1
|
Sannemann L, Bartels C, Brosseron F, Buerger K, Fliessbach K, Freiesleben SD, Frommann I, Glanz W, Heneka MT, Janowitz D, Kilimann I, Kleineidam L, Lammerding D, Laske C, Munk MHJ, Perneczky R, Peters O, Priller J, Rauchmann BS, Rostamzadeh A, Roy-Kluth N, Schild AK, Schneider A, Schneider LS, Spottke A, Spruth EJ, Teipel S, Wagner M, Wiltfang J, Wolfsgruber S, Duezel E, Jessen F. Symptomatic Clusters Related to Amyloid Positivity in Cognitively Unimpaired Individuals. J Alzheimers Dis 2024; 100:193-205. [PMID: 38848176 DOI: 10.3233/jad-231335] [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: 06/09/2024]
Abstract
Background The NIA-AA Research Framework on Alzheimer's disease (AD) proposes a transitional stage (stage 2) characterized by subtle cognitive decline, subjective cognitive decline (SCD) and mild neurobehavioral symptoms (NPS). Objective To identify participant clusters based on stage 2 features and assess their association with amyloid positivity in cognitively unimpaired individuals. Methods We included baseline data of N = 338 cognitively unimpaired participants from the DELCODE cohort with data on cerebrospinal fluid biomarkers for AD. Classification into the AD continuum (i.e., amyloid positivity, A+) was based on Aβ42/40 status. Neuropsychological test data were used to assess subtle objective cognitive dysfunction (OBJ), the subjective cognitive decline interview (SCD-I) was used to detect SCD, and the Neuropsychiatric Inventory Questionnaire (NPI-Q) was used to assess NPS. A two-step cluster analysis was carried out and differences in AD biomarkers between clusters were analyzed. Results We identified three distinct participant clusters based on presented symptoms. The highest rate of A+ participants (47.6%) was found in a cluster characterized by both OBJ and SCD. A cluster of participants that presented with SCD and NPS (A+:26.6%) and a cluster of participants with overall few symptoms (A+:19.7%) showed amyloid positivity in a range that was not higher than the expected A+ rate for the age group. Across the full sample, participants with a combination of SCD and OBJ in the memory domain showed a lower Aβ42/ptau181 ratio compared to those with neither SCD nor OBJ. Conclusions The cluster characterized by participants with OBJ and concomitant SCD was enriched for amyloid pathology.
Collapse
Affiliation(s)
- Lena Sannemann
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
| | - Claudia Bartels
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Göttingen, Germany
| | | | - Katharina Buerger
- German Center for Neurodegenerative Diseases - DZNE, Munich, Germany
- Institute for Stroke and Dementia Research - ISD, University Hospital, LMU Munich, Munich, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases - DZNE, Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Silka Dawn Freiesleben
- German Center for Neurodegenerative Diseases - DZNE, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin-Department of Psychiatry and Neurosciences, Berlin, Germany
| | - Ingo Frommann
- German Center for Neurodegenerative Diseases - DZNE, Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases - DZNE, Magdeburg, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine - LCSB, University of Luxembourg, Belvaux, Luxembourg
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research - ISD, University Hospital, LMU Munich, Munich, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases - DZNE, Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases - DZNE, Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Dominik Lammerding
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin-Department of Psychiatry and Neurosciences, Berlin, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases - DZNE, Tübingen, Germany
- Department of Psychiatry and Psychotherapy, Section for Dementia Research, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Matthias H J Munk
- German Center for Neurodegenerative Diseases - DZNE, Tübingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases - DZNE, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology - SyNergy, Munich, Munich, Germany
- Ageing Epidemiology Research Unit - AGE, School of Public Health, Imperial College London, London, UK
| | - Oliver Peters
- German Center for Neurodegenerative Diseases - DZNE, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin-Department of Psychiatry and Neurosciences, Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases - DZNE, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, Berlin, Germany
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Sheffield Institute for Translational Neuroscience - SITraN, University of Sheffield, Sheffield, UK
- Department of Neuroradiology, University Hospital LMU, Munich, Germany
| | - Ayda Rostamzadeh
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
| | - Nina Roy-Kluth
- German Center for Neurodegenerative Diseases - DZNE, Bonn, Germany
| | - Ann-Katrin Schild
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases - DZNE, Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Luisa-Sophie Schneider
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin-Department of Psychiatry and Neurosciences, Berlin, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases - DZNE, Bonn, Germany
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases - DZNE, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, Berlin, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases - DZNE, Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases - DZNE, Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases - DZNE, Göttingen, Germany
- Department of Medical Sciences, Neurosciences and Signaling Group, Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases - DZNE, Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Emrah Duezel
- German Center for Neurodegenerative Diseases - DZNE, Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research - IKND, Otto-von-Guericke University, Magdeburg, Germany
| | - Frank Jessen
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
- German Center for Neurodegenerative Diseases - DZNE, Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases - CECAD, University of Cologne, Cologne, Germany
| |
Collapse
|
2
|
Gauthreaux K, Mock C, Teylan MA, Culhane JE, Chen YC, Chan KCG, Katsumata Y, Nelson PT, Kukull WA. Symptomatic Profile and Cognitive Performance in Autopsy-Confirmed Limbic-Predominant Age-Related TDP-43 Encephalopathy With Comorbid Alzheimer Disease. J Neuropathol Exp Neurol 2022; 81:975-987. [PMID: 36264254 PMCID: PMC9677237 DOI: 10.1093/jnen/nlac093] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Transactive response DNA-binding protein 43 kDa (TDP-43) proteinopathy is the hallmark of limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC). LATE-NC is a common copathology with Alzheimer disease neuropathologic change (ADNC). Data from the National Alzheimer's Coordinating Center were analyzed to compare clinical features and copathologies of autopsy-confirmed ADNC with versus without comorbid LATE-NC. A total of 735 participants with ADNC alone and 365 with ADNC with LATE-NC were included. Consistent with prior work, brains with LATE-NC had more severe ADNC, more hippocampal sclerosis, and more brain arteriolosclerosis copathologies. Behavioral symptoms and cognitive performance on neuropsychological tests were compared, stratified by ADNC severity (low/intermediate vs high). Participants with ADNC and LATE-NC were older, had higher ADNC burden, and had worse cognitive performance than participants with ADNC alone. In the low/intermediate ADNC strata, participants with comorbid LATE-NC had higher prevalence of behavioral symptoms (apathy, disinhibition, agitation, personality change). They also had worsened performance in episodic memory and language/semantic memory. Differences narrowed in the high ADNC strata, with worsened performance in only episodic memory in the comorbid LATE-NC group. The co-occurrence of LATE-NC with ADNC is associated with a different pattern of behavioral and cognitive performance than ADNC alone, particularly in people with low/intermediate ADNC burden.
Collapse
Affiliation(s)
- Kathryn Gauthreaux
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
| | - Charles Mock
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
| | - Merilee A Teylan
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
| | - Jessica E Culhane
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
| | - Yen-Chi Chen
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Kwun C G Chan
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA
| | - Walter A Kukull
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
| |
Collapse
|
3
|
Zimmerman SC, Brenowitz WD, Calmasini C, Ackley SF, Graff RE, Asiimwe SB, Staffaroni AM, Hoffmann TJ, Glymour MM. Association of Genetic Variants Linked to Late-Onset Alzheimer Disease With Cognitive Test Performance by Midlife. JAMA Netw Open 2022; 5:e225491. [PMID: 35377426 PMCID: PMC8980909 DOI: 10.1001/jamanetworkopen.2022.5491] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
IMPORTANCE Identifying the youngest age when Alzheimer disease (AD) influences cognition and the earliest affected cognitive domains will improve understanding of the natural history of AD and approaches to early diagnosis. OBJECTIVE To evaluate the age at which cognitive differences between individuals with higher compared with lower genetic risk of AD are first apparent and which cognitive assessments show the earliest difference. DESIGN, SETTING, AND PARTICIPANTS This cross-sectional study used data from UK Biobank participants of European genetic ancestry, aged 40 years or older, who contributed genotypic and cognitive test data from January 1, 2006, to December 31, 2015. Data analysis was performed from March 10, 2020, to January 4, 2022. EXPOSURE The AD genetic risk score (GRS), which is a weighted sum of 23 single-nucleotide variations. MAIN OUTCOMES AND MEASURES Seven cognitive tests were administered via touchscreen at in-person visits or online. Cognitive domains assessed included fluid intelligence, episodic memory, processing speed, executive functioning, and prospective memory. Multiple cognitive measures were derived from some tests, yielding 32 separate measures. Interactions between age and AD-GRS for each of the 32 cognitive measures were tested with linear regression using a Bonferroni-corrected P value threshold. For cognitive measures with significant evidence of age by AD-GRS interaction, the youngest age of interaction was assessed with new regression models, with nonlinear specification of age terms. Models with youngest age of interaction from 40 to 70 years, in 1-year increments, were compared, and the best-fitting model for each cognitive measure was chosen. Results across cognitive measures were compared to determine which cognitive indicators showed earliest AD-related change. RESULTS A total of 405 050 participants (mean [SD] age, 57.1 [7.9] years; 54.1% female) were included. Sample sizes differed across cognitive tests (from 12 455 to 404 682 participants). The AD-GRS significantly modified the association with age on 13 measures derived from the pairs matching (range in difference in mean cognition per decade increase in age for 1-SD higher AD-GRS, 2.5%-11.5%), symbol digit substitution (range in difference in mean cognition per decade increase in age for 1-SD higher AD-GRS, 2.0%-5.8%), and numeric memory tests (difference in mean cognition per decade increase in age for 1-SD higher AD-GRS, 8.8%) (P = 1.56 × 10-3). Best-fitting models suggested that cognitive scores of individuals with a high vs low AD-GRS began to diverge by 56 years of age for all 13 measures and by 47 years of age for 9 measures. CONCLUSIONS AND RELEVANCE In this cross-sectional study, by early midlife, subtle differences in memory and attention were detectable among individuals with higher genetic risk of AD.
Collapse
Affiliation(s)
- Scott C. Zimmerman
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Willa D. Brenowitz
- Department of Epidemiology and Biostatistics, University of California, San Francisco
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
| | - Camilla Calmasini
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Sarah F. Ackley
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Rebecca E. Graff
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Stephen B. Asiimwe
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Adam M. Staffaroni
- Weill Institute for Neurosciences, Department of Neurology, Memory and Aging Center, University of California, San Francisco
| | - Thomas J. Hoffmann
- Department of Epidemiology and Biostatistics, University of California, San Francisco
- Institute for Human Genetics, University of California, San Francisco
| | - M. Maria Glymour
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| |
Collapse
|
4
|
Ali DG, Bahrani AA, Barber JM, El Khouli RH, Gold BT, Harp JP, Jiang Y, Wilcock DM, Jicha GA. Amyloid-PET Levels in the Precuneus and Posterior Cingulate Cortices Are Associated with Executive Function Scores in Preclinical Alzheimer's Disease Prior to Overt Global Amyloid Positivity. J Alzheimers Dis 2022; 88:1127-1135. [PMID: 35754276 PMCID: PMC10349398 DOI: 10.3233/jad-220294] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Global amyloid-β (Aβ) deposition in the brain can be quantified by Aβ-PET scans to support or refute a diagnosis of preclinical Alzheimer's disease (pAD). Yet, Aβ-PET scans enable quantitative evaluation of regional Aβ elevations in pAD, potentially allowing even earlier detection of pAD, long before global positivity is achieved. It remains unclear as to whether such regional changes are clinically meaningful. OBJECTIVE Test the hypothesis that early focal regional amyloid deposition in the brain is associated with cognitive performance in specific cognitive domain scores in pAD. METHODS Global and regional standardized uptake value ratios (SUVr) from 18F-florbetapir PET/CT scanning were determined using the Siemens Syngo.via® Neurology software package across a sample of 99 clinically normal participants with Montreal Cognitive Assessment (MoCA) scores≥23. Relationships between regional SUVr and cognitive test scores were analyzed using linear regression models adjusted for age, sex, and education. Participants were divided into two groups based on SUVr in the posterior cingulate and precuneus gyri (SUVR≥1.17). Between group differences in cognitive test scores were analyzed using ANCOVA models. RESULTS Executive function performance was associated with increased regional SUVr in the precuneus and posterior cingulate regions only (p < 0.05). There were no significant associations between memory and Aβ-PET SUVr in any regions of the brain. CONCLUSION These data demonstrate that increased Aβ deposition in the precuneus and posterior cingulate (the earliest brain regions affected with Aβ pathology) is associated with changes in executive function that may precede memory decline in pAD.
Collapse
Affiliation(s)
- Doaa G. Ali
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
- Department of Behavioral Science, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| | - Ahmed A. Bahrani
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| | - Justin M. Barber
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| | - Riham H. El Khouli
- Department of Radiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Brian T. Gold
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jordan P. Harp
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| | - Yang Jiang
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
- Department of Behavioral Science, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| | - Donna M. Wilcock
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - Gregory A. Jicha
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
- Department of Behavioral Science, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| |
Collapse
|
5
|
Appiah F, Charnigo RJ. A Comparison of Methods for Predicting Future Cognitive Status: Mixture Modeling, Latent Class Analysis, and Competitors. Alzheimer Dis Assoc Disord 2021; 35:306-314. [PMID: 34224419 PMCID: PMC8605986 DOI: 10.1097/wad.0000000000000462] [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: 08/10/2020] [Accepted: 05/17/2021] [Indexed: 11/25/2022]
Abstract
PURPOSE The present work compares various methods for using baseline cognitive performance data to predict eventual cognitive status of longitudinal study participants at the University of Kentucky's Alzheimer's Disease Center. METHODS Cox proportional hazards models examined time to cognitive transition as predicted by risk strata derived from normal mixture modeling, latent class analysis, and a 1-SD thresholding approach. An additional comparator involved prediction directly from a numeric value for baseline cognitive performance. RESULTS A normal mixture model suggested 3 risk strata based on Consortium to Establish a Registry for Alzheimer's Disease (CERAD) T scores: high, intermediate, and low risk. Cox modeling of time to cognitive decline based on posterior probabilities for risk stratum membership yielded an estimated hazard ratio of 4.00 with 95% confidence interval 1.53-10.44 in comparing high risk membership to low risk; for intermediate risk membership versus low risk, the modeling yielded hazard ratio=2.29 and 95% confidence interval=0.98-5.33. Latent class analysis produced 3 groups, which did not have a clear ordering in terms of risk; however, one group exhibited appreciably greater hazard of cognitive decline. All methods for generating predictors of cognitive transition yielded statistically significant likelihood ratio statistics but modest concordance statistics. CONCLUSION Posterior probabilities from mixture modeling allow for risk stratification that is data-driven and, in the case of CERAD T scores, modestly predictive of later cognitive decline. Incorporating other covariates may enhance predictions.
Collapse
Affiliation(s)
- Frank Appiah
- Program, Management, Analytics and Technology, Greenwood Village, CO
| | - Richard J Charnigo
- Departments of Biostatistics
- Statistics, University of Kentucky, Lexington, KY
| |
Collapse
|
6
|
Teylan MA, Mock C, Gauthreaux K, Culhane JE, Jicha G, Chen YC, Chan KCG, Kukull WA, Nelson PT, Katsumata Y. Differences in Symptomatic Presentation and Cognitive Performance Among Participants With LATE-NC Compared to FTLD-TDP. J Neuropathol Exp Neurol 2021; 80:1024–1032. [PMID: 34597386 DOI: 10.1093/jnen/nlab098] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Transactive response DNA-binding protein 43 kDa (TDP-43) is aberrantly aggregated and phosphorylated in frontotemporal lobar degeneration of the TDP-43 type (FTLD-TDP), and in limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC). We examined data from the National Alzheimer's Coordinating Center to compare clinical features of autopsy-confirmed LATE-NC and FTLD-TDP. A total of 265 LATE-NC and 92 FTLD-TDP participants were included. Cognitive and behavioral symptoms were compared, stratified by level of impairment based on global clinical dementia rating (CDR) score. LATE-NC participants were older at death, more likely to carry APOE ε4, more likely to have Alzheimer disease neuropathology, and had lower (i.e. less severe) final CDR global scores than those with FTLD-TDP. Participants with FTLD-TDP were more likely to present with primary progressive aphasia, or behavior problems such as apathy, disinhibition, and personality changes. Among participants with final CDR score of 2-3, those with LATE-NC were more likely to have visuospatial impairment, delusions, and/or visual hallucinations. These differences were robust after sensitivity analyses excluding older (≥80 years at death), LATE-NC stage 3, or severe Alzheimer cases. Overall, FTLD-TDP was more globally severe, and affected younger participants, whereas psychoses were more common in LATE-NC.
Collapse
Affiliation(s)
- Merilee A Teylan
- From the National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, Washington, USA (MAT, CM, KG, JEC, Y-CC, KCGC, WAK); Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA (GJ, PTN, YK); Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA (PTN); Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA (YK)
| | - Charles Mock
- From the National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, Washington, USA (MAT, CM, KG, JEC, Y-CC, KCGC, WAK); Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA (GJ, PTN, YK); Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA (PTN); Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA (YK)
| | - Kathryn Gauthreaux
- From the National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, Washington, USA (MAT, CM, KG, JEC, Y-CC, KCGC, WAK); Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA (GJ, PTN, YK); Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA (PTN); Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA (YK)
| | - Jessica E Culhane
- From the National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, Washington, USA (MAT, CM, KG, JEC, Y-CC, KCGC, WAK); Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA (GJ, PTN, YK); Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA (PTN); Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA (YK)
| | - Gregory Jicha
- From the National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, Washington, USA (MAT, CM, KG, JEC, Y-CC, KCGC, WAK); Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA (GJ, PTN, YK); Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA (PTN); Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA (YK)
| | - Yen-Chi Chen
- From the National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, Washington, USA (MAT, CM, KG, JEC, Y-CC, KCGC, WAK); Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA (GJ, PTN, YK); Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA (PTN); Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA (YK)
| | - Kwun C G Chan
- From the National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, Washington, USA (MAT, CM, KG, JEC, Y-CC, KCGC, WAK); Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA (GJ, PTN, YK); Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA (PTN); Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA (YK)
| | - Walter A Kukull
- From the National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, Washington, USA (MAT, CM, KG, JEC, Y-CC, KCGC, WAK); Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA (GJ, PTN, YK); Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA (PTN); Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA (YK)
| | - Peter T Nelson
- From the National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, Washington, USA (MAT, CM, KG, JEC, Y-CC, KCGC, WAK); Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA (GJ, PTN, YK); Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA (PTN); Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA (YK)
| | - Yuriko Katsumata
- From the National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, Washington, USA (MAT, CM, KG, JEC, Y-CC, KCGC, WAK); Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA (GJ, PTN, YK); Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA (PTN); Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA (YK)
| |
Collapse
|
7
|
Williams BD, Pendleton N, Chandola T. Does the association between cognition and education differ between older adults with gradual or rapid trajectories of cognitive decline? NEUROPSYCHOLOGY, DEVELOPMENT, AND COGNITION. SECTION B, AGING, NEUROPSYCHOLOGY AND COGNITION 2021; 29:1-21. [PMID: 33683174 DOI: 10.1080/13825585.2021.1889958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 02/09/2021] [Indexed: 10/22/2022]
Abstract
Education is associated with improved baseline cognitive performance in older adults, but the association with maintenance of cognitive function is less clear. Education may be associated with different types of active cognitive reserve in those following different cognitive trajectories. We used data on n = 5642 adults aged >60 from the English Longitudinal Study of Aging (ELSA) over 5 waves (8 years). We used growth mixture models to test if the association between educational attainment and rate of change in verbal fluency or immediate recall varied by latent class trajectory. For recall, 91.5% (n = 5164) of participants were in a gradual decline class and 8.5% (n = 478) in a rapid decline class. For fluency, 90.0% (n = 4907) were in a gradual decline class and 10.0% (n = 561) were in a rapid decline class. Educational attainment was associated with improved baseline performance for both verbal fluency and recall. In the rapidly declining classes, educational attainment was not associated with rate of change for either outcome. In the verbal fluency gradual decline class, education was associated with higher (an additional 0.05-0.38 words per 2 years) or degree level education (an additional 0.04-0.42 words per 2 years) when compared to those with no formal qualifications. We identified no evidence of a protective effect of education against rapid cognitive decline. There was some evidence of active cognitive reserve for verbal fluency but not recall, which may reflect a small degree of domain-specific protection against age-related cognitive decline.
Collapse
Affiliation(s)
| | - Neil Pendleton
- Cathie Marsh Institute for Social Research, University of Manchester, Manchester, UK
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - Tarani Chandola
- Cathie Marsh Institute for Social Research, University of Manchester, Manchester, UK
| |
Collapse
|
8
|
Mock C, Teylan M, Beecham G, Besser L, Cairns NJ, Crary JF, Katsumata Y, Nelson PT, Kukull W. The Utility of the National Alzheimer's Coordinating Center's Database for the Rapid Assessment of Evolving Neuropathologic Conditions. Alzheimer Dis Assoc Disord 2020; 34:105-111. [PMID: 32304374 PMCID: PMC7242145 DOI: 10.1097/wad.0000000000000380] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The field of dementia research is rapidly evolving, especially with regards to our understanding of the diversity of neuropathologic changes that underlie cognitive decline. Definitions and criteria for known conditions are being periodically revised and refined, and new findings are being made about neuropathologic features associated with dementia status. The database maintained by the National Alzheimer's Coordinating Center (NACC) offer researchers a robust, rapid, and statistically well-powered method to evaluate the implications of newly identified neuropathologic conditions with regards to comorbidities, demographic associations, cognitive status, neuropsychologic tests, radiographic findings, and genetics. NACC data derive from dozens of excellent US Alzheimer disease research centers, which collectively follow thousands of research volunteers longitudinally. Many of the research participants are autopsied using state-of-the-art methods. In this article, we describe the NACC database and give examples of its use in evaluating recently revised neuropathologic diagnoses, including primary age-related tauopathy (PART), limbic predominant age-related TDP-43 encephalopathy (LATE), and the preclinical stage of Alzheimer disease neuropathologic change, based on the National Institute on Aging-Alzheimer's Association consensus guidelines. The dementia research community is encouraged to make use of this readily available database as new neuropathologic changes are recognized and defined in this rapidly evolving field.
Collapse
Affiliation(s)
- Charles Mock
- National Alzheimer’s Coordinating Center, University of Washington, WA
| | - Merilee Teylan
- National Alzheimer’s Coordinating Center, University of Washington, WA
| | - Gary Beecham
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL
| | | | - Nigel J. Cairns
- College of Medicine and Health, University of Exeter, Exeter, UK
| | - John F. Crary
- Neuropathology Brain Bank & Research Core, Departments of Pathology & Neuroscience, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY
| | - Peter T. Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY
| | - Walter Kukull
- National Alzheimer’s Coordinating Center, University of Washington, WA
| |
Collapse
|
9
|
Teylan M, Mock C, Gauthreaux K, Chen YC, Chan KCG, Hassenstab J, Besser LM, Kukull WA, Crary JF. Cognitive trajectory in mild cognitive impairment due to primary age-related tauopathy. Brain 2020; 143:611-621. [PMID: 31942622 PMCID: PMC7009602 DOI: 10.1093/brain/awz403] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 11/05/2019] [Accepted: 11/09/2019] [Indexed: 01/18/2023] Open
Abstract
Primary age-related tauopathy is increasingly recognized as a separate neuropathological entity different from Alzheimer's disease. Both share the neuropathological features of tau aggregates and neuronal loss in the temporal lobe, but primary age-related tauopathy lacks the requisite amyloid plaques central to Alzheimer's disease. While both have similar clinical presentations, individuals with symptomatic primary age-related tauopathy are commonly of more advanced ages with milder cognitive dysfunction. Direct comparison of the neuropsychological trajectories of primary age-related tauopathy and Alzheimer's disease has not been thoroughly evaluated and thus, our objective was to determine how cognitive decline differs longitudinally between these two conditions after the onset of clinical symptoms. Data were obtained from the National Alzheimer's Coordinating Center on participants with mild cognitive impairment at baseline and either no neuritic plaques (i.e. primary age-related tauopathy) or moderate to frequent neuritic plaques (i.e. Alzheimer neuropathological change) at subsequent autopsy. For patients with Alzheimer's disease and primary age-related tauopathy, we compared rates of decline in the sum of boxes score from the CDR® Dementia Staging Instrument and in five cognitive domains (episodic memory, attention/working memory, executive function, language/semantic memory, and global composite) using z-scores for neuropsychological tests that were calculated based on scores for participants with normal cognition. The differences in rates of change were tested using linear mixed-effects models accounting for clinical centre clustering and repeated measures by individual. Models were adjusted for sex, age, education, baseline test score, Braak stage, apolipoprotein ε4 (APOE ε4) carrier status, family history of cognitive impairment, and history of stroke, hypertension, or diabetes. We identified 578 participants with a global CDR of 0.5 (i.e. mild cognitive impairment) at baseline, 126 with primary age-related tauopathy and 452 with Alzheimer's disease. Examining the difference in rates of change in CDR sum of boxes and in all domain scores, participants with Alzheimer's disease had a significantly steeper decline after becoming clinically symptomatic than those with primary age-related tauopathy. This remained true after adjusting for covariates. The results of this analysis corroborate previous studies showing that primary age-related tauopathy has slower cognitive decline than Alzheimer's disease across multiple neuropsychological domains, thus adding to the understanding of the neuropsychological burden in primary age-related tauopathy. The study provides further evidence to support the hypothesis that primary age-related tauopathy has distinct neuropathological and clinical features compared to Alzheimer's disease.
Collapse
Affiliation(s)
- Merilee Teylan
- National Alzheimer’s Coordinating Center, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Charles Mock
- National Alzheimer’s Coordinating Center, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Kathryn Gauthreaux
- National Alzheimer’s Coordinating Center, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Yen-Chi Chen
- National Alzheimer’s Coordinating Center, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Kwun C G Chan
- National Alzheimer’s Coordinating Center, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Lilah M Besser
- School of Urban and Regional Planning, Florida Atlantic University, Boca Raton, FL, USA
| | - Walter A Kukull
- National Alzheimer’s Coordinating Center, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - John F Crary
- Neuropathology Brain Bank and Research CoRE, Department of Pathology, Nash Family Department of Neuroscience, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
10
|
Biomarker-Based Signature of Alzheimer's Disease in Pre-MCI Individuals. Brain Sci 2019; 9:brainsci9090213. [PMID: 31450744 PMCID: PMC6769621 DOI: 10.3390/brainsci9090213] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/10/2019] [Accepted: 08/20/2019] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) pathology begins decades before the onset of clinical symptoms. It is recognized as a clinicobiological entity, being detectable in vivo independently of the clinical stage by means of pathophysiological biomarkers. Accordingly, neuropathological studies that were carried out on healthy elderly subjects, with or without subjective experience of cognitive decline, reported evidence of AD pathology in a high proportion of cases. At present, mild cognitive impairment (MCI) represents the only clinically diagnosed pre-dementia stage. Several attempts have been carried out to detect AD as early as possible, when subtle cognitive alterations, still not fulfilling MCI criteria, appear. Importantly, pre-MCI individuals showing the positivity of pathophysiological AD biomarkers show a risk of progression similar to MCI patients. In view of successful treatment with disease modifying agents, in a clinical setting, a timely diagnosis is mandatory. In clinical routine, biomarkers assessment should be taken into consideration whenever a subject with subtle cognitive deficits (pre-MCI), who is aware of his/her decline, requests to know the cause of such disturbances. In this review, we report the available neuropsychological and biomarkers data that characterize the pre-MCI patients, thus proposing pre-MCI as the first clinical manifestation of AD.
Collapse
|
11
|
Martínez-Sánchez F, Meilán JJG, Carro J, Ivanova O. A Prototype for the Voice Analysis Diagnosis of Alzheimer's Disease. J Alzheimers Dis 2019; 64:473-481. [PMID: 29914025 DOI: 10.3233/jad-180037] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Speech variations enable us to map the performance of cognitive processes of syntactic, semantic, phonological, and articulatory planning and execution. Speaking is one of the first functions to be affected by neurodegenerative complaints such as Alzheimer's disease (AD), which makes the speech a highly promising biomarker for detecting the illness before the first preclinical symptoms appear. OBJECTIVE This paper has sought to develop and validate a technological prototype that adopts an automated approach to speech analysis among older people. METHODS It uses a mathematical algorithm based on certain discriminatory variables to estimate the probability of developing AD. RESULTS AND CONCLUSION This device may be used at a preclinical stage by non-expert health professionals to determine the likelihood of the onset of AD.
Collapse
Affiliation(s)
| | | | - Juan Carro
- Psychology Faculty, University of Salamanca, Salamanca, Spain
| | - Olga Ivanova
- Philology Faculty, University of Extremadura, Cáceres, Spain
| |
Collapse
|
12
|
Malek-Ahmadi M, Chen K, Perez SE, He A, Mufson EJ. Cognitive composite score association with Alzheimer's disease plaque and tangle pathology. ALZHEIMERS RESEARCH & THERAPY 2018; 10:90. [PMID: 30205840 PMCID: PMC6134796 DOI: 10.1186/s13195-018-0401-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 07/02/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Cognitive composite scores are used as the primary outcome measures for Alzheimer's disease (AD) prevention trials; however, the extent to which these composite measures correlate with AD pathology has not been fully investigated. Since many on-going AD prevention studies are testing therapies that target either amyloid or tau, we sought to establish an association between a cognitive composite score and the underlying pathology of AD. METHODS Data from 192 older deceased and autopsied persons from the Rush Religious Order Study were used in this study. All participants were classified at their initial evaluations with a clinical diagnosis of no cognitive impairment (NCI). Of these individuals, 105 remained NCI at the time of their death while the remaining 87 progressed to mild cognitive impairment (MCI) or AD. A cognitive composite score composed of eight cognitive tests was used as the outcome measure. Individuals were classified into groups based on Consortium to Establish a Registry for Alzheimer's Disease (CERAD) neuropathological diagnosis and Braak stage. RESULTS The rate of annualized composite score decline was significantly greater for the high CERAD (p < 0.001, d = 0.56) and Braak (p < 0.001, d = 0.55) groups compared with the low CERAD and Braak groups, respectively. Mixed-model repeated measure (MMRM) analyses revealed a significantly greater difference in composite score change from baseline for the high CERAD group relative to the low CERAD group after 5 years (Δ = -2.74, 95% confidence interval (CI) -5.01 to -0.47; p = 0.02). A similar analysis between low and high Braak stage groups found no significant difference in change from baseline (Δ = -0.69, 95% CI -3.03 to 1.66; p = 0.56). CONCLUSIONS These data provide evidence that decreased cognitive composite scores were significantly associated with increased AD pathology and provide support for the use of cognitive composite scores in AD prevention trials.
Collapse
Affiliation(s)
| | - Kewei Chen
- Banner Alzheimer's Institute, 901 E. Willetta St, Phoenix, AZ, USA
| | - Sylvia E Perez
- Department of Neurobiology and Neurology, Barrow Neurological Institute, 350 W. Thomas Rd, Phoenix, AZ, 85013, USA
| | - Anna He
- Banner Alzheimer's Institute, 901 E. Willetta St, Phoenix, AZ, USA
| | - Elliott J Mufson
- Department of Neurobiology and Neurology, Barrow Neurological Institute, 350 W. Thomas Rd, Phoenix, AZ, 85013, USA.
| |
Collapse
|
13
|
Malek-Ahmadi M, Mufson EJ, Perez SE, Chen K. Statistical considerations for assessing cognition and neuropathology associations in preclinical Alzheimer's disease. ACTA ACUST UNITED AC 2017. [DOI: 10.1080/24709360.2017.1342186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
| | - Elliott J. Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Sylvia E. Perez
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| |
Collapse
|
14
|
Brayet P, Petit D, Baril AA, Gosselin N, Gagnon JF, Soucy JP, Gauthier S, Kergoat MJ, Carrier J, Rouleau I, Montplaisir J. Brain perfusion during rapid-eye-movement sleep successfully identifies amnestic mild cognitive impairment. Sleep Med 2017; 34:134-140. [PMID: 28522082 DOI: 10.1016/j.sleep.2017.01.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/20/2017] [Accepted: 01/24/2017] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Prodromal markers of Alzheimer's disease (AD) have been derived from wakefulness. However, brain perfusion during rapid-eye movement (REM) sleep could be a sensitive marker of amnestic mild cognitive impairment (aMCI), as activation of REM sleep relies more on the cholinergic system. METHODS Eight subjects with aMCI, and 16 controls, underwent two single-photon emission computed tomography (SPECT) scans with tracer injected during REM sleep then wakefulness. RESULTS Perfusion in the anterior cingulate cortex was significantly decreased in aMCI cases compared to controls for both conditions. That defect was much larger and more severe in REM sleep (1795 voxels) compared to wakefulness (398 voxels), and extended to the middle cingulate cortex and the olfactory cortex. Hypoperfusion in the anterior cingulate cortex during REM sleep allowed better classification than hypoperfusion found in wakefulness (93.8 vs 81.3%). CONCLUSION REM sleep imaging is a valuable tool with which to identify individuals at risk of developing AD.
Collapse
Affiliation(s)
- Pauline Brayet
- Department of Psychology, Université du Québec à Montréal, Montréal, Canada; Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, Canada
| | - Dominique Petit
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, Canada
| | - Andrée-Ann Baril
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, Canada; Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Nadia Gosselin
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, Canada; Department of Psychology, Université de Montréal, Montréal, Canada
| | - Jean-François Gagnon
- Department of Psychology, Université du Québec à Montréal, Montréal, Canada; Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, Canada
| | - Jean-Paul Soucy
- Montreal Neurological Institute, McGill University, Montréal, Canada; PERFORM Centre, Concordia University, Montréal, Canada
| | - Serge Gauthier
- McGill University Research Centre for Studies in Aging, Douglas Hospital, Verdun, Canada
| | - Marie-Jeanne Kergoat
- Faculty of Medicine, Université de Montréal, Montréal, Canada; Research Center, Institut universitaire de gériatrie de Montréal, Montreal, Canada
| | - Julie Carrier
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, Canada; Department of Psychology, Université de Montréal, Montréal, Canada
| | - Isabelle Rouleau
- Department of Psychology, Université du Québec à Montréal, Montréal, Canada; Neurology Service, Hôpital Notre-Dame du CHUM, Montréal, Canada
| | - Jacques Montplaisir
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, Canada; Faculty of Medicine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
15
|
The Differential Effects of Alzheimer's Disease and Lewy Body Pathology on Cognitive Performance: a Meta-analysis. Neuropsychol Rev 2016; 27:1-17. [PMID: 27878426 DOI: 10.1007/s11065-016-9334-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 09/29/2016] [Indexed: 10/20/2022]
Abstract
Differential diagnosis of Alzheimer's disease (AD) from normal aging and other dementia etiologies is imperative for disease specific treatment options and long-term care planning. Neuropathological confirmation is the gold standard for neurodegenerative disease diagnosis, yet most published studies examining the use of neuropsychological tests in the differential diagnosis of dementia rely upon clinical diagnostic outcomes. The present study undertook a meta-analytic review of the literature to identify cognitive tests and domains that allow for the differentiation of individuals with AD pathology from individuals with dementia with Lewy Bodies (DLB) pathology and pathology-free individuals. A comprehensive literature search yielded 14 studies that met the inclusion criteria for the present meta-analysis. Six studies comprised 222 decedents with AD compared to 433 normal controls, and eight studies comprised 431 cases of AD compared to 155 decedents with DLB. Results revealed that the effect of having neuropathologically confirmed AD versus DLB lowered performance in the memory domain, and having DLB decreased performance in the visuospatial domain. No single test differed significantly across the AD and DLB groups. For the AD and pathology free comparison, results indicated that that AD was associated with poorer performance on the memory and language domains. With respect to specific cognitive tests, AD produced lower scores on list learning tests, category fluency, and the Digit Symbol substitution test. The limited number of studies meeting inclusion criteria warrants formulation of guidelines for reporting in clinico-pathological studies; suggested guidelines are provided.
Collapse
|
16
|
Baker JE, Lim YY, Pietrzak RH, Hassenstab J, Snyder PJ, Masters CL, Maruff P. Cognitive impairment and decline in cognitively normal older adults with high amyloid-β: A meta-analysis. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2016; 6:108-121. [PMID: 28239636 PMCID: PMC5315443 DOI: 10.1016/j.dadm.2016.09.002] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION This meta-analysis aimed to characterize the nature and magnitude of amyloid (Aβ)-related cognitive impairment and decline in cognitively normal (CN) older individuals. METHOD MEDLINE Ovid was searched from 2012 to June 2016 for studies reporting relationships between cerebrospinal fluid or positron emission tomography (PET) Aβ levels and cognitive impairment (cross-sectional) and decline (longitudinal) in CN older adults. Neuropsychological data were classified into domains of episodic memory, executive function, working memory, processing speed, visuospatial function, semantic memory, and global cognition. Type of Aβ measure, how Aβ burden was analyzed, inclusion of control variables, and clinical criteria used to exclude participants, were considered as moderators. Random-effects models were used for analyses with effect sizes expressed as Cohen's d. RESULTS A total of 38 studies met inclusion criteria contributing 30 cross-sectional (N = 5005) and 14 longitudinal (N = 2584) samples. Aβ-related cognitive impairment was observed for global cognition (d = 0.32), visuospatial function (d = 0.25), processing speed (d = 0.18), episodic memory, and executive function (both d's = 0.15), with decline observed for global cognition (d = 0.30), semantic memory (d = 0.28), visuospatial function (d = 0.25), and episodic memory (d = 0.24). Aβ-related impairment was moderated by age, amyloid measure, type of analysis, and inclusion of control variables and decline moderated by amyloid measure, type of analysis, inclusion of control variables, and exclusion criteria used. DISCUSSION CN older adults with high Aβ show a small general cognitive impairment and small to moderate decline in episodic memory, visuospatial function, semantic memory, and global cognition.
Collapse
Affiliation(s)
- Jenalle E. Baker
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- CRC for Mental Health, Carlton South, Victoria, Australia
| | - Yen Ying Lim
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Cogstate Ltd., Melbourne, Victoria, Australia
| | - Robert H. Pietrzak
- U.S. Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Peter J. Snyder
- Department of Neurology, Warren Alpert School of Medicine, Brown University, Providence, RI, USA
- Department of Neurology, Rhode Island Hospital & Alpert Medical School of Brown University, Providence, RI, USA
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Paul Maruff
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- CRC for Mental Health, Carlton South, Victoria, Australia
- Cogstate Ltd., Melbourne, Victoria, Australia
| |
Collapse
|
17
|
Ritchie K, Ropacki M, Albala B, Harrison J, Kaye J, Kramer J, Randolph C, Ritchie CW. Recommended cognitive outcomes in preclinical Alzheimer's disease: Consensus statement from the European Prevention of Alzheimer's Dementia project. Alzheimers Dement 2016; 13:186-195. [PMID: 27702619 DOI: 10.1016/j.jalz.2016.07.154] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/23/2016] [Accepted: 07/22/2016] [Indexed: 10/20/2022]
Abstract
The Horizon 2020/IMI European Prevention of Alzheimer's Dementia (EPAD) project will undertake large-scale proof-of-concept trials in predementia Alzheimer's disease (AD). Within EPAD, the monitoring of cognitive trajectories in the preclinical period will constitute a central outcome measure; however, there are currently no clear guidelines as to how this should be achieved as most measures have been developed for the period around dementia diagnosis. The EPAD Scientific Advisory Group for Clinical and Cognitive Outcomes identified appropriate cognitive measures based on a literature search covering both cognitive correlates of preclinical brain changes from imaging studies and cognitive changes observed over time in nondementia population cohorts developing incident dementia. These measures were evaluated according to the following criteria: validity, coherence with biomarker changes, psychometric properties, cross-cultural suitability, availability of alternative forms, and normative data limited practice effects. The resulting consensus statement provides recommendations for both future drug trials and research into preclinical Alzheimer's disease.
Collapse
Affiliation(s)
- Karen Ritchie
- Institut National de la Sante et de la Recherche Medicale, U1061 Neuropsychiatrie, Montpellier, France; University of Montpellier, Montpellier, France; Centre for Dementia Prevention, University of Edinburgh, Edinburgh, UK.
| | - Michael Ropacki
- Clinical Research, Neurosciences, Janssen, South San Francisco, CA, USA
| | - Bruce Albala
- Clinical Research, Neuroscience and General Medicine, Eisai Inc, Woodcliff Lake, NJ, USA
| | - John Harrison
- Metis Cognition Ltd, Kilmington Common, UK; Alzheimer Center VUmc, Amsterdam, Holland
| | - Jeffrey Kaye
- Neurology and Biomedical Engineering, Oregon Health and Science University, Portand, OR, USA
| | - Joel Kramer
- Department of Neurology Memory and Ageing Centre, University of California at San Francisco, San Francisco, CA, USA
| | | | - Craig W Ritchie
- Centre for Dementia Prevention, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
18
|
Detecting cognitive changes in preclinical Alzheimer's disease: A review of its feasibility. Alzheimers Dement 2016; 13:468-492. [PMID: 27702618 DOI: 10.1016/j.jalz.2016.06.2365] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/31/2016] [Accepted: 06/18/2016] [Indexed: 11/23/2022]
Abstract
Significant progress has been made in characterizing the biological changes occurring in preclinical Alzheimer's disease (AD). Cognitive dysfunction has been viewed, however, as a late-stage phenomenon, despite increasing evidence that changes may be detected in the decades preceding dementia. In the absence of comprehensive evidence-based guidelines for preclinical cognitive assessment, longitudinal cohort and neuroimaging studies have been reviewed to determine the temporal order and brain biomarker correlates of specific cognitive functions. Episodic memory decline was observed to be the most salient cognitive function, correlating with high levels of amyloid deposition and hypoconnectivity across large-scale brain networks. Prospective studies point to early decline in both episodic and semantic memory processing as well as executive functions in the predementia period. The cognitive tests have, however, been principally those used to diagnose dementia. New procedures are required which target more finely the medial temporal lobe subregions first affected by clinically silent AD pathology.
Collapse
|
19
|
Three Dimensional Human Neuro-Spheroid Model of Alzheimer's Disease Based on Differentiated Induced Pluripotent Stem Cells. PLoS One 2016; 11:e0163072. [PMID: 27684569 PMCID: PMC5042502 DOI: 10.1371/journal.pone.0163072] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/01/2016] [Indexed: 12/15/2022] Open
Abstract
The testing of candidate drugs to slow progression of Alzheimer’s disease (AD) requires clinical trials that are lengthy and expensive. Efforts to model the biochemical milieu of the AD brain may be greatly facilitated by combining two cutting edge technologies to generate three-dimensional (3D) human neuro-spheroid from induced pluripotent stem cells (iPSC) derived from AD subjects. We created iPSC from blood cells of five AD patients and differentiated them into 3D human neuronal culture. We characterized neuronal markers of our 3D neurons by immunocytochemical staining to validate the differentiation status. To block the generation of pathologic amyloid β peptides (Aβ), the 3D-differentiated AD neurons were treated with inhibitors targeting β-secretase (BACE1) and γ-secretases. As predicted, both BACE1 and γ-secretase inhibitors dramatically decreased Aβ generation in iPSC-derived neural cells derived from all five AD patients, under standard two-dimensional (2D) differentiation conditions. However, BACE1 and γ-secretase inhibitors showed less potency in decreasing Aβ levels in neural cells differentiated under 3D culture conditions. Interestingly, in a single subject AD1, we found that BACE1 inhibitor treatment was not able to significantly reduce Aβ42 levels. To investigate underlying molecular mechanisms, we performed proteomic analysis of 3D AD human neuronal cultures including AD1. Proteomic analysis revealed specific reduction of several proteins that might contribute to a poor inhibition of BACE1 in subject AD1. To our knowledge, this is the first iPSC-differentiated 3D neuro-spheroid model derived from AD patients’ blood. Our results demonstrate that our 3D human neuro-spheroid model can be a physiologically relevant and valid model for testing efficacy of AD drug.
Collapse
|
20
|
Calderón-Garcidueñas L, San Juan Chávez V, Vacaseydel-Aceves NB, Calderón-Sánchez R, Macías-Escobedo E, Frías C, Giacometto M, Velasquez L, Félix-Villarreal R, Martin JD, Draheim C, Engle RW. Chocolate, Air Pollution and Children's Neuroprotection: What Cognition Tools should be at Hand to Evaluate Interventions? Front Pharmacol 2016; 7:232. [PMID: 27563291 PMCID: PMC4980563 DOI: 10.3389/fphar.2016.00232] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/18/2016] [Indexed: 12/19/2022] Open
Abstract
Millions of children across the world are exposed to multiple sources of indoor and outdoor air pollutants, including high concentrations of fine particulate matter (PM2.5) and ozone (O3). The established link between exposure to PM2.5, brain structural, volumetric and metabolic changes, severe cognitive deficits (1.5-2 SD from average IQ) in APOE 4 heterozygous females with >75 − < 94% BMI percentiles, and the presence of Alzheimer's disease (AD) hallmarks in urban children and young adults necessitates exploration of ways to protect these individuals from the deleterious neural effects of pollution exposure. Emerging research suggests that cocoa interventions may be a viable option for neuroprotection, with evidence suggesting that early cocoa interventions could limit the risk of cognitive and developmental concerns including: endothelial dysfunction, cerebral hypoperfusion, neuroinflammation, and metabolic detrimental brain effects. Currently, however, it is not clear how early we should implement consumption of cocoa to optimize its neuroprotective effects. Moreover, we have yet to identify suitable instruments for evaluating cognitive responses to these interventions in clinically healthy children, teens, and young adults. An approach to guide the selection of cognitive tools should take into account neuropsychological markers of cognitive declines in patients with Alzheimer's neuropathology, the distinct patterns of memory impairment between early and late onset AD, and the key literature associating white matter integrity and poor memory binding performance in cases of asymptomatic familial AD. We highlight potential systemic and neural benefits of cocoa consumption. We also highlight Working Memory Capacity (WMC) and attention control tasks as opened avenues for exploration in the air pollution scenario. Exposures to air pollutants during brain development have serious brain consequences in the short and long term and reliable cognition tools should be at hand to evaluate interventions.
Collapse
Affiliation(s)
- Lilian Calderón-Garcidueñas
- Biomedical Sciences, University of MontanaMissoula, MT, USA; Universidad del Valle de MéxicoCiudad de México, Mexico
| | | | | | | | | | | | | | - Luis Velasquez
- Facultad de Medicina, Universidad Andrés Bello Santiago de Chile, Chile
| | | | - Jessie D Martin
- School of Psychology, Georgia Institute of Technology Atlanta, GA, USA
| | | | - Randall W Engle
- School of Psychology, Georgia Institute of Technology Atlanta, GA, USA
| |
Collapse
|
21
|
Kazim SF, Iqbal K. Neurotrophic factor small-molecule mimetics mediated neuroregeneration and synaptic repair: emerging therapeutic modality for Alzheimer's disease. Mol Neurodegener 2016; 11:50. [PMID: 27400746 PMCID: PMC4940708 DOI: 10.1186/s13024-016-0119-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 07/02/2016] [Indexed: 11/10/2022] Open
Abstract
Alzheimer's disease (AD) is an incurable and debilitating chronic progressive neurodegenerative disorder which is the leading cause of dementia worldwide. AD is a heterogeneous and multifactorial disorder, histopathologically characterized by the presence of amyloid β (Aβ) plaques and neurofibrillary tangles composed of Aβ peptides and abnormally hyperphosphorylated tau protein, respectively. Independent of the various etiopathogenic mechanisms, neurodegeneration is a final common outcome of AD neuropathology. Synaptic loss is a better correlate of cognitive impairment in AD than Aβ or tau pathologies. Thus a highly promising therapeutic strategy for AD is to shift the balance from neurodegeneration to neuroregeneration and synaptic repair. Neurotrophic factors, by virtue of their neurogenic and neurotrophic activities, have potential for the treatment of AD. However, the clinical therapeutic usage of recombinant neurotrophic factors is limited because of the insurmountable hurdles of unfavorable pharmacokinetic properties, poor blood-brain barrier (BBB) permeability, and severe adverse effects. Neurotrophic factor small-molecule mimetics, in this context, represent a potential strategy to overcome these short comings, and have shown promise in preclinical studies. Neurotrophic factor small-molecule mimetics have been the focus of intense research in recent years for AD drug development. Here, we review the relevant literature regarding the therapeutic beneficial effect of neurotrophic factors in AD, and then discuss the recent status of research regarding the neurotrophic factor small-molecule mimetics as therapeutic candidates for AD. Lastly, we summarize the preclinical studies with a ciliary neurotrophic factor (CNTF) small-molecule peptide mimetic, Peptide 021 (P021). P021 is a neurogenic and neurotrophic compound which enhances dentate gyrus neurogenesis and memory processes via inhibiting leukemia inhibitory factor (LIF) signaling pathway and increasing brain-derived neurotrophic factor (BDNF) expression. It robustly inhibits tau abnormal hyperphosphorylation via increased BDNF mediated decrease in glycogen synthase kinase-3β (GSK-3β, major tau kinase) activity. P021 is a small molecular weight, BBB permeable compound with suitable pharmacokinetics for oral administration, and without adverse effects associated with native CNTF or BDNF molecule. P021 has shown beneficial therapeutic effect in several preclinical studies and has emerged as a highly promising compound for AD drug development.
Collapse
Affiliation(s)
- Syed Faraz Kazim
- />Department of Neurochemistry, and SUNY Downstate/NYSIBR Program in Developmental Neuroscience, New York State Institute for Basic Research (NYSIBR), 1050 Forest Hill Road, Staten Island, NY 10314 USA
- />Graduate Program in Neural and Behavioral Science, and Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203 USA
| | - Khalid Iqbal
- />Department of Neurochemistry, and SUNY Downstate/NYSIBR Program in Developmental Neuroscience, New York State Institute for Basic Research (NYSIBR), 1050 Forest Hill Road, Staten Island, NY 10314 USA
| |
Collapse
|
22
|
Downer B, Vickers BN, Al Snih S, Raji M, Markides KS. Effects of Comorbid Depression and Diabetes Mellitus on Cognitive Decline in Older Mexican Americans. J Am Geriatr Soc 2016; 64:109-17. [PMID: 26782859 DOI: 10.1111/jgs.13883] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVES To assess the relationship between comorbid depression, diabetes mellitus (DM), and cognitive decline in Mexican Americans aged 65 and older. DESIGN Retrospective cohort study with longitudinal analysis. SETTING Texas, New Mexico, Colorado, Arizona, and California. PARTICIPANTS Hispanic Established Populations for the Epidemiologic Study of the Elderly. MEASUREMENTS Cognition was assessed using the Mini-Mental State Examination (MMSE). Depression was defined as a score of 16 or greater on the Center for Epidemiologic Studies Depression Scale. DM was defined as according to self-reported history or taking insulin or oral hypoglycemic medication. RESULTS Participants with depression and DM declined an average of 6.5 points on the MMSE; depression only, 4.4 points; DM only, 7.8 points; and neither condition, 4.2 points across the six examination waves. Participants with DM declined an average of 0.18 more points on the MMSE per year (P=.001) than those with neither DM nor depression, and those with comorbid DM and depression declined 0.25 more points per year (P=.002). Depression was associated with significantly greater cognitive decline (β^=-0.11, P=.05) after excluding participants with baseline cognitive impairment (MMSE score≤17). Participants with DM were 1.08 (95% CI=1.03-1.12) times as likely as those with neither DM nor depression, and those with comorbid DM and depression were 1.08 (95% CI=1.01-1.15) times as likely as those with neither DM nor depression to develop severe cognitive impairment per year. CONCLUSION DM and comorbid depression and DM are risk factors for cognitive decline in older Mexican Americans. Interventions that reduce the prevalence of depression and DM in Mexican Americans may decrease the number of older adults who experience cognitive decline.
Collapse
Affiliation(s)
- Brian Downer
- Sealy Center on Aging, University of Texas Medical Branch, Galveston, Texas
| | - Benjamin N Vickers
- Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, Texas
| | - Soham Al Snih
- Department of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Mukaila Raji
- Division of Geriatrics, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Kyriakos S Markides
- Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
23
|
Mufson EJ, Malek-Ahmadi M, Snyder N, Ausdemore J, Chen K, Perez SE. Braak stage and trajectory of cognitive decline in noncognitively impaired elders. Neurobiol Aging 2016; 43:101-10. [PMID: 27255819 DOI: 10.1016/j.neurobiolaging.2016.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 12/24/2022]
Abstract
In a previous cross-sectional study, we found that nondemented elderly participants from the Rush Religious Orders Study (RROS) displayed a wide range of Braak neurofibrillary tangle and amyloid plaque pathology similar to that seen in prodromal and frank Alzheimer's disease. Here, we examined longitudinal changes in cognitive domains in subjects from this cohort grouped by Braak stage using linear mixed effects models. We found that the trajectory of episodic memory composite (EMC), executive function composite (EFC), and global cognitive composite scores (GCS: average of EMC and EFC scores) was significantly associated with age at visit over time, but not with Braak stage, apolipoprotein E (APOE) ε4 status or plaque pathology alone. By contrast, the combined effects of Braak stage, APOE status, and age at visit were strongly correlated with the trajectory of EMC, EFC and GCS performance over time. These data suggest that age and APOE ε4 status, rather than Alzheimer's disease-related pathology, play a more prominent role in the trajectory of cognitive decline over time in this elderly nondemented population. However, the findings reported require confirmation in a larger cohort of cases.
Collapse
Affiliation(s)
- Elliott J Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA.
| | | | | | | | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Sylvia E Perez
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
24
|
Dubois B, Hampel H, Feldman HH, Scheltens P, Aisen P, Andrieu S, Bakardjian H, Benali H, Bertram L, Blennow K, Broich K, Cavedo E, Crutch S, Dartigues JF, Duyckaerts C, Epelbaum S, Frisoni GB, Gauthier S, Genthon R, Gouw AA, Habert MO, Holtzman DM, Kivipelto M, Lista S, Molinuevo JL, O'Bryant SE, Rabinovici GD, Rowe C, Salloway S, Schneider LS, Sperling R, Teichmann M, Carrillo MC, Cummings J, Jack CR. Preclinical Alzheimer's disease: Definition, natural history, and diagnostic criteria. Alzheimers Dement 2016; 12:292-323. [PMID: 27012484 PMCID: PMC6417794 DOI: 10.1016/j.jalz.2016.02.002] [Citation(s) in RCA: 1197] [Impact Index Per Article: 149.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
During the past decade, a conceptual shift occurred in the field of Alzheimer's disease (AD) considering the disease as a continuum. Thanks to evolving biomarker research and substantial discoveries, it is now possible to identify the disease even at the preclinical stage before the occurrence of the first clinical symptoms. This preclinical stage of AD has become a major research focus as the field postulates that early intervention may offer the best chance of therapeutic success. To date, very little evidence is established on this "silent" stage of the disease. A clarification is needed about the definitions and lexicon, the limits, the natural history, the markers of progression, and the ethical consequence of detecting the disease at this asymptomatic stage. This article is aimed at addressing all the different issues by providing for each of them an updated review of the literature and evidence, with practical recommendations.
Collapse
Affiliation(s)
- Bruno Dubois
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France.
| | - Harald Hampel
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France; AXA Research Fund & UPMC Chair, Paris, France
| | | | - Philip Scheltens
- Department of Neurology and Alzheimer Center, VU University Medical Center and Neuroscience Campus, Amsterdam, The Netherlands
| | - Paul Aisen
- University of Southern California San Diego, CA, USA
| | - Sandrine Andrieu
- UMR1027, INSERM, Université Toulouse III, Toulouse University Hospital, France
| | - Hovagim Bakardjian
- IHU-A-ICM-Institut des Neurosciences translationnelles de Paris, Paris, France
| | - Habib Benali
- INSERM U1146-CNRS UMR 7371-UPMC UM CR2, Site Pitié-Salpêtrière, Paris, France
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), Institutes of Neurogenetics and Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany; School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Lab, Department of Neuroscience and Physiology, University of Gothenburg, Mölndal Hospital, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Enrica Cavedo
- AXA Research Fund & UPMC Chair, Paris, France; Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Sebastian Crutch
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | | | - Charles Duyckaerts
- University Pierre et Marie Curie, Assistance Publique des Hôpitaux de Paris, Alzheimer-Prion Team Institut du Cerveau et de la Moelle (ICM), Paris, France
| | - Stéphane Epelbaum
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France
| | - Giovanni B Frisoni
- University Hospitals and University of Geneva, Geneva, Switzerland; IRCCS Fatebenefratelli, Brescia, Italy
| | - Serge Gauthier
- McGill Center for Studies in Aging, Douglas Mental Health Research Institute, Montreal, Canada
| | - Remy Genthon
- Fondation pour la Recherche sur Alzheimer, Hôpital Pitié-Salpêtrière, Paris, France
| | - Alida A Gouw
- UMR1027, INSERM, Université Toulouse III, Toulouse University Hospital, France; Department of Clinical Neurophysiology/MEG Center, VU University Medical Center, Amsterdam
| | - Marie-Odile Habert
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, Département de Médecine Nucléaire, Paris, France
| | - David M Holtzman
- Department of Neurology, Washington University, Hope Center for Neurological Disorders, St. Louis, MO, USA; Department of Neurology, Washington University, Knight Alzheimer's Disease Research Center, St. Louis, MO, USA
| | - Miia Kivipelto
- Center for Alzheimer Research, Karolinska Institutet, Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden; Institute of Clinical Medicine/ Neurology, University of Eastern Finland, Kuopio, Finland
| | | | - José-Luis Molinuevo
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Sid E O'Bryant
- Center for Alzheimer's & Neurodegenerative Disease Research, University of North Texas Health Science Center, TX, USA
| | - Gil D Rabinovici
- Memory & Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Christopher Rowe
- Department of Molecular Imaging, Austin Health, University of Melbourne, Australia
| | - Stephen Salloway
- Memory and Aging Program, Butler Hospital, Alpert Medical School of Brown University, USA; Department of Neurology, Alpert Medical School of Brown University, USA; Department of Psychiatry, Alpert Medical School of Brown University, USA
| | - Lon S Schneider
- Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Reisa Sperling
- Harvard Medical School, Memory Disorders Unit, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, USA; Harvard Medical School, Memory Disorders Unit, Center for Alzheimer Research and Treatment, Massachusetts General Hospital, Boston, USA
| | - Marc Teichmann
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France
| | - Maria C Carrillo
- The Alzheimer's Association Division of Medical & Scientific Relations, Chicago, USA
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Cliff R Jack
- Department of Radiology, Mayo Clinic, Rochester MN, USA
| |
Collapse
|
25
|
A Mendelian randomization study of testosterone and cognition in men. Sci Rep 2016; 6:21306. [PMID: 26864717 PMCID: PMC4749999 DOI: 10.1038/srep21306] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/19/2016] [Indexed: 11/08/2022] Open
Abstract
Testosterone replacement for older men is increasingly common, with some observations suggesting a protective effect on cognitive function. We examined the association of endogenous testosterone with cognitive function among older men in a Mendelian randomization study using a separate-sample instrumental variable (SSIV) analysis estimator to minimize confounding and reverse causality. A genetic score predicting testosterone was developed in 289 young Chinese men from Hong Kong, based on selected testosterone-related single nucleotide polymorphisms (rs10046, rs1008805 and rs1256031). The association of genetically predicted testosterone with delayed 10-word recall score and Mini-Mental State Examination (MMSE) score was assessed at baseline and follow-up using generalized estimating equation among 4,212 older Chinese men from the Guangzhou Biobank Cohort Study. Predicted testosterone was not associated with delayed 10-word recall score (-0.02 per nmol/L testosterone, 95% confidence interval (CI) -0.06-0.02) or MMSE score (0.06, 95% CI -0.002-0.12). These estimates were similar after additional adjustment for age, education, smoking, use of alcohol, body mass index and the Framingham score. Our findings do not corroborate observed protective effects of testosterone on cognitive function among older men.
Collapse
|
26
|
Neuropsychological Markers of Cognitive Decline in Persons With Alzheimer Disease Neuropathology. J Neuropathol Exp Neurol 2016; 74:1086-92. [PMID: 26469250 DOI: 10.1097/nen.0000000000000254] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
To evaluate cognitive performance among persons who did and did not develop clinical Alzheimer disease (AD) but had AD neuropathology at autopsy, we examined neuropsychological performance in cognitively healthy (Clinical Dementia Rating [CDR] = 0) participants who returned for at least 1 follow-up and died within 2 years of their last assessment. Nonprogressors remained at CDR = 0 until death; progressors developed symptomatic AD during life (CDR > 0). Cognitive performance at baseline was compared between progressors and nonprogressors on a global cognitive composite and 4 domain-specific composites (episodic memory, language, attention/working memory, and executive function). Models adjusted for age, education, sex, and non-AD neuropathology. Progressors (n = 173) had worse performance than nonprogressors (n = 141) in nearly all cognitive domains. Progressors scored lower on composites of global cognition (P < 0.001), executive function (P = 0.0006), language (P < 0.0001), and episodic memory (P = 0.0006) but not on attention/working memory (P = 0.91). These data indicate that individuals with underlying AD neuropathology who are clinically healthy but who later develop symptomatic AD have worse performance in a wide range of domains versus individuals with underlying AD neuropathology who are clinically healthy but do not become symptomatic during life. Therefore, subtle cognitive decline at baseline may indicate an increased risk of progression to symptomatic AD.
Collapse
|
27
|
Downer B, Fardo DW, Schmitt FA. A Summary Score for the Framingham Heart Study Neuropsychological Battery. J Aging Health 2015; 27:1199-222. [PMID: 25804903 DOI: 10.1177/0898264315577590] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To calculate three summary scores of the Framingham Heart Study neuropsychological battery and determine which score best differentiates between subjects classified as having normal cognition, test-based impaired learning and memory, test-based multidomain impairment, and dementia. METHOD The final sample included 2,503 participants. Three summary scores were assessed: (a) composite score that provided equal weight to each subtest, (b) composite score that provided equal weight to each cognitive domain assessed by the neuropsychological battery, and (c) abbreviated score comprised of subtests for learning and memory. Receiver operating characteristic analysis was used to determine which summary score best differentiated between the four cognitive states. RESULTS The summary score that provided equal weight to each subtest best differentiated between the four cognitive states. DISCUSSION A summary score that provides equal weight to each subtest is an efficient way to utilize all of the cognitive data collected by a neuropsychological battery.
Collapse
Affiliation(s)
- Brian Downer
- University of Texas Medical Branch, Galveston, USA
| | | | | |
Collapse
|
28
|
Malek-Ahmadi M, Chen K, Davis K, Belden CM, Powell J, Jacobson SA, Sabbagh MN. Sensitivity to change and prediction of global change for the Alzheimer's Questionnaire. ALZHEIMERS RESEARCH & THERAPY 2015; 7:1. [PMID: 26584966 PMCID: PMC4652427 DOI: 10.1186/s13195-014-0092-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 12/22/2014] [Indexed: 11/10/2022]
Abstract
INTRODUCTION Longitudinal assessment of cognitive decline in amnestic mild cognitive impairment (aMCI) and Alzheimer's disease (AD) often involves the use of both informant-based and objective cognitive assessments. As efforts have focused on identifying individuals in pre-clinical stages, instruments that are sensitive to subtle cognitive changes are needed. The Alzheimer's Questionnaire (AQ) has demonstrated high sensitivity and specificity in identifying aMCI and AD; however its ability to measure longitudinal change has not been assessed. The aims of this study are to assess the sensitivity to change of the AQ and to determine whether the AQ predicts change in global cognition and function in cognitively normal (CN), aMCI, and AD subjects. METHODS Data from 202 individuals participating in a brain and body donation program were utilized for this study (101 CN, 62 aMCI, 39 AD). AD and aMCI individuals were matched on age, education, and gender to CN individuals. Sensitivity to change of the AQ was assessed in addition to the AQ's ability to predict change in global cognition and function. The Mini Mental State Exam (MMSE) and Functional Activities Questionnaire (FAQ) were used as gold standard comparisons of cognition and function. Sample size calculations for a 25% treatment effect were also carried out for all three groups. RESULTS The AQ demonstrated small sensitivity to change in the aMCI and CN groups (d = 0.33, d = 0.23, respectively) and moderate sensitivity to change in the AD group (d = 0.43). The AQ was associated with increases in the Clinical Dementia Rating Global Score (OR = 1.20 (1.09, 1.32), P <0.001). Sample size calculations found that the AQ would require substantially fewer subjects than the MMSE given a 25% treatment effect. CONCLUSIONS Although the AQ demonstrated small sensitivity to change in aMCI and CN individuals in terms of effect size, the AQ may be superior to objective cognitive tests in terms of required sample size for a clinical trial. As clinicians and researchers continue to identify and treat individuals in earlier stages of AD, there is a need for instruments that are sensitive to cognitive changes in these earlier stages.
Collapse
Affiliation(s)
- Michael Malek-Ahmadi
- The Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ, 85351, USA.
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, 85006, USA.
| | - Kathryn Davis
- The Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ, 85351, USA.
| | - Christine M Belden
- The Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ, 85351, USA.
| | - Jessica Powell
- The Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ, 85351, USA.
| | - Sandra A Jacobson
- The Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ, 85351, USA.
| | - Marwan N Sabbagh
- The Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ, 85351, USA.
| |
Collapse
|
29
|
Langbaum JB, Hendrix SB, Ayutyanont N, Chen K, Fleisher AS, Shah RC, Barnes LL, Bennett DA, Tariot PN, Reiman EM. An empirically derived composite cognitive test score with improved power to track and evaluate treatments for preclinical Alzheimer's disease. Alzheimers Dement 2014; 10:666-74. [PMID: 24751827 PMCID: PMC4201904 DOI: 10.1016/j.jalz.2014.02.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 01/18/2014] [Accepted: 02/11/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND There is growing interest in the evaluation of preclinical Alzheimer's disease (AD) treatments. As a result, there is a need to identify a cognitive composite that is sensitive to track preclinical AD decline to be used as a primary endpoint in treatment trials. METHODS Longitudinal data from initially cognitively normal, 70- to 85-year-old participants in three cohort studies of aging and dementia from the Rush Alzheimer's Disease Center were examined to empirically define a composite cognitive endpoint that is sensitive to detect and track cognitive decline before the onset of cognitive impairment. The mean-to-standard deviation ratios (MSDRs) of change over time were calculated in a search for the optimal combination of cognitive tests/subtests drawn from the neuropsychological battery in cognitively normal participants who subsequently progressed to clinical stages of AD during 2- and 5-year periods, using data from those who remained unimpaired during the same period to correct for aging and practice effects. Combinations that performed well were then evaluated for representation of relevant cognitive domains, robustness across individual years before diagnosis, and occurrence of selected items within top performing combinations. RESULTS The optimal composite cognitive test score comprised seven cognitive tests/subtests with an MSDR = 0.964. By comparison, the most sensitive individual test score was Logical Memory Delayed Recall with an MSDR = 0.64. CONCLUSIONS We have identified a composite cognitive test score representing multiple cognitive domains that has improved power compared with the most sensitive single test item to track preclinical AD decline and evaluate preclinical AD treatments. We are confirming the power of the composite in independent cohorts and with other analytical approaches, which may result in refinements, have designated it as the primary endpoint in the Alzheimer's Prevention Initiative's preclinical treatment trials for individuals at high imminent risk for developing symptoms due to late-onset AD.
Collapse
Affiliation(s)
- Jessica B Langbaum
- Banner Alzheimer's Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| | | | - Napatkamon Ayutyanont
- Banner Alzheimer's Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA; Department of Mathematics and Statistics, Arizona State University, Tempe, AZ, USA
| | - Adam S Fleisher
- Banner Alzheimer's Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Raj C Shah
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Family Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Pierre N Tariot
- Banner Alzheimer's Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA; Department of Psychiatry, University of Arizona, Tucson, AZ, USA
| | - Eric M Reiman
- Banner Alzheimer's Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA; Department of Psychiatry, University of Arizona, Tucson, AZ, USA; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| |
Collapse
|
30
|
Early neuropsychological detection of Alzheimer's disease. Eur J Clin Nutr 2014; 68:1192-9. [PMID: 25182019 DOI: 10.1038/ejcn.2014.176] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 07/28/2014] [Indexed: 02/08/2023]
Abstract
Lifestyle modification offers a promising way of preventing or delaying Alzheimer's disease (AD). In particular, nutritional interventions can contribute to decrease the risk of dementia. The efficacy of such interventions should be assessed in individuals thought to be prone to AD. It is therefore necessary to identify markers that may help detecting AD as early as possible. This review will focus on subtle neuropsychological changes that may already exist in the predementia phase, and that could point to individuals at risk of dementia. Episodic memory decline appears consistently as the earliest sign of incipient typical AD. An episodic memory test that ensures deep encoding of information and assesses retrieval with free as well as cued recall appears as a useful tool to detect patients at an early stage of AD. Beyond the memory domain, category verbal fluency has been shown to decline early and to predict progression to AD. Moreover, in line with current diagnosis criteria for prodromal AD, combining neuropsychological scores and neuroimaging data allows a better discrimination of future AD patients than neuroimaging or neuropsychological data alone. Altogether, the detection of cognitive changes that are predictive of the typical form of probable AD already in the predementia stage points to at risk people who are the best target for therapeutic interventions, such as nutrition or physical exercise counseling or dietary interventions.
Collapse
|
31
|
Monsell SE, Mock C, Hassenstab J, Roe CM, Cairns NJ, Morris JC, Kukull W. Neuropsychological changes in asymptomatic persons with Alzheimer disease neuropathology. Neurology 2014; 83:434-40. [PMID: 24951474 DOI: 10.1212/wnl.0000000000000650] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether asymptomatic persons with Alzheimer disease (AD) neuropathologic change differ in the trajectory of their cognitive performance compared to asymptomatic persons without AD neuropathologic change. METHODS Longitudinal performance on standard neuropsychological tests was examined in participants who died within 2 years of their last cognitive assessment and who were never diagnosed with mild cognitive impairment or dementia (Clinical Dementia Rating global score of 0 at all assessments). Using cognitive and neuropathologic data collected between 2005 and 2013 from the 34 National Institute on Aging-sponsored Alzheimer's Disease Centers, cognitive trajectories were compared for persons with and without evidence of AD neuropathologic change. We evaluated rates of decline in 4 domains (episodic memory, language, attention/working memory, executive function). The significance of the differences (β) in rates of decline was tested using linear regression, adjusting for age, education, sex, and other neuropathologic lesions. RESULTS Participants who had low to high levels of AD neuropathologic change (n = 131) showed a greater rate of decline on the attention/working memory domain score (β = -0.11; 95% confidence interval = -0.19, -0.02; p = 0.02) when compared to 80 participants who died without evidence of AD neuropathologic change. CONCLUSIONS Clinically normal individuals who come to autopsy with AD neuropathologic change exhibit subtle evidence of declining cognitive trajectories for attention/working memory.
Collapse
Affiliation(s)
- Sarah E Monsell
- From the National Alzheimer's Coordinating Center (S.E.M., C.M., W.K.), University of Washington, Seattle; and Knight Alzheimer's Disease Research Center (J.H., C.M.R., N.J.C., J.C.M.), Department of Neurology, Washington University School of Medicine, St. Louis, MO.
| | - Charles Mock
- From the National Alzheimer's Coordinating Center (S.E.M., C.M., W.K.), University of Washington, Seattle; and Knight Alzheimer's Disease Research Center (J.H., C.M.R., N.J.C., J.C.M.), Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Jason Hassenstab
- From the National Alzheimer's Coordinating Center (S.E.M., C.M., W.K.), University of Washington, Seattle; and Knight Alzheimer's Disease Research Center (J.H., C.M.R., N.J.C., J.C.M.), Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Catherine M Roe
- From the National Alzheimer's Coordinating Center (S.E.M., C.M., W.K.), University of Washington, Seattle; and Knight Alzheimer's Disease Research Center (J.H., C.M.R., N.J.C., J.C.M.), Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Nigel J Cairns
- From the National Alzheimer's Coordinating Center (S.E.M., C.M., W.K.), University of Washington, Seattle; and Knight Alzheimer's Disease Research Center (J.H., C.M.R., N.J.C., J.C.M.), Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - John C Morris
- From the National Alzheimer's Coordinating Center (S.E.M., C.M., W.K.), University of Washington, Seattle; and Knight Alzheimer's Disease Research Center (J.H., C.M.R., N.J.C., J.C.M.), Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Walter Kukull
- From the National Alzheimer's Coordinating Center (S.E.M., C.M., W.K.), University of Washington, Seattle; and Knight Alzheimer's Disease Research Center (J.H., C.M.R., N.J.C., J.C.M.), Department of Neurology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
32
|
Ayutyanont N, Langbaum JB, Hendrix SB, Chen K, Fleisher AS, Friesenhahn M, Ward M, Aguirre C, Acosta-Baena N, Madrigal L, Muñoz C, Tirado V, Moreno S, Tariot PN, Lopera F, Reiman EM. The Alzheimer's prevention initiative composite cognitive test score: sample size estimates for the evaluation of preclinical Alzheimer's disease treatments in presenilin 1 E280A mutation carriers. J Clin Psychiatry 2014; 75:652-60. [PMID: 24816373 PMCID: PMC4331113 DOI: 10.4088/jcp.13m08927] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 02/13/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To identify a cognitive composite that is sensitive to tracking preclinical Alzheimer's disease decline to be used as a primary end point in treatment trials. METHOD We capitalized on longitudinal data collected from 1995 to 2010 from cognitively unimpaired presenilin 1 (PSEN1) E280A mutation carriers from the world's largest known early-onset autosomal dominant Alzheimer's disease kindred to identify a composite cognitive test with the greatest statistical power to track preclinical Alzheimer's disease decline and estimate the number of carriers age 30 years and older needed to detect a treatment effect in the Alzheimer's Prevention Initiative's (API) preclinical Alzheimer's disease treatment trial. The mean-to-standard-deviation ratios (MSDRs) of change over time were calculated in a search for the optimal combination of 1 to 7 cognitive tests/subtests drawn from the neuropsychological test battery in cognitively unimpaired mutation carriers during a 2- and 5-year follow-up period (n = 78 and 57), using data from noncarriers (n = 31 and 56) during the same time period to correct for aging and practice effects. Combinations that performed well were then evaluated for robustness across follow-up years, occurrence of selected items within top-performing combinations, and representation of relevant cognitive domains. RESULTS The optimal test combination included Consortium to Establish a Registry for Alzheimer's Disease (CERAD) Word List Recall, CERAD Boston Naming Test (high frequency items), Mini-Mental State Examination (MMSE) Orientation to Time, CERAD Constructional Praxis, and Raven's Progressive Matrices (Set A), with an MSDR of 1.62. This composite is more sensitive than using either the CERAD Word List Recall (MSDR = 0.38) or the entire CERAD-Col battery (MSDR = 0.76). A sample size of 75 cognitively normal PSEN1 E280A mutation carriers aged 30 years and older per treatment arm allows for a detectable treatment effect of 29% in a 60-month trial (80% power, P = .05). CONCLUSIONS We have identified a composite cognitive test score representing multiple cognitive domains that, compared to the most sensitive single test item, has improved power to track preclinical Alzheimer's disease decline in autosomal dominant Alzheimer's disease mutation carriers and to evaluate preclinical Alzheimer's disease treatments. This API composite cognitive test score will be used as the primary end point in the first API trial in cognitively unimpaired autosomal dominant Alzheimer's disease carriers within 15 years of their estimated age at clinical onset. We have independently confirmed our findings in a separate cohort of cognitively healthy older adults who progressed to the clinical stages of late-onset Alzheimer's disease, described in a separate report, and continue to refine the composite in independent cohorts and compared with other analytic approaches.
Collapse
Affiliation(s)
| | - Jessica B. Langbaum
- Banner Alzheimer’s Institute, Phoenix, AZ,Arizona Alzheimer’s Consortium, Phoenix, AZ
| | | | - Kewei Chen
- Banner Alzheimer’s Institute, Phoenix, AZ,Department of Mathematics and Statistics, Arizona State University, Tempe, AZ,Arizona Alzheimer’s Consortium, Phoenix, AZ
| | - Adam S. Fleisher
- Banner Alzheimer’s Institute, Phoenix, AZ,Department of Neurology, University of California, San Diego, CA,Arizona Alzheimer’s Consortium, Phoenix, AZ
| | | | - Michael Ward
- F Hoffmann-La Roche, Ltd, South San Francisco, CA
| | - Camilo Aguirre
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia
| | - Natalia Acosta-Baena
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia
| | - Lucìa Madrigal
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia
| | - Claudia Muñoz
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia
| | - Victoria Tirado
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia
| | - Sonia Moreno
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia
| | - Pierre N. Tariot
- Banner Alzheimer’s Institute, Phoenix, AZ,Department of Psychiatry, University of Arizona, Tucson, AZ,Arizona Alzheimer’s Consortium, Phoenix, AZ
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia
| | - Eric M. Reiman
- Banner Alzheimer’s Institute, Phoenix, AZ,Department of Psychiatry, University of Arizona, Tucson, AZ,Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ,Arizona Alzheimer’s Consortium, Phoenix, AZ
| |
Collapse
|
33
|
Webster SJ, Bachstetter AD, Nelson PT, Schmitt FA, Van Eldik LJ. Using mice to model Alzheimer's dementia: an overview of the clinical disease and the preclinical behavioral changes in 10 mouse models. Front Genet 2014; 5:88. [PMID: 24795750 PMCID: PMC4005958 DOI: 10.3389/fgene.2014.00088] [Citation(s) in RCA: 494] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/01/2014] [Indexed: 01/17/2023] Open
Abstract
The goal of this review is to discuss how behavioral tests in mice relate to the pathological and neuropsychological features seen in human Alzheimer's disease (AD), and present a comprehensive analysis of the temporal progression of behavioral impairments in commonly used AD mouse models that contain mutations in amyloid precursor protein (APP). We begin with a brief overview of the neuropathological changes seen in the AD brain and an outline of some of the clinical neuropsychological assessments used to measure cognitive deficits associated with the disease. This is followed by a critical assessment of behavioral tasks that are used in AD mice to model the cognitive changes seen in the human disease. Behavioral tests discussed include spatial memory tests [Morris water maze (MWM), radial arm water maze (RAWM), Barnes maze], associative learning tasks (passive avoidance, fear conditioning), alternation tasks (Y-Maze/T-Maze), recognition memory tasks (Novel Object Recognition), attentional tasks (3 and 5 choice serial reaction time), set-shifting tasks, and reversal learning tasks. We discuss the strengths and weaknesses of each of these behavioral tasks, and how they may correlate with clinical assessments in humans. Finally, the temporal progression of both cognitive and non-cognitive deficits in 10 AD mouse models (PDAPP, TG2576, APP23, TgCRND8, J20, APP/PS1, TG2576 + PS1 (M146L), APP/PS1 KI, 5×FAD, and 3×Tg-AD) are discussed in detail. Mouse models of AD and the behavioral tasks used in conjunction with those models are immensely important in contributing to our knowledge of disease progression and are a useful tool to study AD pathophysiology and the resulting cognitive deficits. However, investigators need to be aware of the potential weaknesses of the available preclinical models in terms of their ability to model cognitive changes observed in human AD. It is our hope that this review will assist investigators in selecting an appropriate mouse model, and accompanying behavioral paradigms to investigate different aspects of AD pathology and disease progression.
Collapse
Affiliation(s)
- Scott J Webster
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA ; Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of Kentucky Lexington, KY, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA ; Department of Neurology, University of Kentucky Lexington, KY, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA ; Department of Anatomy and Neurobiology, University of Kentucky Lexington, KY, USA
| |
Collapse
|
34
|
|
35
|
Schmitt FA, Nelson PT, Abner E, Scheff S, Jicha GA, Smith C, Cooper G, Mendiondo M, Danner DD, Van Eldik LJ, Caban-Holt A, Lovell MA, Kryscio RJ. University of Kentucky Sanders-Brown healthy brain aging volunteers: donor characteristics, procedures and neuropathology. Curr Alzheimer Res 2012; 9:724-33. [PMID: 22471862 DOI: 10.2174/156720512801322591] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 12/28/2011] [Accepted: 01/13/2012] [Indexed: 11/22/2022]
Abstract
Cognitively intact elderly research volunteers at the University of Kentucky have been recruited, followed longitudinally, and autopsied with extensive neuropathological evaluations since 1989. To date, the cohort has recruited 1,030 individuals with 552 participants being actively followed, 363 deceased, and 273 autopsied. An extensive database has been constructed with continuous updates that include textured clinical, neuropsychological, neuroimaging, and pathological information. The history, demographics, clinical observations, and pathological features of this research cohort are described. We also explain some of the evolving methodologies and the academic contributions that have been made due to this motivated group of older Kentuckians.
Collapse
Affiliation(s)
- Frederick A Schmitt
- Department of Neurology and the Sanders-Brown Center on Aging, 303 Sanders-Brown Building, 800 S. Limestone, University of Kentucky, Lexington, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Balasubramanian AB, Kawas CH, Peltz CB, Brookmeyer R, Corrada MM. Alzheimer disease pathology and longitudinal cognitive performance in the oldest-old with no dementia. Neurology 2012; 79:915-21. [PMID: 22895581 DOI: 10.1212/wnl.0b013e318266fc77] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE It has been hypothesized that individuals without dementia with Alzheimer disease (AD) neuropathology may be in the preclinical stages of dementia and could be experiencing subtle cognitive decline. The purpose of this study was to compare longitudinal cognitive performance in oldest-old individuals without dementia with and without AD neuropathology. METHODS The study included 58 individuals without dementia from The 90+ Autopsy Study, a population-based study of aging and dementia in individuals aged 90 and older. Participants had neurologic and neuropsychological testing every 6 months with an average of 3 years of follow-up. We compared the trajectory of cognitive performance on the Modified Mini-Mental State Examination (3MS) and the California Verbal Learning Test II (CVLT) by level of AD neuropathology. Based on Consortium to Establish a Registry for Alzheimer's Disease plaque staging, individuals were categorized as having low (none or sparse) or high (moderate or frequent) plaques. Based on Braak and Braak staging, participants were classified as having low (stages I-III) or high (IV-VI) tangles. RESULTS No significant differences were found in 3MS or CVLT cognitive performance over time based on plaque or tangle staging. Both high and low pathology groups showed modest improvements on the 3MS and CVLT consistent with learning effects. CONCLUSIONS AD neuropathology at autopsy is not associated with the trajectory of cognitive performance in the 3 years before death in oldest-old without dementia. Despite the presence of AD neuropathology at death, oldest-old without dementia display learning effects on cognitive tests. Further research is necessary to understand factors other than AD neuropathology that may affect cognition in the oldest-old.
Collapse
|