ, Seo SW, Na DL. Amyloid Positivity in the Alzheimer/Subcortical-Vascular Spectrum. Neurology 2021; 96:e2201-e2211. [PMID: 33722997 DOI: 10.1212/wnl.0000000000011833] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 01/28/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE We investigated the frequency of β-amyloid (Aβ) positivity in 9 groups classified according to a combination of 3 different cognition states and 3 distinct levels of white matter hyperintensities (WMH) (minimal, moderate, and severe) and aimed to determine which factors were associated with Aβ after controlling for WMH and vice versa. METHODS A total of 1,047 individuals with subjective cognitive decline (SCD, n = 294), mild cognitive impairment (MCI, n = 237), or dementia (n = 516) who underwent Aβ PET scans were recruited from the memory clinic at Samsung Medical Center in Seoul, Korea. We investigated the following: (1) Aβ positivity in the 9 groups, (2) the relationship between Aβ positivity and WMH severity, and (3) clinical and genetic factors independently associated with Aβ or WMH. RESULTS Aβ positivity increased as the severity of cognitive impairment increased (SCD [15.7%], MCI [43.5%], and dementia [76.2%]), whereas it decreased as the severity of WMH increased (minimal [54.5%], moderate [53.9%], and severe [41.0%]) or the number of lacunes (0 [59.0%], 1-3 [42.0%], and >3 [23.4%]) increased. Aβ positivity was associated with higher education, absence of diabetes, and presence of APOE ε4 after controlling for cognitive and WMH status. CONCLUSION Our analysis of Aβ positivity involving a large sample classified according to the stratified cognitive states and WMH severity indicates that Alzheimer and cerebral small vessel diseases lie on a continuum. Our results offer clinicians insightful information about the association among Aβ, WMH, and cognition.
Collapse
Affiliation(s)
- Sung Hoon Kang
- From the Department of Neurology, Sungkyunkwan University School of Medicine (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), Neuroscience Center (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), and Stem Cell & Regenerative Medicine Institute (D.L.N.), Samsung Medical Center; Department of Neurology (S.H.K.), Korea University Guro Hospital, Korea University College of Medicine, Seoul; Chicago College of Osteopathic Medicine (M.E.K.), Midwestern University, IL; New York University (H.K.), NY; and Department of Health Sciences and Technology, SAIHST (D.L.N.), Sungkyunkwan University, Seoul, Korea
| | - Monica Eunseo Kim
- From the Department of Neurology, Sungkyunkwan University School of Medicine (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), Neuroscience Center (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), and Stem Cell & Regenerative Medicine Institute (D.L.N.), Samsung Medical Center; Department of Neurology (S.H.K.), Korea University Guro Hospital, Korea University College of Medicine, Seoul; Chicago College of Osteopathic Medicine (M.E.K.), Midwestern University, IL; New York University (H.K.), NY; and Department of Health Sciences and Technology, SAIHST (D.L.N.), Sungkyunkwan University, Seoul, Korea
| | - Hyemin Jang
- From the Department of Neurology, Sungkyunkwan University School of Medicine (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), Neuroscience Center (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), and Stem Cell & Regenerative Medicine Institute (D.L.N.), Samsung Medical Center; Department of Neurology (S.H.K.), Korea University Guro Hospital, Korea University College of Medicine, Seoul; Chicago College of Osteopathic Medicine (M.E.K.), Midwestern University, IL; New York University (H.K.), NY; and Department of Health Sciences and Technology, SAIHST (D.L.N.), Sungkyunkwan University, Seoul, Korea
| | - Hojeong Kwon
- From the Department of Neurology, Sungkyunkwan University School of Medicine (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), Neuroscience Center (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), and Stem Cell & Regenerative Medicine Institute (D.L.N.), Samsung Medical Center; Department of Neurology (S.H.K.), Korea University Guro Hospital, Korea University College of Medicine, Seoul; Chicago College of Osteopathic Medicine (M.E.K.), Midwestern University, IL; New York University (H.K.), NY; and Department of Health Sciences and Technology, SAIHST (D.L.N.), Sungkyunkwan University, Seoul, Korea
| | - Hyejoo Lee
- From the Department of Neurology, Sungkyunkwan University School of Medicine (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), Neuroscience Center (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), and Stem Cell & Regenerative Medicine Institute (D.L.N.), Samsung Medical Center; Department of Neurology (S.H.K.), Korea University Guro Hospital, Korea University College of Medicine, Seoul; Chicago College of Osteopathic Medicine (M.E.K.), Midwestern University, IL; New York University (H.K.), NY; and Department of Health Sciences and Technology, SAIHST (D.L.N.), Sungkyunkwan University, Seoul, Korea
| | - Hee Jin Kim
- From the Department of Neurology, Sungkyunkwan University School of Medicine (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), Neuroscience Center (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), and Stem Cell & Regenerative Medicine Institute (D.L.N.), Samsung Medical Center; Department of Neurology (S.H.K.), Korea University Guro Hospital, Korea University College of Medicine, Seoul; Chicago College of Osteopathic Medicine (M.E.K.), Midwestern University, IL; New York University (H.K.), NY; and Department of Health Sciences and Technology, SAIHST (D.L.N.), Sungkyunkwan University, Seoul, Korea
| | - Sang Won Seo
- From the Department of Neurology, Sungkyunkwan University School of Medicine (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), Neuroscience Center (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), and Stem Cell & Regenerative Medicine Institute (D.L.N.), Samsung Medical Center; Department of Neurology (S.H.K.), Korea University Guro Hospital, Korea University College of Medicine, Seoul; Chicago College of Osteopathic Medicine (M.E.K.), Midwestern University, IL; New York University (H.K.), NY; and Department of Health Sciences and Technology, SAIHST (D.L.N.), Sungkyunkwan University, Seoul, Korea
| | - Duk L Na
- From the Department of Neurology, Sungkyunkwan University School of Medicine (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), Neuroscience Center (S.H.K., H.J., H.L., H.J.K., S.W.S., D.L.N.), and Stem Cell & Regenerative Medicine Institute (D.L.N.), Samsung Medical Center; Department of Neurology (S.H.K.), Korea University Guro Hospital, Korea University College of Medicine, Seoul; Chicago College of Osteopathic Medicine (M.E.K.), Midwestern University, IL; New York University (H.K.), NY; and Department of Health Sciences and Technology, SAIHST (D.L.N.), Sungkyunkwan University, Seoul, Korea.
| |
Collapse
|
27
|
Alvares Pereira G, Silva Nunes MV, Alzola P, Contador I. Cognitive reserve and brain maintenance in aging and dementia: An integrative review. APPLIED NEUROPSYCHOLOGY-ADULT 2021; 29:1615-1625. [PMID: 33492168 DOI: 10.1080/23279095.2021.1872079] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This research is an integrative review of scientific evidence differentiating between cognitive reserve (CR) and brain maintenance concepts. Thus, we have examined how CR socio-behavioral proxies (i.e. education, occupational attainment, and leisure activities) may help to cope with age-related cognitive decline and negative consequences of brain pathology. We also analyze lifestyle factors associated with brain maintenance or the relative absence of change in neural resources over time. Medline and Web of Science databases were used for the bibliographic search in the last 20 years. Observational cohort studies were selected to analyze the effect of different CR proxies on cognitive decline, including dementia incidence, whereas studies employing functional neuroimaging (fMRI) were used to display the existence of compensation mechanisms. Besides, structural MRI studies were used to test the association between lifestyle factors and neural changes. Our findings suggest that education, leisure activities, and occupational activity are protective factors against cognitive decline and dementia. Moreover, functional neuroimaging studies have verified the existence of brain networks that may underlie CR. Therefore, CR may be expressed either through a more efficient utilization (neural reserve) of brain networks or the recruitment of additional brain regions (compensation). Finally, lifestyle factors such as abstaining from smoking, lower alcohol consumption, and physical activity contributed to brain maintenance and were associated with the preservation of cognitive function. Advances in multimodal neuroimaging studies, preferably longitudinal design, will allow a better understanding of the neural mechanisms associated with the prevention of cognitive decline and preservation of neural resources in aging.
Collapse
Affiliation(s)
| | - Maria Vânia Silva Nunes
- Health Sciences Institute, Portuguese Catholic University, Lisbon, Portugal.,Interdisciplinary Health Research Center, Lisbon, Portugal
| | - Patricia Alzola
- Department of Basic Psychology, Psychobiology and Methodology, University of Salamanca, Salamanca, Spain
| | - Israel Contador
- Department of Basic Psychology, Psychobiology and Methodology, University of Salamanca, Salamanca, Spain
| |
Collapse
|
28
|
The age-dependent associations of white matter hyperintensities and neurofilament light in early- and late-stage Alzheimer's disease. Neurobiol Aging 2020; 97:10-17. [PMID: 33070094 DOI: 10.1016/j.neurobiolaging.2020.09.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 01/19/2023]
Abstract
Neurofilament light (NFL) is an emerging marker of axonal degeneration. This study investigated the relationship between white matter hyperintensities (WMHs) and plasma NFL in a large elderly cohort with, and without, cognitive impairment. We used the Alzheimer's Disease Neuroimaging Initiative and included 163 controls, 103 participants with a significant memory concern, 279 with early mild cognitive impairment (EMCI), 152 with late mild cognitive impairment (LMCI), and 130 with Alzheimer's disease, with 3T MRI and plasma NFL data. Multiple linear regression models examined the relationship between WMHs and NFL, with and without age adjustment. We used smoking status, history of hypertension, history of diabetes, and BMI as additional covariates to examine the effect of vascular risk. We found increases of between 20% and 41% in WMH volume per 1SD increase in NFL in significant memory concern, early mild cognitive impairment, late mild cognitive impairment, and Alzheimer's disease groups (p < 0.02). Marked attenuation of the positive associations between WMHs and NFL were seen after age adjustment, suggesting that a significant proportion of the association between NFL and WMHs is age-related. No effect of vascular risk was observed. These results are supportive of a link between WMH and axonal degeneration in early to late disease stages, in an age-dependent, but vascular risk-independent manner.
Collapse
|
29
|
Zhong X, Frost DC, Yu Q, Li M, Gu TJ, Li L. Mass Defect-Based DiLeu Tagging for Multiplexed Data-Independent Acquisition. Anal Chem 2020; 92:11119-11126. [PMID: 32649829 PMCID: PMC7438256 DOI: 10.1021/acs.analchem.0c01136] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The unbiased selection of peptide precursors makes data-independent acquisition (DIA) an advantageous alternative to data-dependent acquisition (DDA) for discovery proteomics, but traditional multiplexed quantification approaches employing mass difference labeling or isobaric tagging are incompatible with DIA. Here, we describe a strategy that permits multiplexed quantification by DIA using mass defect-based N,N-dimethyl leucine (mdDiLeu) tags and high-resolution tandem mass spectrometry (MS2) analysis. Millidalton mass differences between mdDiLeu isotopologues produce fragment ion multiplet peaks separated in mass by as little as 5.8 mDa, enabling up to 4-plex quantification in DIA MS2 spectra. Quantitative analysis of yeast samples displayed comparable accuracy and precision for MS2-based DIA and MS1-based DDA methods. Multiplexed DIA analysis of cerebrospinal fluid revealed the dynamic proteome changes in Alzheimer's disease, demonstrating its utility for discovery of potential clinical biomarkers. We show that the mdDiLeu tagging approach for multiplexed DIA is a viable methodology for investigating proteome changes, particularly for low-abundance proteins, in different biological matrices.
Collapse
Affiliation(s)
- Xiaofang Zhong
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Dustin C. Frost
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Qinying Yu
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Miyang Li
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Ting-Jia Gu
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705, USA
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| |
Collapse
|
30
|
Arenaza-Urquijo EM, Przybelski SA, Machulda MM, Knopman DS, Lowe VJ, Mielke MM, Reddy AL, Geda YE, Jack CR, Petersen RC, Vemuri P. Better stress coping associated with lower tau in amyloid-positive cognitively unimpaired older adults. Neurology 2020; 94:e1571-e1579. [PMID: 31964689 PMCID: PMC7251516 DOI: 10.1212/wnl.0000000000008979] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 10/16/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Research in animals has shown that chronic stress exacerbates tau pathology. In humans, psychological stress has been associated with higher risk of Alzheimer disease clinical syndrome. The objective of this cross-sectional study was to assess the hypothesis that stress coping ability (assessed via the Brief Resilience Scale [BRS]) is associated with tau burden and to evaluate whether these associations differed by sex and amyloid status (A+/A-) in cognitively unimpaired (CU) older adults. METHODS We included 225 CU participants (mean age 70.4 ± 10.2 years, 48% female) enrolled in the population-based Mayo Clinic Study of Aging who completed the BRS and underwent amyloid-PET (Pittsburgh compound B-PET) and tau-PET (AV1451-PET). We fitted multiple regression and analysis of covariance models to assess the associations between BRS and tau-PET and the interaction with amyloid status and sex. We focused on entorhinal cortex (ERC) tau burden and also performed voxel-wise analyses. Age, sex, education, depression, and anxiety were considered as covariates. RESULTS Higher stress coping ability was associated with lower tau burden in the medial temporal lobe (including ERC) and occipito-temporal and cuneal/precuneal cortices. The association was present in both A+ and A- but weaker in A- CU older adults. There was an interaction between amyloid status and stress coping ability that was restricted to the medial temporal lobe tau such that A+ CU older adults with lower stress coping abilities showed higher tau. There were no significant interactions between stress coping and sex. CONCLUSIONS A faster termination of the stress response (higher coping ability) may limit the negative effects of stress on tau deposition. Conversely, lower stress coping ability may be an early sign of accumulating tau pathology. Longitudinal studies are warranted to clarify whether stress mechanisms act to exacerbate tau pathology or tau influences stress-related brain mechanisms and lowers the ability to cope with stress.
Collapse
Affiliation(s)
- Eider M Arenaza-Urquijo
- From the Departments of Radiology (E.M.A.-U., V.J.L., A.L.R., C.R.J., P.V.), Health Sciences Research (S.A.P., M.M.M.), Psychiatry and Psychology (M.M. Machulda), and Neurology (D.S.K., M.M. Mielke, R.C.P.), Mayo Clinic, Rochester, MN; and Department of Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ
| | - Scott A Przybelski
- From the Departments of Radiology (E.M.A.-U., V.J.L., A.L.R., C.R.J., P.V.), Health Sciences Research (S.A.P., M.M.M.), Psychiatry and Psychology (M.M. Machulda), and Neurology (D.S.K., M.M. Mielke, R.C.P.), Mayo Clinic, Rochester, MN; and Department of Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ
| | - Mary M Machulda
- From the Departments of Radiology (E.M.A.-U., V.J.L., A.L.R., C.R.J., P.V.), Health Sciences Research (S.A.P., M.M.M.), Psychiatry and Psychology (M.M. Machulda), and Neurology (D.S.K., M.M. Mielke, R.C.P.), Mayo Clinic, Rochester, MN; and Department of Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ
| | - David S Knopman
- From the Departments of Radiology (E.M.A.-U., V.J.L., A.L.R., C.R.J., P.V.), Health Sciences Research (S.A.P., M.M.M.), Psychiatry and Psychology (M.M. Machulda), and Neurology (D.S.K., M.M. Mielke, R.C.P.), Mayo Clinic, Rochester, MN; and Department of Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ
| | - Val J Lowe
- From the Departments of Radiology (E.M.A.-U., V.J.L., A.L.R., C.R.J., P.V.), Health Sciences Research (S.A.P., M.M.M.), Psychiatry and Psychology (M.M. Machulda), and Neurology (D.S.K., M.M. Mielke, R.C.P.), Mayo Clinic, Rochester, MN; and Department of Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ
| | - Michelle M Mielke
- From the Departments of Radiology (E.M.A.-U., V.J.L., A.L.R., C.R.J., P.V.), Health Sciences Research (S.A.P., M.M.M.), Psychiatry and Psychology (M.M. Machulda), and Neurology (D.S.K., M.M. Mielke, R.C.P.), Mayo Clinic, Rochester, MN; and Department of Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ
| | - Ashritha L Reddy
- From the Departments of Radiology (E.M.A.-U., V.J.L., A.L.R., C.R.J., P.V.), Health Sciences Research (S.A.P., M.M.M.), Psychiatry and Psychology (M.M. Machulda), and Neurology (D.S.K., M.M. Mielke, R.C.P.), Mayo Clinic, Rochester, MN; and Department of Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ
| | - Yonas E Geda
- From the Departments of Radiology (E.M.A.-U., V.J.L., A.L.R., C.R.J., P.V.), Health Sciences Research (S.A.P., M.M.M.), Psychiatry and Psychology (M.M. Machulda), and Neurology (D.S.K., M.M. Mielke, R.C.P.), Mayo Clinic, Rochester, MN; and Department of Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ
| | - Clifford R Jack
- From the Departments of Radiology (E.M.A.-U., V.J.L., A.L.R., C.R.J., P.V.), Health Sciences Research (S.A.P., M.M.M.), Psychiatry and Psychology (M.M. Machulda), and Neurology (D.S.K., M.M. Mielke, R.C.P.), Mayo Clinic, Rochester, MN; and Department of Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ
| | - Ronald C Petersen
- From the Departments of Radiology (E.M.A.-U., V.J.L., A.L.R., C.R.J., P.V.), Health Sciences Research (S.A.P., M.M.M.), Psychiatry and Psychology (M.M. Machulda), and Neurology (D.S.K., M.M. Mielke, R.C.P.), Mayo Clinic, Rochester, MN; and Department of Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ
| | - Prashanthi Vemuri
- From the Departments of Radiology (E.M.A.-U., V.J.L., A.L.R., C.R.J., P.V.), Health Sciences Research (S.A.P., M.M.M.), Psychiatry and Psychology (M.M. Machulda), and Neurology (D.S.K., M.M. Mielke, R.C.P.), Mayo Clinic, Rochester, MN; and Department of Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ.
| |
Collapse
|
31
|
Rodriguez Then FS, Jackson J, Ware C, Churchyard R, Hanseeuw B. Interdisciplinary and Transdisciplinary Perspectives: On the Road to a Holistic Approach to Dementia Prevention and Care. J Alzheimers Dis Rep 2020; 4:39-48. [PMID: 32206756 PMCID: PMC7081086 DOI: 10.3233/adr-180070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Dementia, of which the most frequent form is Alzheimer's disease, is a chronic and terminal condition with multi-factorial causes and numerous consequences on a patient's life. Combining perspectives from different disciplines seems necessary for unraveling dementia's entangled issues. Current dementia management is a multidisciplinary effort; however, integrating different disciplines as a holistic treatment process is often hindered due to different responsibilities, various conceptual approaches, and distinctive research methods. With this paper, we raise some of the challenges that need to be addressed in order to initiate an interdisciplinary or even transdisciplinary research agenda. We also outline recommendations on how to integrate multiple disciplinary perspectives in dementia care and research. We see opportunities for young investigators to draw from different fields of research in dementia as their research focus is still developing. By establishing common objectives with investigators from other fields, we can pursue the goal of improving treatment and care as a team-meaning accomplishing different tasks but sharing a common purpose. It is necessary to address the communication between fields that limits the understanding of connections between cognitive symptoms, biological processes, treatment, lifestyle, and care giving in order to reach the aim of developing a holistic, person-centered, patient-first approach. Associating biomedical research to field experience from care professionals and the study of human science will promote a more independent, social, and sustainable lifestyle for people with dementia.
Collapse
Affiliation(s)
- Francisca S Rodriguez Then
- German Center for Neurodegenerative Diseases (DZNE), RG Psychosocial Epidemiology & Public Health, Greifswald, Germany.,Edward R. Roybal Institute on Aging, University of Southern California, Los Angeles, CA, USA.,Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, Leipzig, Germany
| | - Jonathan Jackson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Caitlin Ware
- Department of Psychoanalytical Studies, Université de Paris, CRPMS, F-75013, Paris, France.,Institut National de la Santé et de la Recherche Médicale, (INSERM) Inserm UMR-S 1237, Normandie Univ, UNICAEN, GIP Cyceron, France
| | - Rebekah Churchyard
- Factor-Inwentash Faculty of Social Work, University of Toronto, Toronto, ON, Canada
| | - Bernard Hanseeuw
- Cliniques Universitaires Saint-Luc, Department of Neurology, Brussels, Belgium.,Université Catholique de Louvain, Institute of Neurosciences, Brussels, Belgium.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, and the GordonCenter for Medical Imaging, Charlestown, MA, USA
| | | |
Collapse
|
32
|
Rodriguez FS, Zheng L, Chui HC. Psychometric Characteristics of Cognitive Reserve: How High Education Might Improve Certain Cognitive Abilities in Aging. Dement Geriatr Cogn Disord 2020; 47:335-344. [PMID: 31466060 DOI: 10.1159/000501150] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/23/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The capacity to mitigate dementia symptomology despite the prevailing brain pathology has been attributed to cognitive reserve. OBJECTIVES This study aimed to investigate how psychometric performance differs between individuals with a high school versus college education (surrogate measures for medium and high cognitive reserves) given the same level of brain pathology assessed using quantitative structural MRI. METHODS We used data from the Aging Brain: Vasculature, Ischemia, and Behavior Study (ABVIB). Cognition was assessed using a neuropsychological battery that included those contained in the National Alzheimer's Coordinating Center (NACC) uniform data set. Participants with a medium and high cognitive reserve were matched by level of structural MRI changes, gender, and age. RESULTS Matched-pair regression analyses indicated that individuals with a higher education had a significantly better performance in recognition and verbal fluency animals, working memory, and processing speed in complex tasks. Moreover, they had a better performance in interference trails compared to individuals with a high school education (medium cognitive reserve). CONCLUSIONS Our findings suggest that, given the same level of brain pathology, individuals with a higher education (cognitive reserve) benefit from a superior performance in semantic memory and executive functioning. Differences in these cognitive domains may be key pathways explaining how individuals with a high cognitive reserve are able to diminish dementia symptomatology despite physical changes in the brain.
Collapse
Affiliation(s)
- Francisca S Rodriguez
- Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, Leipzig, Germany, .,Center for Cognitive Science, University of Kaiserslautern, Kaiserslautern, Germany,
| | - Ling Zheng
- USC Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Helena C Chui
- USC Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | |
Collapse
|
33
|
Pettigrew C, Soldan A, Zhu Y, Cai Q, Wang MC, Moghekar A, Miller MI, Singh B, Martinez O, Fletcher E, DeCarli C, Albert M. Cognitive reserve and rate of change in Alzheimer's and cerebrovascular disease biomarkers among cognitively normal individuals. Neurobiol Aging 2019; 88:33-41. [PMID: 31932050 DOI: 10.1016/j.neurobiolaging.2019.12.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/27/2019] [Accepted: 12/06/2019] [Indexed: 01/03/2023]
Abstract
We examined whether cognitive reserve (CR) impacts level of, or rate of change in, biomarkers of Alzheimer's disease (AD) and small-vessel cerebrovascular disease in >250 individuals who were cognitively normal and middle-aged and older at the baseline. The four primary biomarker categories commonly examined in studies of AD were measured longitudinally: cerebrospinal fluid measures of amyloid (A) and tau (T); cerebrospinal fluid and neuroimaging measures of neuronal injury (N); and neuroimaging measures of white matter hyperintensities (WMHs) to assess cerebrovascular pathology (V). CR was indexed by a composite score including years of education, reading, and vocabulary test performance. Higher CR was associated with lower levels of WMHs, particularly among those who subsequently progressed from normal cognition to MCI. CR was not associated with WMH trajectories. In addition, CR was not associated with either levels of, or rate of change in, A/T/N biomarkers. This may suggest that higher CR is associated with lifestyle factors that reduce levels of cerebrovascular disease, allowing individuals with higher CR to better tolerate other types of pathology.
Collapse
Affiliation(s)
- Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuxin Zhu
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Qing Cai
- Lyft, Inc., San Francisco, CA, USA
| | - Mei-Cheng Wang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael I Miller
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Baljeet Singh
- Department of Neurology, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Oliver Martinez
- Department of Neurology, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Evan Fletcher
- Department of Neurology, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Charles DeCarli
- Department of Neurology, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
34
|
Menardi A, Pascual-Leone A, Fried PJ, Santarnecchi E. The Role of Cognitive Reserve in Alzheimer's Disease and Aging: A Multi-Modal Imaging Review. J Alzheimers Dis 2019; 66:1341-1362. [PMID: 30507572 DOI: 10.3233/jad-180549] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Comforts in modern society have generally been associated with longer survival rates, enabling individuals to reach advanced age as never before in history. With the increase in longevity, however, the incidence of neurodegenerative diseases, especially Alzheimer's disease, has also doubled. Nevertheless, most of the observed variance, in terms of time of clinical diagnosis and progression, often remains striking. Only recently, differences in the social, educational and occupational background of the individual, as proxies of cognitive reserve (CR), have been hypothesized to play a role in accounting for such discrepancies. CR is a well-established concept in literature; lots of studies have been conducted in trying to better understand its underlying neural substrates and associated biomarkers, resulting in an incredible amount of data being produced. Here, we aimed to summarize recent relevant published work addressing the issue, gathering evidence for the existence of a common path across research efforts that might ease future investigations by providing a general perspective on the actual state of the arts. An innovative model is hereby proposed, addressing the role of CR across structural and functional evidences, as well as the potential implementation of non-invasive brain stimulation techniques in the causal validation of such theoretical frame.
Collapse
Affiliation(s)
- Arianna Menardi
- Brain Investigation and Neuromodulation Lab, Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy.,Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alvaro Pascual-Leone
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Peter J Fried
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Emiliano Santarnecchi
- Brain Investigation and Neuromodulation Lab, Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy.,Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Abstract
The concept of cognitive reserve (CR) was proposed to account for the discrepancy between levels of brain pathologic process or damage and clinical and cognitive function. We provide a detailed review of prospective longitudinal studies that have investigated the interaction between CR and Alzheimer disease (AD) biomarkers on clinical and cognitive outcomes among individuals with normal cognition at baseline. Current evidence is consistent with the view that higher levels of CR are associated with a delay in the onset of symptoms of mild cognitive impairment and that there may be multiple pathways by which CR exerts its protective effects.
Collapse
|
36
|
Ramanan VK, Castillo AM, Knopman DS, Graff-Radford J, Lowe VJ, Petersen RC, Jack CR, Mielke MM, Vemuri P. Association of Apolipoprotein E ɛ4, Educational Level, and Sex With Tau Deposition and Tau-Mediated Metabolic Dysfunction in Older Adults. JAMA Netw Open 2019; 2:e1913909. [PMID: 31642932 PMCID: PMC6820045 DOI: 10.1001/jamanetworkopen.2019.13909] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022] Open
Abstract
Importance While amyloidosis is an early event in the Alzheimer disease (AD) biomarker cascade, a complex interplay among the apolipoprotein E (APOE) ɛ4 allele, educational levels, and sex may be associated with an individual's resilience to dementia. Objective To assess whether APOE ɛ4, educational levels, and sex are associated with regional tau deposition and tau-mediated metabolic dysfunction in older adults. Design, Setting, and Participants Population-based cohort study of individuals aged 65 years and older enrolled between January 1, 2004, and May 1, 2018, in the Mayo Clinic Study of Aging, a prospective longitudinal study of cognitive aging in Olmsted County, Minnesota. Main Outcomes and Measures The primary outcomes were cross-sectional tau burden and the fluorodeoxyglucose (FDG) to tau ratio (as a measure of tau-mediated metabolic dysfunction) assessed by positron emission tomography for 43 atlas-defined regions, with specific focus on the entorhinal, inferior temporal, and posterior cingulate cortices. Exposures Using linear regression, APOE ɛ4 status and years of education were the primary exposure variables, with sex additionally investigated through interaction models. Results The sample included 325 individuals (173 [53%] male; mean [SD] age, 76.1 [7.2] years; 291 [90%] cognitively unimpaired). Although APOE ɛ4 was nominally associated with higher tau deposition (β = 0.05 [95% CI, 0.02-0.09]; P = .001; Cohen d = 0.40) and lower FDG to tau ratio (β = -0.05 [95% CI, -0.08 to -0.01]; P = .008; Cohen d = 0.33) in the entorhinal cortex, these associations were completely attenuated after controlling for global amyloid burden. Education was not associated with regional tau burden or FDG to tau ratio. In the 3 regions of interest, global amyloid burden accounted for the largest proportion of variance in tau deposition among the candidate variables assessed. In the entorhinal cortex, significant interactions were identified between APOE ɛ4 and global amyloid burden on tau (β = 0.25; SE = 0.06; P < .001) and between sex and tau burden on FDG metabolism (β = 0.10; SE = 0.05; P = .049). Conclusions and Relevance These results suggest that (1) tau deposition is most significantly associated with amyloidosis; (2) in the presence of abundant amyloidosis, APOE ɛ4 may be associated with accelerated entorhinal cortex tau deposition; and (3) women may have lower resilience to tau, manifested by a higher degree of metabolic dysfunction in the entorhinal cortex in response to tau pathology.
Collapse
Affiliation(s)
- Vijay K Ramanan
- Department of Neurology, Mayo Clinic–Rochester, Rochester, Minnesota
| | - Anna M. Castillo
- Department of Health Sciences Research, Mayo Clinic–Rochester, Rochester, Minnesota
| | - David S. Knopman
- Department of Neurology, Mayo Clinic–Rochester, Rochester, Minnesota
| | | | - Val J. Lowe
- Department of Radiology, Mayo Clinic–Rochester, Rochester, Minnesota
| | - Ronald C. Petersen
- Department of Neurology, Mayo Clinic–Rochester, Rochester, Minnesota
- Department of Health Sciences Research, Mayo Clinic–Rochester, Rochester, Minnesota
| | - Clifford R. Jack
- Department of Radiology, Mayo Clinic–Rochester, Rochester, Minnesota
| | - Michelle M. Mielke
- Department of Neurology, Mayo Clinic–Rochester, Rochester, Minnesota
- Department of Health Sciences Research, Mayo Clinic–Rochester, Rochester, Minnesota
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic–Rochester, Rochester, Minnesota
| |
Collapse
|
37
|
Gazzina S, Grassi M, Premi E, Cosseddu M, Alberici A, Archetti S, Gasparotti R, Van Swieten J, Galimberti D, Sanchez-Valle R, Laforce RJ, Moreno F, Synofzik M, Graff C, Masellis M, Tartaglia MC, Rowe JB, Vandenberghe R, Finger E, Tagliavini F, de Mendonça A, Santana I, Butler CR, Ducharme S, Gerhard A, Danek A, Levin J, Otto M, Frisoni G, Sorbi S, Padovani A, Rohrer JD, Borroni B. Education modulates brain maintenance in presymptomatic frontotemporal dementia. J Neurol Neurosurg Psychiatry 2019; 90:1124-1130. [PMID: 31182509 DOI: 10.1136/jnnp-2019-320439] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Cognitively engaging lifestyles have been associated with reduced risk of conversion to dementia. Multiple mechanisms have been advocated, including increased brain volumes (ie, brain reserve) and reduced disease progression (ie, brain maintenance). In cross-sectional studies of presymptomatic frontotemporal dementia (FTD), higher education has been related to increased grey matter volume. Here, we examine the effect of education on grey matter loss over time. METHODS Two-hundred twenty-nine subjects at-risk of carrying a pathogenic mutation leading to FTD underwent longitudinal cognitive assessment and T1-weighted MRI at baseline and at 1 year follow-up. The first principal component score of the graph-Laplacian Principal Component Analysis on 112 grey matter region-of-interest volumes was used to summarise the grey matter volume (GMV). The effects of education on cognitive performances and GMV at baseline and on the change between 1 year follow-up and baseline (slope) were tested by Structural Equation Modelling. RESULTS Highly educated at-risk subjects had better cognition and higher grey matter volume at baseline; moreover, higher educational attainment was associated with slower loss of grey matter over time in mutation carriers. CONCLUSIONS This longitudinal study demonstrates that even in presence of ongoing pathological processes, education may facilitate both brain reserve and brain maintenance in the presymptomatic phase of genetic FTD.
Collapse
Affiliation(s)
- Stefano Gazzina
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Mario Grassi
- Department of Brain and Behavioral Science, Medical and Genomic Statistics Unit, University of Pavia, Pavia, Italy
| | - Enrico Premi
- Stroke Unit, Neurology Unit, Spedali Civili Hospital, Brescia, Italy
| | | | - Antonella Alberici
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvana Archetti
- Biotechnology Laboratory, Department of Diagnostics, Spedali Civili Hospital, Brescia, Italy
| | | | - John Van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Daniela Galimberti
- Centro Dino Ferrari, University of Milan, Milan, Italy.,Neurodegenerative Diseases Unit, Fondazione IRCSS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Raquel Sanchez-Valle
- Neurology Department, Hospital Clinic, Institut d'Investigacions Biomèdiques, Barcelona, Spain
| | - Robert Jr Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| | - Fermin Moreno
- Department of Neurology, Hospital Universitario Donostia, San Sebastian, Gipuzkoa, Spain
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research & Center of Neurology, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Caroline Graff
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogenetics, Stockholm, Sweden
| | - Mario Masellis
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Maria Carmela Tartaglia
- Toronto Western Hospital, Tanz Centre for Research in Neurodegenerative Disease, Toronto, Ontario, Canada
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Rik Vandenberghe
- Department of Neurosciences, Laboratory for Cognitive Neurology, KU Leuven, Leuven, Belgium
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Fabrizio Tagliavini
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Isabel Santana
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | | | - Simon Ducharme
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, Quebec, Canada.,McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Alex Gerhard
- Institute of Brain, Behaviour and Mental Health, The University of Manchester, Withington, Manchester, United Kingdom
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, Munich, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, Munich, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Markus Otto
- Department of Neurology, University Hospital Ulm, Ulm, Germany
| | - Giovanni Frisoni
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Memory Clinic and LANVIE-Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) "Don Gnocchi", Florence, Italy
| | - Alessandro Padovani
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Jonathan D Rohrer
- Dementia Research Centre, UCL Institute of Neurology, London, United Kingdom
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | | |
Collapse
|
38
|
Giovacchini G, Giovannini E, Borsò E, Lazzeri P, Riondato M, Leoncini R, Duce V, Mansi L, Ciarmiello A. The brain cognitive reserve hypothesis: A review with emphasis on the contribution of nuclear medicine neuroimaging techniques. J Cell Physiol 2019; 234:14865-14872. [PMID: 30784080 DOI: 10.1002/jcp.28308] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/09/2019] [Accepted: 01/24/2019] [Indexed: 01/24/2023]
Abstract
Neuropathological and clinical evidence indicates that the clinical expression of Alzheimer's disease (AD) occurs as neuropathology exceeds the brain reserve capacity. The brain or cognitive reserve (BCR) hypothesis states that high premorbid intelligence, education, and an active and stimulating lifestyle provide reserve capacity, which acts as a buffer against the cognitive deficits due to accumulating neuropathology. Neuroimaging studies that assessed the BCR hypothesis are critically reviewed with emphasis on study design and statistical analysis. Many studies were performed in the last two decades owing to the increasing availability of positron emission tomography (PET) and PET/computed tomography scanners and to the synthesis of new radiopharmaceuticals, including tracers for amyloid and tau proteins. Studies with different tracers provided complementary consistent results supporting the BCR hypothesis. Many studies were appropriately designed with a measure of reserve, a measure of brain anatomy/function/neuropathology, and a measure of cognitive functions that are necessary. Most of the early studies were performed with PET and [ 18 F]fluorodeoxyglucose, and occasionally with [ 15 O]water, reporting a significant association between higher occupation/education and lower glucose metabolism (blood flow) in associative temporo-parietal cortex in patients with AD and also in patients with MCI, after correcting for the degree in the cognitive impairment. On the contrary, performances on several neuropsychological tests increased with increasing education for participants with elevated [ 11 C]PiB uptake. Studies with the tracers specific for tau protein showed that patients with AD with elevated tau deposits had higher cognitive performances compared with patients with similar levels of tau deposits. BCR in AD is also associated with a preserved cholinergic function. The BCR hypothesis has been validated with methodologically sound study designs and sophisticated neuroimaging techniques using different radiotracers and providing an explanation for neuropathological and clinical observations on patients with AD.
Collapse
Affiliation(s)
| | | | - Elisa Borsò
- Department of Nuclear Medicine, S. Andrea Hospital, La Spezia, Italy
| | - Patrizia Lazzeri
- Department of Nuclear Medicine, S. Andrea Hospital, La Spezia, Italy
| | - Mattia Riondato
- Department of Nuclear Medicine, S. Andrea Hospital, La Spezia, Italy
| | - Rossella Leoncini
- Department of Nuclear Medicine, S. Andrea Hospital, La Spezia, Italy
| | - Valerio Duce
- Department of Nuclear Medicine, S. Andrea Hospital, La Spezia, Italy
| | - Luigi Mansi
- Department of Nuclear Medicine, S. Andrea Hospital, La Spezia, Italy
| | - Andrea Ciarmiello
- Department of Nuclear Medicine, S. Andrea Hospital, La Spezia, Italy
| |
Collapse
|
39
|
Vadini F, Santilli F, Casalini G, dell'Isola M, Iuliani O, D'Ardes D, Lattanzio L, Di Nicola M, Di Iorio G, Accorsi P. Homocysteine and education but not lipoprotein (a) predict estimated 10-year risk of cardiovascular disease in blood donors: a community based cross-sectional study. BMC Cardiovasc Disord 2019; 19:177. [PMID: 31349819 PMCID: PMC6660663 DOI: 10.1186/s12872-019-1157-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/15/2019] [Indexed: 11/24/2022] Open
Abstract
Background With aging of the population, screening and prevention health programs for blood donors will increasingly be a priority. We aimed at: assessing the 10 year-cardiovascular disease (CVD) risk in blood donors, according to Italian CUORE risk score (CRS); determining the association of homocysteine (Hcy), lipoprotein (Lp)(a) and socio-demographic or lifestyle variables with estimated 10-year CVD risk. Methods Between June 2015 and July 2017, 1,447 (61.2% men) unselected blood donors (aged 18–69 years) were enrolled at the Blood Transfusion Service of the Pescara General Hospital, Italy. The project entailed evaluation of unalterable (age and gender) and modifiable CV risk factors (total cholesterol, HDL, LDL, triglycerides, fasting glucose, smoking, hypertension). The educational attainment, socio-demographic and lifestyle behavior information were obtained through a structured self-report questionnaire, and Health-related quality of life (HRQoL) through the Short Form Survey (SF-12). Plasma Hcy and Lp(a) were determined in the fasting state. Results A CRS within the moderate-high risk range was reported in 21.7% donors. Multivariate logistic regression, after adjustment for clinical and demographic variables, showed that Hcy [OR (95% CI): 1.09 (1.04–1.13); p < 0.001) and low educational attainment [1.71 (1.09–2.73); p = 0.019] are independent risk factors for moderate-to-high CVD risk. Instead, Lp(a), evaluated in 774 donors, was > 30 mg/dL in 22.4% of the examined population, but without any significant correlation with CRS. Conclusions Our study highlights a previously unappreciated need for CV risk assessment in blood donors, which may include evaluation of educational attainment as a non-traditional risk marker.
Collapse
Affiliation(s)
- Francesco Vadini
- FIDAS (Italian Federation of Associations of Blood Donors), Pescara, Italy.,Psychoinfectivology Service, Infectious Disease Unit, Pescara General Hospital, Pescara, Italy
| | - Francesca Santilli
- Department of Medicine and Aging, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy. .,Center of Aging Science and Translational Medicine (CESI-Met), "G. D'Annunzio" University Foundation, Via Luigi Polacchi, 66013, Chieti, Italy.
| | - Giuseppe Casalini
- Department of Hematology, Transfusion Medicine and Biotechnologies, Pescara General Hospital, Pescara, Italy
| | - Mario dell'Isola
- Department of Hematology, Transfusion Medicine and Biotechnologies, Pescara General Hospital, Pescara, Italy
| | - Ornella Iuliani
- Department of Hematology, Transfusion Medicine and Biotechnologies, Pescara General Hospital, Pescara, Italy
| | - Damiano D'Ardes
- Department of Medicine and Aging, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Luisa Lattanzio
- FIDAS (Italian Federation of Associations of Blood Donors), Pescara, Italy
| | - Marta Di Nicola
- Department of Medicine and Aging, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | | | - Patrizia Accorsi
- Department of Hematology, Transfusion Medicine and Biotechnologies, Pescara General Hospital, Pescara, Italy
| |
Collapse
|
40
|
Wang N, Chen J, Xiao H, Wu L, Jiang H, Zhou Y. Application of artificial neural network model in diagnosis of Alzheimer's disease. BMC Neurol 2019; 19:154. [PMID: 31286894 PMCID: PMC6613238 DOI: 10.1186/s12883-019-1377-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Background Alzheimer’s disease has become a public health crisis globally due to its increasing incidence. The purpose of this study was to establish an early warning model using artificial neural network (ANN) for early diagnosis of AD and to explore early sensitive markers for AD. Methods A population based nested case-control study design was used. 89 new AD cases with good compliance who were willing to provide urine and blood specimen were selected from the cohort of 2482 community-dwelling elderly aged 60 years and over from 2013 to 2016. For each case, two controls living nearby were identified. Biomarkers for AD in urine and blood, neuropsychological functions and epidemiological parameters were included to analyze potential risk factors of AD. Compared with logistic regression, k-Nearest Neighbor (kNN) and support vector machine (SVM) model, back-propagation neural network of three-layer topology structures was applied to develop the early warning model. The performance of all models were measured by sensitivity, specificity, accuracy, positive prognostic value (PPV), negative prognostic value (NPV), the area under curve (AUC), and were validated using bootstrap resampling. Results The average age of AD group was about 5 years older than the non-AD controls (P < 0.001). Patients with AD included a significantly larger proportion of subjects with family history of dementia, compared with non-AD group. After adjusting for age and gender, the concentrations of urinary AD7c-NTP and aluminum in blood were significantly higher in AD group than non-AD group (2.01 ± 1.06 vs 1.03 ± 0.43, 1.74 ± 0.62 vs 1.24 ± 0.41 respectively), but the concentration of Selenium in AD group (2.26 ± 0.59) was significantly lower than that in non-AD group (2.61 ± 1.07). All the models were established using 18 variables that were significantly different between AD patients and controls as independent variables. The ANN model outperformed the other classifiers. The AUC for this ANN was 0.897 and the model obtained the accuracy of 92.13%, the sensitivity of 87.28% and the specificity of 94.74% on the average. Conclusions Increased risk of AD may be associated with higher age among senior citizens in urban communities. Urinary AD7c-NTP is clinically valuable for the early diagnosis. The established ANN model obtained a high accuracy and diagnostic efficiency, which could be a low-cost practicable tool for the screening and diagnosis of AD for citizens.
Collapse
Affiliation(s)
- Naibo Wang
- Jiangxi Province Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, 330006, People's Republic of China.,Jiangxi Centre for Health Education and Promotion, Nanchang, China
| | - Jinghua Chen
- Jiangxi Province Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Hui Xiao
- Jiangxi Province Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Lei Wu
- Jiangxi Province Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, 330006, People's Republic of China.
| | - Han Jiang
- Second Affiliated Hospital, Nanchang University, Nanchang, China.
| | - Yueping Zhou
- Jiangxi Province Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, 330006, People's Republic of China
| |
Collapse
|
41
|
Erickson CM, Schultz SA, Oh JM, Darst BF, Ma Y, Norton D, Betthauser T, Gallagher CL, Carlsson CM, Bendlin BB, Asthana S, Hermann BP, Sager MA, Blennow K, Zetterberg H, Engelman CD, Christian BT, Johnson SC, Dubal DB, Okonkwo OC. KLOTHO heterozygosity attenuates APOE4-related amyloid burden in preclinical AD. Neurology 2019; 92:e1878-e1889. [PMID: 30867273 PMCID: PMC6550504 DOI: 10.1212/wnl.0000000000007323] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 12/05/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE To examine whether the KLOTHO gene variant KL-VS attenuates APOE4-associated β-amyloid (Aβ) accumulation in a late-middle-aged cohort enriched with Alzheimer disease (AD) risk factors. METHODS Three hundred nine late-middle-aged adults from the Wisconsin Registry for Alzheimer's Prevention and the Wisconsin Alzheimer's Disease Research Center were genotyped to determine KL-VS and APOE4 status and underwent CSF sampling (n = 238) and/or 11C-Pittsburgh compound B (PiB)-PET imaging (n = 183). Covariate-adjusted regression analyses were used to investigate whether APOE4 exerted expected effects on Aβ burden. Follow-up regression analyses stratified by KL-VS genotype (i.e., noncarrier vs heterozygous; there were no homozygous individuals) evaluated whether the influence of APOE4 on Aβ was different among KL-VS heterozygotes compared to noncarriers. RESULTS APOE4 carriers exhibited greater Aβ burden than APOE4-negative participants. This effect was stronger in CSF (t = -5.12, p < 0.001) compared with PiB-PET (t = 3.93, p < 0.001). In the stratified analyses, this APOE4 effect on Aβ load was recapitulated among KL-VS noncarriers (CSF: t = -5.09, p < 0.001; PiB-PET: t = 3.77, p < 0 .001). In contrast, among KL-VS heterozygotes, APOE4-positive individuals did not exhibit higher Aβ burden than APOE4-negative individuals (CSF: t = -1.03, p = 0.308; PiB-PET: t = 0.92, p = 0.363). These differential APOE4 effects remained after KL-VS heterozygotes and noncarriers were matched on age and sex. CONCLUSION In a cohort of at-risk late-middle-aged adults, KL-VS heterozygosity was associated with an abatement of APOE4-associated Aβ aggregation, suggesting KL-VS heterozygosity confers protections against APOE4-linked pathways to disease onset in AD.
Collapse
Affiliation(s)
- Claire M Erickson
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Stephanie A Schultz
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Jennifer M Oh
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Burcu F Darst
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Yue Ma
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Derek Norton
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Tobey Betthauser
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Catherine L Gallagher
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Cynthia M Carlsson
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Barbara B Bendlin
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Sanjay Asthana
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Bruce P Hermann
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Mark A Sager
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Kaj Blennow
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Henrik Zetterberg
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Corinne D Engelman
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Bradley T Christian
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Sterling C Johnson
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Dena B Dubal
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Ozioma C Okonkwo
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco.
| |
Collapse
|
42
|
Haapalinna F, Kokki M, Jääskeläinen O, Hallikainen M, Helisalmi S, Koivisto A, Kokki H, Paajanen T, Penttinen J, Pikkarainen M, Rautiainen M, Soininen H, Solje E, Remes AM, Herukka SK. Subtle Cognitive Impairment and Alzheimer's Disease-Type Pathological Changes in Cerebrospinal Fluid are Common Among Neurologically Healthy Subjects. J Alzheimers Dis 2019; 62:165-174. [PMID: 29439329 DOI: 10.3233/jad-170534] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND The neuropathology of Alzheimer's disease (AD) has previously been shown to be rather common among the elderly. OBJECTIVE The aim of this study was to inspect the associations between cerebrospinal fluid (CSF) AD biomarker concentrations, age, the APOEɛ4 allele, cardiovascular diseases, diabetes, and cognitive performance in a cohort of a neurologically healthy population. METHODS This study included 93 subjects (42 men, mean age 67 years) without previous neurological symptoms or subjective cognitive complaints. Their cognition was assessed, and CSF biomarkers and APOEɛ4 status were analyzed. RESULTS Of the studied subjects, 8.6% (n = 8) had a pathological CSF AD biomarker profile. An increase in age correlated positively with CSF tau pathology and negatively with global cognitive performance. CONCLUSION AD-type pathological changes in CSF and subtle cognitive impairment are common within a population with no previous memory complaints. Age was the main risk factor for the changes.
Collapse
Affiliation(s)
- Fanni Haapalinna
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Merja Kokki
- Department of Anesthesia and Operative Services, Kuopio University Hospital, Kuopio, Finland
| | - Olli Jääskeläinen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Merja Hallikainen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Seppo Helisalmi
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Anne Koivisto
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland.,Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Hannu Kokki
- Institute of Clinical Medicine, Anesthesiology and Intensive Care, University of Eastern Finland, Kuopio, Finland
| | - Teemu Paajanen
- Research and Service Centre for Occupational Health, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Janne Penttinen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Maria Pikkarainen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Minna Rautiainen
- Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Hilkka Soininen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland.,Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Eino Solje
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Anne M Remes
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland.,Department of Neurology, Kuopio University Hospital, Kuopio, Finland.,Medical Research Center, Oulu University Hospital, Oulu, Finland.,Research Unit of Clinical Neuroscience, Neurology, University of Oulu, Oulu, Finland
| | - Sanna-Kaisa Herukka
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
43
|
Zhong X, Yu Q, Ma F, Frost DC, Lu L, Chen Z, Zetterberg H, Carlsson C, Okonkwo O, Li L. HOTMAQ: A Multiplexed Absolute Quantification Method for Targeted Proteomics. Anal Chem 2019; 91:2112-2119. [PMID: 30608134 PMCID: PMC6379083 DOI: 10.1021/acs.analchem.8b04580] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Absolute quantification in targeted proteomics is challenging due to a variety of factors, including low specificity in complex backgrounds, limited analytical throughput, and wide dynamic range. To address these problems, we developed a hybrid offset-triggered multiplex absolute quantification (HOTMAQ) strategy that combines cost-effective mass difference and isobaric tags to enable simultaneous construction of an internal standard curve in the MS1 precursor scan, real-time identification of peptides at the MS2 level, and mass offset-triggered accurate quantification of target proteins in synchronous precursor selection (SPS)-MS3 spectra. This approach increases the analytical throughput of targeted quantitative proteomics by up to 12-fold. The HOTMAQ strategy was employed to verify candidate protein biomarkers in preclinical Alzheimer's disease with high accuracy. The greatly enhanced throughput and quantitative performance, paired with sample flexibility, makes HOTMAQ broadly applicable to targeted peptidomics, proteomics, and phosphoproteomics.
Collapse
Affiliation(s)
- Xiaofang Zhong
- School of Pharmacy , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Qinying Yu
- School of Pharmacy , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Fengfei Ma
- School of Pharmacy , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Dustin C Frost
- School of Pharmacy , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Lei Lu
- School of Pharmacy , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Zhengwei Chen
- Department of Chemistry , University of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Sahlgrenska Academy , University of Gothenburg , S-431 80 , Gothenburg , Sweden
- Clinical Neurochemistry Laboratory , Sahlgrenska University Hospital , S-431 80 , Mölndal , Sweden
- Department of Molecular Neuroscience , UCL Institute of Neurology , Queen Square, London , WC1N 3BG , U.K
- UK Dementia Research Institute at UCL , London , WC1N 3BG , U.K
| | - Cynthia Carlsson
- School of Medicine and Public Health , University of Wisconsin , Madison , Wisconsin 53705 , United States
| | - Ozioma Okonkwo
- School of Medicine and Public Health , University of Wisconsin , Madison , Wisconsin 53705 , United States
| | - Lingjun Li
- School of Pharmacy , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
- Department of Chemistry , University of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW The aim of this review is to summarize current conceptual models of cognitive reserve (CR) and related concepts and to discuss evidence for these concepts within the context of aging and Alzheimer's disease. RECENT FINDINGS Evidence to date supports the notion that higher levels of CR, as measured by proxy variables reflective of lifetime experiences, are associated with better cognitive performance, and with a reduced risk of incident mild cognitive impairment/dementia. However, the impact of CR on longitudinal cognitive trajectories is unclear and may be influenced by a number of factors. Although there is promising evidence that some proxy measures of CR may influence structural brain measures, more research is needed. The protective effects of CR may provide an important mechanism for preserving cognitive function and cognitive well-being with age, in part because it can be enhanced throughout the lifespan. However, more research on the mechanisms by which CR is protective is needed.
Collapse
Affiliation(s)
- Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, 1620 McElderry St., Reed Hall 1-West, Baltimore, MD, 21205, USA
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, 1620 McElderry St., Reed Hall 1-West, Baltimore, MD, 21205, USA.
| |
Collapse
|
45
|
Zhong X, Wang J, Carlsson C, Okonkwo O, Zetterberg H, Li L. A Strategy for Discovery and Verification of Candidate Biomarkers in Cerebrospinal Fluid of Preclinical Alzheimer's Disease. Front Mol Neurosci 2019; 11:483. [PMID: 30666187 PMCID: PMC6330998 DOI: 10.3389/fnmol.2018.00483] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disease, is characterized by the accumulation of senile plaques, neurofibrillary tangles, and loss of synapses and neurons in the brain. The pathophysiological process of AD begins with a long asymptomatic phase, which provides a potential opportunity for early therapeutic intervention. Therefore, it is crucial to define putative biomarkers via reliable and validated methods for early diagnosis of AD. Here, we characterized candidate biomarkers by discovery proteomics analysis of cerebrospinal fluid (CSF), revealing that 732 and 704 proteins with more than one unique peptide were identified in healthy controls and preclinical AD patients, respectively. Among them, 79 and 98 proteins were significantly altered in preclinical AD for women and men, respectively, many of which have been demonstrated with consistent regulation pattern in patients with mild cognitive impairment or AD dementia. In-house developed 5-plex isotopic N,N-dimethyl leucine (iDiLeu) tags were further utilized to verify candidate biomarkers, neurosecretory protein VGF (VGF) and apolipoprotein E (apoE). By labeling peptide standards with different iDiLeu tags, a four-point internal calibration curve was constructed to allow for determination of the absolute amount of target analytes in CSF through a single liquid chromatography-mass spectrometry run.
Collapse
Affiliation(s)
- Xiaofang Zhong
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States
| | - Jingxin Wang
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Cynthia Carlsson
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ozioma Okonkwo
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
46
|
Lesuis SL, Hoeijmakers L, Korosi A, de Rooij SR, Swaab DF, Kessels HW, Lucassen PJ, Krugers HJ. Vulnerability and resilience to Alzheimer's disease: early life conditions modulate neuropathology and determine cognitive reserve. Alzheimers Res Ther 2018; 10:95. [PMID: 30227888 PMCID: PMC6145191 DOI: 10.1186/s13195-018-0422-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/15/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder with a high prevalence among the elderly and a huge personal and societal impact. Recent epidemiological studies have indicated that the incidence and age of onset of sporadic AD can be modified by lifestyle factors such as education, exercise, and (early) stress exposure. Early life adversity is known to promote cognitive decline at a later age and to accelerate aging, which are both primary risk factors for AD. In rodent models, exposure to 'negative' or 'positive' early life experiences was recently found to modulate various measures of AD neuropathology, such as amyloid-beta levels and cognition at later ages. Although there is emerging interest in understanding whether experiences during early postnatal life also modulate AD risk in humans, the mechanisms and possible substrates underlying these long-lasting effects remain elusive. METHODS We review literature and discuss the role of early life experiences in determining later age and AD-related processes from a brain and cognitive 'reserve' perspective. We focus on rodent studies and the identification of possible early determinants of later AD vulnerability or resilience in relation to early life adversity/enrichment. RESULTS Potential substrates and mediators of early life experiences that may influence the development of AD pathology and cognitive decline are: programming of the hypothalamic-pituitary-adrenal axis, priming of the neuroinflammatory response, dendritic and synaptic complexity and function, overall brain plasticity, and proteins such as early growth response protein 1 (EGR1), activity regulated cytoskeleton-associated protein (Arc), and repressor element-1 silencing transcription factor (REST). CONCLUSIONS We conclude from these rodent studies that the early postnatal period is an important and sensitive phase that influences the vulnerability to develop AD pathology. Yet translational studies are required to investigate whether early life experiences also modify AD development in human studies, and whether similar molecular mediators can be identified in the sensitivity to develop AD in humans.
Collapse
Affiliation(s)
- Sylvie L. Lesuis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Lianne Hoeijmakers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Aniko Korosi
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Susanne R. de Rooij
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
- Department of Clinical Epidemiology, Biostatistics & Bio informatics, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Dick F. Swaab
- The Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Helmut W. Kessels
- The Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
- Department of Cellular and Computational Neuroscience, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Paul J. Lucassen
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
47
|
Racial and Ethnic Disparities in Dementia Risk Among Individuals With Low Education. Am J Geriatr Psychiatry 2018; 26:966-976. [PMID: 30005921 DOI: 10.1016/j.jagp.2018.05.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 11/23/2022]
Abstract
INTRODUCTION As higher dementia prevalence in ethnic minority groups could be attributed to low education, we studied individuals with low education and explored potential factors driving dementia disparities. METHODS We examined differences in dementia risk between low-educated non-Hispanic whites, Hispanics, and African Americans, and the impact of lifetime risk factors using data from the nationally representative Aging, Demographics, and Memory Study (N = 819). RESULTS As indicated by Cox regression modeling, dementia risk of low-educated individuals was not significantly different between ethnic groups but was related to having an APOE e4 allele (hazard ratio [HR] 1.89), depression (HR 1.67), stroke (HR 1.60), and smoking (HR 1.32). Further, even in people with low education, every additional year of education decreased dementia risk (HR 0.95). DISCUSSION Our findings imply that higher dementia prevalence in ethnic minorities may be attributable to low education, especially among Hispanics, in addition to other risk factors.
Collapse
|
48
|
Weiler M, Casseb RF, de Campos BM, de Ligo Teixeira CV, Carletti-Cassani AFMK, Vicentini JE, Magalhães TNC, de Almeira DQ, Talib LL, Forlenza OV, Balthazar MLF, Castellano G. Cognitive Reserve Relates to Functional Network Efficiency in Alzheimer's Disease. Front Aging Neurosci 2018; 10:255. [PMID: 30186154 PMCID: PMC6111617 DOI: 10.3389/fnagi.2018.00255] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 08/02/2018] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia, with no means of cure or prevention. The presence of abnormal disease-related proteins in the population is, in turn, much more common than the incidence of dementia. In this context, the cognitive reserve (CR) hypothesis has been proposed to explain the discontinuity between pathophysiological and clinical expression of AD, suggesting that CR mitigates the effects of pathology on clinical expression and cognition. fMRI studies of the human connectome have recently reported that AD patients present diminished functional efficiency in resting-state networks, leading to a loss in information flow and cognitive processing. No study has investigated, however, whether CR modifies the effects of the pathology in functional network efficiency in AD patients. We analyzed the relationship between CR, pathophysiology and network efficiency, and whether CR modifies the relationship between them. Fourteen mild AD, 28 amnestic mild cognitive impairment (aMCI) due to AD, and 28 controls were enrolled. We used education to measure CR, cerebrospinal fluid (CSF) biomarkers to evaluate pathophysiology, and graph metrics to measure network efficiency. We found no relationship between CR and CSF biomarkers; CR was related to higher network efficiency in all groups; and abnormal levels of CSF protein biomarkers were related to more efficient networks in the AD group. Education modified the effects of tau-related pathology in the aMCI and mild AD groups. Although higher CR might not protect individuals from developing AD pathophysiology, AD patients with higher CR are better able to cope with the effects of pathology—presenting more efficient networks despite pathology burden. The present study highlights that interventions focusing on cognitive stimulation might be useful to slow age-related cognitive decline or dementia and lengthen healthy aging.
Collapse
Affiliation(s)
- Marina Weiler
- Neurophysics Group, Institute of Physics Gleb Wataghin, Cosmic Rays and Chronology Department, University of Campinas (UNICAMP), Campinas, Brazil.,Neuroimaging Laboratory, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Raphael Fernandes Casseb
- Neurophysics Group, Institute of Physics Gleb Wataghin, Cosmic Rays and Chronology Department, University of Campinas (UNICAMP), Campinas, Brazil.,Neuroimaging Laboratory, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Brunno Machado de Campos
- Neuroimaging Laboratory, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | | | | | - Jéssica Elias Vicentini
- Neuroimaging Laboratory, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Débora Queiroz de Almeira
- Neuroimaging Laboratory, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Leda Leme Talib
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Orestes Vicente Forlenza
- Laboratório de Neurociências (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, Brazil
| | | | - Gabriela Castellano
- Neurophysics Group, Institute of Physics Gleb Wataghin, Cosmic Rays and Chronology Department, University of Campinas (UNICAMP), Campinas, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| |
Collapse
|
49
|
Tsai CK, Kao TW, Lee JT, Wang CC, Chou CH, Liang CS, Yang FC, Chen WL. Global-cognitive health metrics: A novel approach for assessing cognition impairment in adult population. PLoS One 2018; 13:e0197691. [PMID: 29813084 PMCID: PMC5973572 DOI: 10.1371/journal.pone.0197691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/07/2018] [Indexed: 12/27/2022] Open
Abstract
Dementia is the supreme worldwide burden for welfare and the health care system in the 21st century. The early identification and control of the modifiable risk factors of dementia are important. Global-cognitive health (GCH) metrics, encompassing controllable cardiovascular health (CVH) and non-CVH risk factors of dementia, is a newly developed approach to assess the risk of cognitive impairment. The components of ideal GCH metrics includes better education, non-obesity, normal blood pressure, no smoking, no depression, ideal physical activity, good social integration, normal glycated hemoglobin (HbA1c), and normal hearing. This study focuses on the association between ideal GCH metrics and the cognitive function in young adults by investigating the Third Health and Nutrition Examination Survey (NHANES III) database, which has not been reported previously. A total of 1243 participants aged 17 to 39 years were recruited in this study. Cognitive functioning was evaluated by the simple reaction time test (SRTT), symbol-digit substitution test (SDST), and serial digit learning test (SDLT). Participants with significantly higher scores of GCH metrics had better cognitive performance (p for trend <0.01 in three cognitive tests). Moreover, better education, ideal physical activity, good social integration and normal glycated hemoglobin were the optimistic components of ideal GCH metrics associated with better cognitive performance after adjusting for covariates (p < 0.05 in three cognitive tests). These findings emphasize the importance of a preventive strategy for modifiable dementia risk factors to enhance cognitive functioning during adulthood.
Collapse
Affiliation(s)
- Chia-Kuang Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Tung-Wei Kao
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital and School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Jiunn-Tay Lee
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chung-Ching Wang
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital and School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Chung-Hsing Chou
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Sung Liang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital and School of Medicine, National Defense Medical Center, Taipei, Taiwan
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Fu-Chi Yang
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Liang Chen
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital and School of Medicine, National Defense Medical Center, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
50
|
Ko K, Byun MS, Yi D, Lee JH, Kim CH, Lee DY. Early-Life Cognitive Activity Is Related to Reduced Neurodegeneration in Alzheimer Signature Regions in Late Life. Front Aging Neurosci 2018; 10:70. [PMID: 29623037 PMCID: PMC5875443 DOI: 10.3389/fnagi.2018.00070] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/01/2018] [Indexed: 01/16/2023] Open
Abstract
Background: Although increased cognitive activity (CA), both current and past, is known to be associated with a decreased occurrence of Alzheimer’s disease (AD) dementia in older adults, the exact neural mechanisms underlying the association between CA during different stages of life and human dementia remain unclear. Therefore, we investigated whether CA during different life stages is associated with cerebral amyloid-beta (Aβ) pathology and AD-related neurodegeneration in non-demented older adults. Methods: Cross-sectional analyses of data collected between April 2014 and March 2016 from the Korean Brain Aging Study for Early Diagnosis and Prediction of Alzheimer’s Disease (KBASE), an ongoing prospective cohort. In total, 321 community-dwelling, non-demented older adults were involved in this study. Cerebral Aβ deposition and Aβ positivity were measured using 11C-Pittsburgh compound B (PiB)-positron emission tomography (PET). AD-signature region cerebral glucose metabolism (AD-CMglu) and AD-signature region neurodegeneration (AD-ND) positivity were measured using 18F-fluorodeoxyglucose (FDG)-PET. In addition, CA in early, mid, and late life was systematically evaluated using a structured questionnaire. Results: Of the 321 participants, 254 were cognitively normal (CN) and 67 had mild cognitive impairment (MCI). The mean age of participants was 69.6 years old [standard deviation (SD) = 8.0]. Higher early-life CA (CAearly) was associated with significantly increased AD-CMglu (B = 0.035, SE = 0.013, P = 0.009) and a decreasing trend of AD-ND positivity (OR = 0.65, 95% CI 0.43–0.98, P = 0.04) but was not associated with Aβ deposition or positivity. We observed no association between midlife CA (CAmid) and any AD-related brain changes. Late-life CA (CAlate) showed an association with both global Aβ deposition and AD-CMglu, although it was not statistically significant. Sensitivity analyses controlling for current depression or conducted only for CN individuals revealed similar results. Conclusion: Our results suggest that CA in early life may be protective against late-life AD-related neurodegeneration, independently of cerebral Aβ pathology.
Collapse
Affiliation(s)
- Kang Ko
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea.,Department of Psychiatry, Yonsei University College of Medicine, Seoul, South Korea
| | - Min Soo Byun
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Jun Ho Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| | - Chan Hyung Kim
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, South Korea.,Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea.,Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, South Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|