1
|
Bode M, Kalbe E, Liepelt-Scarfone I. Cognition and Activity of Daily Living Function in people with Parkinson's disease. J Neural Transm (Vienna) 2024; 131:1159-1186. [PMID: 38976044 PMCID: PMC11489248 DOI: 10.1007/s00702-024-02796-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/08/2024] [Indexed: 07/09/2024]
Abstract
The ability to perform activities of daily living (ADL) function is a multifaceted construct that reflects functionality in different daily life situations. The loss of ADL function due to cognitive impairment is the core feature for the diagnosis of Parkinson's disease dementia (PDD). In contrast to Alzheimer's disease, ADL impairment in PD can be compromised by various factors, including motor and non-motor aspects. This narrative review summarizes the current state of knowledge on the association of cognition and ADL function in people with PD and introduces the concept of "cognitive ADL" impairment for those problems in everyday life that are associated with cognitive deterioration as their primary cause. Assessment of cognitive ADL impairment is challenging because self-ratings, informant-ratings, and performance-based assessments seldomly differentiate between "cognitive" and "motor" aspects of ADL. ADL function in PD is related to multiple cognitive domains, with attention, executive function, and memory being particularly relevant. Cognitive ADL impairment is characterized by behavioral anomalies such as trial-and-error behavior or task step omissions, and is associated with lower engagement in everyday behaviors, as suggested by physical activity levels and prolonged sedentary behavior. First evidence shows that physical and multi-domain interventions may improve ADL function, in general, but the evidence is confounded by motor aspects. Large multicenter randomized controlled trials with cognitive ADL function as primary outcome are needed to investigate which pharmacological and non-pharmacological interventions can effectively prevent or delay deterioration of cognitive ADL function, and ultimately the progression and conversion to PDD.
Collapse
Affiliation(s)
- Merle Bode
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, Eberhard Karls University Tübingen, Hoppe-Seyler Str. 3, 72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Elke Kalbe
- Medical Psychology | Neuropsychology and Gender Studies & Center for Neuropsychological Diagnostics and Intervention (CeNDI), University Hospital Cologne, Cologne, Germany
- Medical Faculty, University of Cologne, Cologne, Germany
| | - Inga Liepelt-Scarfone
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, Eberhard Karls University Tübingen, Hoppe-Seyler Str. 3, 72076, Tübingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
- IB-Hochschule, Stuttgart, Germany.
| |
Collapse
|
2
|
Ali P, Renaud P, Montero-Odasso M, Gautier J, Dinomais M, Annweiler C. Gait performance in older adults across the cognitive spectrum: Results from the GAIT cohort. J Am Geriatr Soc 2024. [PMID: 39206968 DOI: 10.1111/jgs.19162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Gait performance can provide valuable insights into cognitive functioning in older adult and may be used to screen for cognitive impairment. However, the optimal test condition and spatiotemporal parameter for accuracy have not yet been determined. This study aims to determine the gait measure with the highest accuracy identifying cognitive decline. METHODS A total of 711 participants were recruited, including 332 cognitively healthy individuals, 264 with mild cognitive impairment (MCI), and 115 with dementia, with a mean age of 72 years (interquartile range 69-76), and 43% (n = 307) of women. The participants underwent gait assessment in three different conditions, including a single task and dual tasks of counting backward by ones and naming animals. RESULTS Gait performance was deteriorated as cognitive impairment progressed. The gait test performed during naming animals condition was the most accurate in differentiating between cognitive groups. Specifically, the naming animals gait speed was more accurate in discriminating control participants from those with cognitive impairment (area under the curve [AUC] = 76.9% for MCI and 99.7% for people with dementia with control group as reference). The coefficient of stride length variability while naming animals was the most effective parameter in discriminating between MCI and dementia groups (AUC = 96.7%). CONCLUSIONS The naming animals dual-task gait test can be a valuable assessment for screening cognitive impairment in older adults, regardless of their cognitive abilities. The test is useful in clinical settings for subjects with a range of cognitive profiles.
Collapse
Affiliation(s)
- Pauline Ali
- Robarts Research Institute, Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Laboratoire Angevin de Recherche en Ingénierie des Systèmes, EA7315, University of Angers, Angers, France
- Department of Physical and Rehabilitation Medicine, Angers University Hospital, University of Angers, Angers, France
| | - Pauline Renaud
- Department of Physical and Rehabilitation Medicine, Angers University Hospital, University of Angers, Angers, France
| | - Manuel Montero-Odasso
- Gait and Brain Lab, St. Joseph's Health Care, Parkwood Institute, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Medicine, Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jennifer Gautier
- UNIV ANGERS, UPRES EA 4638, University of Angers, Angers, France
- Department of Geriatric Medicine and Rehabilitation, Research Center on Autonomy and Longevity, University Hospital, Angers, France
| | - Mickaël Dinomais
- Laboratoire Angevin de Recherche en Ingénierie des Systèmes, EA7315, University of Angers, Angers, France
- Department of Physical and Rehabilitation Medicine, Angers University Hospital, University of Angers, Angers, France
| | - Cédric Annweiler
- Robarts Research Institute, Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- UNIV ANGERS, UPRES EA 4638, University of Angers, Angers, France
- Department of Geriatric Medicine and Rehabilitation, Research Center on Autonomy and Longevity, University Hospital, Angers, France
| |
Collapse
|
3
|
Zhang LL, Numbers K, Brodaty H, Lam BCP, Mahalingam G, Reppermund S. Does Mild Functional Impairment Predict Dementia in Older Adults With Normal Cognition? Alzheimer Dis Assoc Disord 2024; 38:257-264. [PMID: 39177170 DOI: 10.1097/wad.0000000000000638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 07/09/2024] [Indexed: 08/24/2024]
Abstract
OBJECTIVES Functional impairment can be an early indicator of cognitive decline. However, its predictive utility in cognitively normal (CN) older adults remains unclear. This study aimed to determine whether mild functional impairment (MFI) in CN older adults could predict incident dementia over 6 years, in addition to assessing its association with cognitive performance. DESIGN A longitudinal study with a 6-year follow-up. PARTICIPANTS A cohort of 296 community-dwelling CN older adults. MEASUREMENTS MFI was defined by cutoffs for impairment on an objective performance-based and/or subjective questionnaire-based functional assessment. Cox regression analysis was conducted to assess the relationship between MFI and risk of incident dementia and cognitive performances over 6 years. Linear regression analysis examined the association between MFI and baseline cognitive performance. RESULTS There were no significant longitudinal associations between MFI and incident dementia or changes in cognitive performance over 6 years. Defining MFI using both performance-based and informant-reported assessments was predictive of dementia. Cross-sectional analyses demonstrated significant associations between MFI and poorer baseline global cognition and performance in attention, visuospatial ability, and executive functioning. CONCLUSIONS CN older adults with MFI were not at an increased risk of developing dementia over 6 years. A definition of functional impairment requiring both performance-based and informant-based assessments may be useful in predicting dementia.
Collapse
Affiliation(s)
- Lei Lei Zhang
- School of Psychiatry, Centre for Healthy Brain Ageing (CHeBA)
| | - Katya Numbers
- School of Psychiatry, Centre for Healthy Brain Ageing (CHeBA)
| | - Henry Brodaty
- School of Psychiatry, Centre for Healthy Brain Ageing (CHeBA)
| | - Ben C P Lam
- School of Psychiatry, Centre for Healthy Brain Ageing (CHeBA)
- School of Psychology and Public Health, La Trobe University, Melbourne, Vic., Australia
| | | | - Simone Reppermund
- School of Psychiatry, Centre for Healthy Brain Ageing (CHeBA)
- Department of Developmental Disability Neuropsychiatry (3DN), Discipline of Psychiatry & Mental Health, School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW
| |
Collapse
|
4
|
Galvin JE, Cummings JL, Benea ML, de Moor C, Allegri RF, Atri A, Chertkow H, Paquet C, Porter VR, Ritchie CW, Sikkes SAM, Smith MR, Grassi CM, Rubino I. Generating real-world evidence in Alzheimer's disease: Considerations for establishing a core dataset. Alzheimers Dement 2024; 20:4331-4341. [PMID: 38706421 PMCID: PMC11180865 DOI: 10.1002/alz.13785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/18/2024] [Accepted: 02/12/2024] [Indexed: 05/07/2024]
Abstract
Ongoing assessment of patients with Alzheimer's disease (AD) in postapproval studies is important for mapping disease progression and evaluating real-world treatment effectiveness and safety. However, interpreting outcomes in the real world is challenging owing to variation in data collected across centers and specialties and greater heterogeneity of patients compared with trial participants. Here, we share considerations for observational postapproval studies designed to collect harmonized longitudinal data from individuals with mild cognitive impairment or mild dementia stage of disease who receive therapies targeting the underlying pathological processes of AD in routine practice. This paper considers key study design parameters, including proposed aims and objectives, study populations, approaches to data collection, and measures of cognition, functional abilities, neuropsychiatric status, quality of life, health economics, safety, and drug utilization. Postapproval studies that capture these considerations will be important to provide standardized data on AD treatment effectiveness and safety in real-world settings.
Collapse
Affiliation(s)
- James E. Galvin
- Comprehensive Center for Brain HealthDepartment of NeurologyUniversity of Miami Miller School of MedicineBoca RatonFloridaUSA
| | - Jeffrey L. Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | | | | | - Ricardo F. Allegri
- Instituto de Investigaciones Neurológicas FleniBuenos AiresArgentina
- Departamento de NeurocienciasUniversidad De La Costa (CUC), BarranquillaAtlánticoColombia
| | - Alireza Atri
- Banner Sun Health Research InstituteSun CityArizonaUSA
- Center for Brain/Mind Medicine, Department of NeurologyBrigham and Women's Hospital – Main CampusBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Howard Chertkow
- Rotman Research Institute, Baycrest Health SciencesTorontoOntarioCanada
| | - Claire Paquet
- Université de Paris GHU AP‐HP Nord Lariboisière HospitalParisFrance
| | - Verna R. Porter
- Pacific Brain Health Center, Pacific Neuroscience InstituteSanta MonicaCaliforniaUSA
- Saint John's Cancer InstituteSanta MonicaCaliforniaUSA
| | | | - Sietske A. M. Sikkes
- Alzheimer Center AmsterdamAmsterdam University Medical CenterAmsterdamThe Netherlands
- Department of Clinical, Neuro‐ and Developmental PsychologyVrije Universiteit (VU) AmsterdamAmsterdamThe Netherlands
| | | | | | | |
Collapse
|
5
|
O'Leary TP, Brown RE. Age-related changes in species-typical behaviours in the 5xFAD mouse model of Alzheimer's disease from 4 to 16 months of age. Behav Brain Res 2024; 465:114970. [PMID: 38531510 DOI: 10.1016/j.bbr.2024.114970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 03/28/2024]
Abstract
Alzheimer's disease (AD) patients show age-related decreases in the ability to perform activities of daily living and the decline in these activities is related to the severity of neurobiological deterioration underlying the disease. The 5xFAD mouse model of AD shows age-related impairments in sensory- motor and cognitive function, but little is known about changes in species-typical behaviours that may model activities of daily living in AD patients. Therefore, we examined species-typical behaviours used as indices of exploration (rearing) and compulsivity (grooming) across six tests of anxiety-like behaviour or motor function in female 5xFAD mice from 3 to 16 months of age. Robust decreases in rearing were found in 5xFAD mice across all tests after 9 months of age, although few differences were observed in grooming. A fine-scale analysis of grooming, however, revealed a previously unresolved and spatially restricted pattern of grooming in 5xFAD mice at 13-16 months of age. We then examined changes in species-typical behaviours in the home-cage, and show impaired nest building in 5xFAD mice at all ages tested. Lastly, we examined the relationship between reduced species typical behaviours in 5xFAD mice and the presentation of freezing behaviour, a commonly used measure of memory for conditioned fear. These results showed that along with cognitive and sensory-motor behaviour, 5xFAD mice have robust age-related impairments in species-typical behaviours. Therefore, species typical behaviours in 5xFAD mice may help to model the decline in activities of daily living observed in AD patients, and may provide useful behavioural phenotypes for evaluating the pre-clinical efficacy of novel therapeutics for AD.
Collapse
Affiliation(s)
- Timothy P O'Leary
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
6
|
Giri PM, Banerjee A, Ghosal A, Layek B. Neuroinflammation in Neurodegenerative Disorders: Current Knowledge and Therapeutic Implications. Int J Mol Sci 2024; 25:3995. [PMID: 38612804 PMCID: PMC11011898 DOI: 10.3390/ijms25073995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
Neurodegenerative disorders (NDs) have become increasingly common during the past three decades. Approximately 15% of the total population of the world is affected by some form of NDs, resulting in physical and cognitive disability. The most common NDs include Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease. Although NDs are caused by a complex interaction of genetic, environmental, and lifestyle variables, neuroinflammation is known to be associated with all NDs, often leading to permanent damage to neurons of the central nervous system. Furthermore, numerous emerging pieces of evidence have demonstrated that inflammation not only supports the progression of NDs but can also serve as an initiator. Hence, various medicines capable of preventing or reducing neuroinflammation have been investigated as ND treatments. While anti-inflammatory medicine has shown promising benefits in several preclinical models, clinical outcomes are often questionable. In this review, we discuss various NDs with their current treatment strategies, the role of neuroinflammation in the pathophysiology of NDs, and the use of anti-inflammatory agents as a potential therapeutic option.
Collapse
Affiliation(s)
- Paras Mani Giri
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Anurag Banerjee
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Arpita Ghosal
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|
7
|
Bohn L, Drouin SM, McFall GP, Rolfson DB, Andrew MK, Dixon RA. Machine learning analyses identify multi-modal frailty factors that selectively discriminate four cohorts in the Alzheimer's disease spectrum: a COMPASS-ND study. BMC Geriatr 2023; 23:837. [PMID: 38082372 PMCID: PMC10714519 DOI: 10.1186/s12877-023-04546-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Frailty indicators can operate in dynamic amalgamations of disease conditions, clinical symptoms, biomarkers, medical signals, cognitive characteristics, and even health beliefs and practices. This study is the first to evaluate which, among these multiple frailty-related indicators, are important and differential predictors of clinical cohorts that represent progression along an Alzheimer's disease (AD) spectrum. We applied machine-learning technology to such indicators in order to identify the leading predictors of three AD spectrum cohorts; viz., subjective cognitive impairment (SCI), mild cognitive impairment (MCI), and AD. The common benchmark was a cohort of cognitively unimpaired (CU) older adults. METHODS The four cohorts were from the cross-sectional Comprehensive Assessment of Neurodegeneration and Dementia dataset. We used random forest analysis (Python 3.7) to simultaneously test the relative importance of 83 multi-modal frailty indicators in discriminating the cohorts. We performed an explainable artificial intelligence method (Tree Shapley Additive exPlanation values) for deep interpretation of prediction effects. RESULTS We observed strong concurrent prediction results, with clusters varying across cohorts. The SCI model demonstrated excellent prediction accuracy (AUC = 0.89). Three leading predictors were poorer quality of life ([QoL]; memory), abnormal lymphocyte count, and abnormal neutrophil count. The MCI model demonstrated a similarly high AUC (0.88). Five leading predictors were poorer QoL (memory, leisure), male sex, abnormal lymphocyte count, and poorer self-rated eyesight. The AD model demonstrated outstanding prediction accuracy (AUC = 0.98). Ten leading predictors were poorer QoL (memory), reduced olfaction, male sex, increased dependence in activities of daily living (n = 6), and poorer visual contrast. CONCLUSIONS Both convergent and cohort-specific frailty factors discriminated the AD spectrum cohorts. Convergence was observed as all cohorts were marked by lower quality of life (memory), supporting recent research and clinical attention to subjective experiences of memory aging and their potentially broad ramifications. Diversity was displayed in that, of the 14 leading predictors extracted across models, 11 were selectively sensitive to one cohort. A morbidity intensity trend was indicated by an increasing number and diversity of predictors corresponding to clinical severity, especially in AD. Knowledge of differential deficit predictors across AD clinical cohorts may promote precision interventions.
Collapse
Affiliation(s)
- Linzy Bohn
- Department of Psychology, University of Alberta, P217 Biological Sciences Building, Edmonton, AB, T6G 2E9, Canada.
- Neuroscience and Mental Health Institute, University of Alberta, 2-132 Li Ka Shing Center for Health Research Innovation, Edmonton, AB, T6G 2E1, Canada.
| | - Shannon M Drouin
- Department of Psychology, University of Alberta, P217 Biological Sciences Building, Edmonton, AB, T6G 2E9, Canada
- Neuroscience and Mental Health Institute, University of Alberta, 2-132 Li Ka Shing Center for Health Research Innovation, Edmonton, AB, T6G 2E1, Canada
| | - G Peggy McFall
- Department of Psychology, University of Alberta, P217 Biological Sciences Building, Edmonton, AB, T6G 2E9, Canada
- Neuroscience and Mental Health Institute, University of Alberta, 2-132 Li Ka Shing Center for Health Research Innovation, Edmonton, AB, T6G 2E1, Canada
| | - Darryl B Rolfson
- Department of Medicine, Division of Geriatric Medicine, University of Alberta, 13-135 Clinical Sciences Building, Edmonton, AB, T6G 2G3, Canada
| | - Melissa K Andrew
- Department of Medicine, Division of Geriatric Medicine, Dalhousie University, 5955 Veterans' Memorial Lane, Halifax, NS, B3H 2E1, Canada
| | - Roger A Dixon
- Department of Psychology, University of Alberta, P217 Biological Sciences Building, Edmonton, AB, T6G 2E9, Canada
- Neuroscience and Mental Health Institute, University of Alberta, 2-132 Li Ka Shing Center for Health Research Innovation, Edmonton, AB, T6G 2E1, Canada
| |
Collapse
|
8
|
Goverover Y, Salter A, DeLuca J. Assessing everyday functional activity in cognitively impaired people with multiple sclerosis: The use of Actual Reality TM. Mult Scler 2023; 29:1107-1117. [PMID: 37496331 DOI: 10.1177/13524585231185048] [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: 07/28/2023]
Abstract
BACKGROUND There is considerable evidence that persons with multiple sclerosis (PwMS) who experience cognitive impairments (CIs) are at risk of having significant limitations in activities of daily living (ADLs). However, ADL assessment often consists of proxies or self-report of ADLs. This study examined whether the performance of instrumental ADLs (I-ADL) is impaired in PwMS with and without CI. METHODS Participants included 72 PwMS and 48 matched healthy controls (HCs). PwMS were divided into MS-CI (n = 25) and MS-not-impaired (n = 47) groups based on the Brief International Cognitive Assessment for Multiple Sclerosis (BICAMS) scores. All participants performed the Actual RealityTM (AR) test, measuring I-ADL using authentic websites. RESULTS The MS-CI performed significantly worse on AR compared with HC and MS-not-impaired. In addition, the MS-not-impaired performed significantly worse than HC on AR. AR differentiates well between PwMS with and without CI. CONCLUSIONS While CI in MS results in significant limitations in the performance of I-ADL, PwMS who do not show evidence of CI can have limitations in I-ADL. AR assessment is a valid and reliable tool sensitive to CI. It should be used in addition to traditional cognitive assessments to detect early functional deterioration through the course of MS.
Collapse
Affiliation(s)
- Yael Goverover
- Department of Occupational Therapy, New York University, New York, NY, USA/Kessler Foundation, West Orange, NJ, USA
| | - Amber Salter
- The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John DeLuca
- Kessler Foundation, West Orange, NJ, USA/Department of Physical Medicine and Rehabilitation, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| |
Collapse
|
9
|
Bolton CJ, Khan OA, Moore EE, Pechman KR, Taylor Davis L, Liu D, Landman BA, Gifford KA, Hohman TJ, Jefferson AL. Baseline grey matter volumes and white matter hyperintensities predict decline in functional activities in older adults over a 5-year follow-up period. Neuroimage Clin 2023; 38:103393. [PMID: 37003129 PMCID: PMC10102557 DOI: 10.1016/j.nicl.2023.103393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/27/2023] [Accepted: 03/26/2023] [Indexed: 03/31/2023]
Abstract
INTRODUCTION Functional independence is an essential predictor of quality of life in aging, yet few accessible predictors of functional decline have been identified. This study examined associations between baseline structural neuroimaging markers and longitudinal functional status. METHODS Linear mixed effects models with follow-up time interaction terms related baseline grey matter volume and white matter hyperintensities (WMHs) to functional trajectory, adjusting for demographic and medical covariates. Subsequent models assessed interactions with cognitive status and apolipoprotein E (APOE) ε4 status. RESULTS Smaller baseline grey matter volumes, particularly in regions commonly affected by Alzheimer's disease (AD), and greater baseline WMHs were associated with faster functional decline over a mean 5-year follow-up. Effects were stronger in APOE-ε4 carriers on grey matter variables. Cognitive status interacted with most MRI variables. DISCUSSION Greater atrophy in AD-related regions and higher WMH burden at study entry were associated with faster functional decline, particularly among participants at increased risk of AD.
Collapse
Affiliation(s)
- Corey J Bolton
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Omair A Khan
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth E Moore
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kimberly R Pechman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - L Taylor Davis
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dandan Liu
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bennett A Landman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Katherine A Gifford
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Angela L Jefferson
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
10
|
Kim MK, Kang BH, Park JH, Ham SM, Park HY, Hong I. Longitudinal Changes in Dual Decline in Memory and Gait Speed Association with Instrumental Activities of Daily Living: Findings from the National Social Life, Health, and Aging Project. J Aging Health 2023; 35:256-264. [PMID: 36083125 DOI: 10.1177/08982643221125274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Objective: We investigated the association between dual decline (DD) (loss of memory and gait speed) and the instrumental activities of daily living (IADL) degeneration in older adults. Methods: Data were drawn from the National Social Life, Health, and Aging Project (NSHAP) reflecting changes over 5 years. This study used the NSHAP data set wave 2 (2010-2011, N = 3196) and wave 3 (2015-2016, N = 4377). Results: Data from 1640 participants were retrieved. There were 601 people with DD and 1039 people without-DD. The DD group had a 28.4% (95% CI = 1.013-1.626) greater risk of degrading in IADL than the without-DD group (odds ratio = 1.284, p < .05). Conclusion: Current research can be used when establishing intervention programs or policies that can prevent IADL degradation through simple memory training and walking activities for older adults living in the community.
Collapse
Affiliation(s)
- Mi Kyeong Kim
- Dept. of Occupational Therapy, Graduate School, 65448Yonsei University, Republic of Korea
| | - Byoung-Ho Kang
- Dept. of Occupational Therapy, Graduate School, 65448Yonsei University, Republic of Korea
| | - Ji Hyeun Park
- Dept. of Occupational Therapy, Graduate School, 65448Yonsei University, Republic of Korea
| | - Sun Mi Ham
- Dept. of Occupational Therapy, College of Software and Digital Healthcare Convergence, 65448Yonsei University, Republic of Korea
| | - Hae Yean Park
- Dept. of Occupational Therapy, College of Software and Digital Healthcare Convergence, 65448Yonsei University, Republic of Korea
| | - Ickpyo Hong
- Dept. of Occupational Therapy, College of Software and Digital Healthcare Convergence, 65448Yonsei University, Republic of Korea
| |
Collapse
|
11
|
Jutten RJ, Papp KV, Hendrix S, Ellison N, Langbaum JB, Donohue MC, Hassenstab J, Maruff P, Rentz DM, Harrison J, Cummings J, Scheltens P, Sikkes SAM. Why a clinical trial is as good as its outcome measure: A framework for the selection and use of cognitive outcome measures for clinical trials of Alzheimer's disease. Alzheimers Dement 2023; 19:708-720. [PMID: 36086926 PMCID: PMC9931632 DOI: 10.1002/alz.12773] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/29/2022] [Accepted: 07/22/2022] [Indexed: 11/11/2022]
Abstract
A crucial aspect of any clinical trial is using the right outcome measure to assess treatment efficacy. Compared to the rapidly evolved understanding and measurement of pathophysiology in preclinical and early symptomatic stages of Alzheimer's disease (AD), relatively less progress has been made in the evolution of clinical outcome assessments (COAs) for those stages. The current paper aims to provide a benchmark for the design and evaluation of COAs for use in early AD trials. We discuss lessons learned on capturing cognitive changes in predementia stages of AD, including challenges when validating novel COAs for those early stages and necessary evidence for their implementation in clinical trials. Moving forward, we propose a multi-step framework to advance the use of more effective COAs to assess clinically meaningful changes in early AD, which will hopefully contribute to the much-needed consensus around more appropriate outcome measures to assess clinical efficacy of putative treatments. HIGHLIGHTS: We discuss lessons learned on capturing cognitive changes in predementia stages of AD. We propose a framework for the design and evaluation of performance based cognitive tests for use in early AD trials. We provide recommendations to facilitate the implementation of more effective cognitive outcome measures in AD trials.
Collapse
Affiliation(s)
- Roos J. Jutten
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kathryn V. Papp
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | - Michael C. Donohue
- Alzheimer’s Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, California, USA
| | - Jason Hassenstab
- Knight Alzheimer Disease Research Center, Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Paul Maruff
- Cogstate Ltd., Melbourne, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia
| | - Dorene M. Rentz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - John Harrison
- Metis Cognition Ltd., Kilmington, UK
- Department of Psychiatry, Psychology & Neuroscience, King’s College London, UK
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, location VUmc, VU University, Amsterdam, The Netherlands
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, Nevada, USA
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, location VUmc, VU University, Amsterdam, The Netherlands
| | - Sietske A. M. Sikkes
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, location VUmc, VU University, Amsterdam, The Netherlands
- Department of Clinical, Neuro and Developmental Psychology, Faculty of Movement and Behavioral Sciences, VU University, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Becker S, Bode M, Brockmann K, Gasser T, Michaelis K, Solbrig S, Nuerk HC, Schulte C, Maetzler W, Zimmermann M, Berg D, Liepelt-Scarfone I. Cognitive-Driven Activities of Daily Living Impairment as a Predictor for Dementia in Parkinson Disease: A Longitudinal Cohort Study. Neurology 2022; 99:e2548-e2560. [PMID: 36240089 PMCID: PMC9754648 DOI: 10.1212/wnl.0000000000201201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/19/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES One-third of Parkinson disease (PD) patients with PD-mild cognitive impairment (PD-MCI) convert to dementia within a few years. Markers with a high prognostic value for dementia conversion are needed. Loss of everyday function primarily caused by cognitive dysfunction is the core criterion for the diagnosis of PD dementia, with an onset of more complex instrumental activities of daily living (IADL) dysfunction in the prodromal stage. This study evaluated the phenotype associated with cognitive IADL impairment and its predictive value for defining a high-risk group for PD dementia. METHODS An observational longitudinal study using cognitive and clinical scores in addition to genetic and CSF biomarkers was conducted. The Functional Activities Questionnaire quotient (cut-off ≥1), indicating more cognitive than motor-driven IADL impairment, defined cognitive IADL impairment status at baseline. Hazard ratios (HRs) were used to compare the impact of baseline classifications on dementia conversion. RESULTS Of 268 patients with PD assessed at baseline, 108 (40.3%) had PD-MCI. After a period of 3.78 ± 0.84 years, 164 (61.2%) patients were reassessed. At follow-up, 93 (56.7%) patients had no cognitive impairment, 54 (32.9%) fulfilled PD-MCI criteria, and 17 (10.4%) had developed dementia. The HR of baseline cognitive IADL impairment (n = 37) for dementia conversion was descriptively higher than for PD-MCI, but highest in patients with both markers (HR = 12.01, 95% CI 4.47-32.22, p < 0.001). In the follow-up sample, nearly half of the patients (n = 10, 47.6%) with baseline classification of cognitive IADL impairment and PD-MCI converted to dementia. Baseline status of cognitive IADL impairment was associated with higher nonmotor burden, worse cognitive performance, and more severe IADL progression over the study period. DISCUSSION The importance of differentiating between cognitive and motor aspects on ADL function in PD and monitoring cognitive ADL impairment in the prodromal stage of dementia is paramount. Patients with PD-MCI and cognitive IADL impairment may be a valuable target group for clinical trials aiming to slow down the development of dementia. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov NCT03687203. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that impairment of cognitive activities of daily living is associated with progression from MCI to dementia among patients with Parkinson disease.
Collapse
Affiliation(s)
- Sara Becker
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany
| | - Merle Bode
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany
| | - Kathrin Brockmann
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany
| | - Thomas Gasser
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany
| | - Katja Michaelis
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany
| | - Susanne Solbrig
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany
| | - Hans-Christoph Nuerk
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany
| | - Claudia Schulte
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany
| | - Walter Maetzler
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany
| | - Milan Zimmermann
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany
| | - Daniela Berg
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany
| | - Inga Liepelt-Scarfone
- From the Hertie Institute for Clinical Brain Research (S.B., M.B., K.B., T.G., K.M., S.S., C.S., M.Z., D.B., I.L.-S.), Department of Neurodegenerative Diseases, University of Tübingen, Germany; Department of Psychology (S.B.), University of Calgary, AB, Canada; German Center for Neurodegenerative Diseases (DZNE) (M.B., K.B., T.G., K.M., S.S., C.S., M.Z., I.L.-S.), University of Tübingen; Diagnostics and Cognitive Neuropsychology (H.-C.N.), Department of Psychology, Eberhard Karls University Tübingen, Department of Neurology (W.M., D.B.), Christian-Albrechts-University, Kiel; and IB-Hochschule (I.L.-S.), Stuttgart, Germany.
| |
Collapse
|
13
|
Lee D, Lee VMY, Hur SK. Manipulation of the diet-microbiota-brain axis in Alzheimer's disease. Front Neurosci 2022; 16:1042865. [PMID: 36408394 PMCID: PMC9672822 DOI: 10.3389/fnins.2022.1042865] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Several studies investigating the pathogenesis of Alzheimer's disease have identified various interdependent constituents contributing to the exacerbation of the disease, including Aβ plaque formation, tau protein hyperphosphorylation, neurofibrillary tangle accumulation, glial inflammation, and the eventual loss of proper neural plasticity. Recently, using various models and human patients, another key factor has been established as an influential determinant in brain homeostasis: the gut-brain axis. The implications of a rapidly aging population and the absence of a definitive cure for Alzheimer's disease have prompted a search for non-pharmaceutical tools, of which gut-modulatory therapies targeting the gut-brain axis have shown promise. Yet multiple recent studies examining changes in human gut flora in response to various probiotics and environmental factors are limited and difficult to generalize; whether the state of the gut microbiota in Alzheimer's disease is a cause of the disease, a result of the disease, or both through numerous feedback loops in the gut-brain axis, remains unclear. However, preliminary findings of longitudinal studies conducted over the past decades have highlighted dietary interventions, especially Mediterranean diets, as preventative measures for Alzheimer's disease by reversing neuroinflammation, modifying the intestinal and blood-brain barrier (BBB), and addressing gut dysbiosis. Conversely, the consumption of Western diets intensifies the progression of Alzheimer's disease through genetic alterations, impaired barrier function, and chronic inflammation. This review aims to support the growing body of experimental and clinical data highlighting specific probiotic strains and particular dietary components in preventing Alzheimer's disease via the gut-brain axis.
Collapse
Affiliation(s)
- Daniel Lee
- Middleton High School, Middleton, WI, United States
| | - Virginia M-Y. Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Seong Kwon Hur
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
14
|
Dubbelman MA, Verrijp M, Terwee CB, Jutten RJ, Postema MC, Barkhof F, Berckel BNM, Gillissen F, Teeuwen V, Teunissen C, van de Flier WM, Scheltens P, Sikkes SAM. Determining the Minimal Important Change of Everyday Functioning in Dementia: Pursuing Clinical Meaningfulness. Neurology 2022; 99:e954-e964. [PMID: 35641309 PMCID: PMC9502738 DOI: 10.1212/wnl.0000000000200781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/11/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Decline in everyday functioning is a key clinical change in Alzheimer disease and related disorders (ADRD). An important challenge remains the determination of what constitutes a clinically meaningful change in everyday functioning. We aimed to investigate this by establishing the minimal important change (MIC): the smallest amount of change that has a meaningful effect on patients' lives. We retrospectively investigated meaningful change in a memory clinic cohort. METHODS In the first, qualitative part of the study, community-recruited informal caregivers of patients with ADRD and memory clinic clinicians completed a survey in which they judged various situations representing changes in everyday functioning. Their judgments of meaningful change were used to determine thresholds for MIC, both for decline and improvement, on the Amsterdam Instrumental Activities of Daily Living (IADL) Questionnaire. In the second, quantitative part, we applied these values in an independent longitudinal cohort study of unselected memory clinic patients. RESULTS MIC thresholds were established at the average threshold of caregivers (N = 1,629; 62.4 ± 9.5 years; 77% female) and clinicians (N = 13): -2.2 points for clinically meaningful decline and +5.0 points for clinically meaningful improvement. Memory clinic patients (N = 230; 64.3 ± 7.7 years; 39% female; 60% dementia diagnosis) were followed for 1 year, 102 (45%) of whom showed a decline larger than the MIC, after a mean of 6.7 ± 3.5 months. Patients with a dementia diagnosis and more atrophy of the medial temporal lobe had larger odds (odds ratio [OR] = 3.4, 95% CI [1.5-7.8] and OR = 5.0, 95% CI [1.2-20.0], respectively) for passing the MIC threshold for decline than those with subjective cognitive complaints and no atrophy. DISCUSSION We were able to operationalize clinically meaningful decline in IADL by determining the MIC. The usefulness of the MIC was supported by our findings from the clinical sample that nearly half of a sample of unselected memory clinic patients showed a meaningful decline in less than a year. Disease stage and medial temporal atrophy were predictors of functional decline greater than the MIC. Our findings provide guidance in interpreting changes in IADL and may help evaluate treatment effects and monitor disease progression.
Collapse
Affiliation(s)
- Mark A Dubbelman
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands.
| | - Merike Verrijp
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| | - Caroline B Terwee
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| | - Roos J Jutten
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| | - Merel C Postema
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| | - Frederik Barkhof
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| | - Bart N M Berckel
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| | - Freek Gillissen
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| | - Vivianne Teeuwen
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| | - Charlotte Teunissen
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| | - Wiesje M van de Flier
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| | - Philip Scheltens
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| | - Sietske A M Sikkes
- From the Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (M.A.D., M.V., M.C.P., F.G., V.T., W.M.F., P.S., S.A.M.S.); Department of Epidemiology and Data Science, Amsterdam UMC, The Netherlands (C.B.T., W.M.F.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (R.J.J.); Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands (F.B., B.N.M.B.); Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.); Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands (C.T.); and Faculty of Behavioural and Movement Sciences (S.A.M.S.), Clinical Developmental Psychology & Clinical Neuropsychology, Vrije Universiteit Amsterdam, the Netherlands
| |
Collapse
|
15
|
Mattsson P, Cselényi Z, Andrée B, Borg J, Nag S, Halldin C, Farde L. Decreased 5-HT 1A binding in mild Alzheimer's disease - a PET study. Synapse 2022; 76:e22235. [PMID: 35587913 PMCID: PMC9285435 DOI: 10.1002/syn.22235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Decreased 5-HT1A receptor binding has been associated with Alzheimer's disease (AD) and interpreted as a consequence of neuron loss. The purpose of the present study was to compare [11 C]WAY100635 binding to the 5-HT1A receptor in hippocampus, entorhinal cortex, amygdala and pericalcarine cortex in mild AD patients and elderly controls. METHODS AD patients (n = 7) and elderly control subjects (n = 8) were examined with positron emission tomography (PET) and [11 C]WAY100635. PET data acquisition was performed with an ECAT EXACT HR system. Wavelet-aided parametric images of non-displaceable binding potential (BPND ) were generated using Logan's graphical analysis with cerebellum as reference region. Correction for partial volume effects (PVE) was performed with the Müller-Gärtner method (MG). Regions of interest (ROIs) were applied to the individual parametric images and the regional BPND was calculated as the average parametric voxel value within each ROI. Besides comparison between subject groups, correlations between BPND values and scores on Mini Mental State Examination (MMSE), Disability Assessment for Dementia (DAD), and Neuropsychiatric Inventory (NPI) were expressed by Pearson correlation coefficients. RESULTS Mean regional BPND was lower in AD patients compared to control subjects and the difference was statistically significant for hippocampus, entorhinal cortex and amygdala. A statistically significant correlation was obtained between hippocampal BPND values and DAD scores. CONCLUSION The results of the present study corroborate and extend previous findings of decreased 5-HT1A binding in AD and strengthen the support for 5-HT1A receptor PET as a tool for assessment of neurodegenerative changes in mild AD. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Patrik Mattsson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County, Stockholm, Sweden
| | - Zsolt Cselényi
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County, Stockholm, Sweden.,PET Science Centre, Personalized Medicine, R&D, AstraZeneca, Stockholm, Sweden
| | - Bengt Andrée
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County, Stockholm, Sweden
| | - Jacqueline Borg
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County, Stockholm, Sweden
| | - Sangram Nag
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County, Stockholm, Sweden
| | - Christer Halldin
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County, Stockholm, Sweden
| | - Lars Farde
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County, Stockholm, Sweden
| |
Collapse
|
16
|
Milošević V, Malobabić M, Stojanović I, Bašić J. Importance of a functional measure in the evaluation of patients in a memory clinic: Validation of the Serbian version of the Amsterdam Instrumental Activities of Daily Living Questionnaire. Clin Neurol Neurosurg 2022; 214:107165. [DOI: 10.1016/j.clineuro.2022.107165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 01/11/2022] [Accepted: 02/01/2022] [Indexed: 11/03/2022]
|
17
|
Custodio N, Montesinos R, Chambergo-Michilot D, Herrera-Perez E, Pintado-Caipa M, Seminario G W, Cuenca J, Mesía L, Failoc-Rojas VE, Diaz MM. A Functional Assessment Tool to Distinguish Controls From Alzheimer's Disease in Lima, Peru. Am J Alzheimers Dis Other Demen 2022; 37:15333175221104354. [PMID: 35656899 PMCID: PMC10581136 DOI: 10.1177/15333175221104354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND The Alzheimer's Disease Cooperative Study-Activities of Daily Living (ADCS-ADL) scale is a versatile functional assessment tool for patients with Alzheimer's disease (ad). We evaluated its performance in controls, Peruvians with MCI or AD. METHODS A cross-sectional study of older adults attending a neurology institute in Lima (Peru) with mild cognitive impairment (MCI), ad or cognitively healthy. Test-retest reliability (intraclass correlation coefficient, ICC; internal consistency, Cronbach's alpha) and validity were assessed. RESULTS We enrolled 276 individuals (ad: 113, MCI: 68, controls: 95) with no age, sex, educational level, and depressive symptom differences. Reliability was ideal (ICC: .996), and Cronbach's alpha was adequate (.937). The ADCS-ADL could not differentiate MCI from controls but did differentiate ad severity. The ADCS-ADL correlated highly with nearly all tools. CONCLUSIONS The ADCS-ADL scale is reliable in a population with ad in Lima, Peru. Future work may validate a tool for Peruvians with lower educational levels.
Collapse
Affiliation(s)
- Nilton Custodio
- Servicio de Neurología, Instituto Peruano de Neurociencias, Lima, Perú
- Unidad de Diagnóstico de Deterioro Cognitivo y Prevención de Demencia, Instituto Peruano de Neurociencias, Lima, Perú
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Perú
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima, Perú
| | - Rosa Montesinos
- Unidad de Diagnóstico de Deterioro Cognitivo y Prevención de Demencia, Instituto Peruano de Neurociencias, Lima, Perú
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Perú
- Servicio de Rehabilitación, Instituto Peruano de Neurociencias, Lima, Perú
| | | | - Eder Herrera-Perez
- Unidad de Diagnóstico de Deterioro Cognitivo y Prevención de Demencia, Instituto Peruano de Neurociencias, Lima, Perú
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Perú
- Grupo de Investigación Molident, Universidad San Ignacio de Loyola, Lima, Perú
| | - Maritza Pintado-Caipa
- Servicio de Neurología, Instituto Peruano de Neurociencias, Lima, Perú
- Unidad de Diagnóstico de Deterioro Cognitivo y Prevención de Demencia, Instituto Peruano de Neurociencias, Lima, Perú
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Perú
- Atlantic Fellow for Equity in Brain Health, Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Wendy Seminario G
- Servicio de Neurología, Instituto Peruano de Neurociencias, Lima, Perú
- Unidad de Diagnóstico de Deterioro Cognitivo y Prevención de Demencia, Instituto Peruano de Neurociencias, Lima, Perú
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Perú
| | - José Cuenca
- Unidad de Diagnóstico de Deterioro Cognitivo y Prevención de Demencia, Instituto Peruano de Neurociencias, Lima, Perú
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Perú
- Servicio de Neuropsicología, Instituto Peruano de Neurociencias, Lima, Peru
- Carrera de Psicología, Facultad de Ciencias de La Salud, Universidad Privada Del Norte, Lima, Perú
| | - Laura Mesía
- Unidad de Diagnóstico de Deterioro Cognitivo y Prevención de Demencia, Instituto Peruano de Neurociencias, Lima, Perú
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Perú
| | - Virgilio E Failoc-Rojas
- Unidad de Diagnóstico de Deterioro Cognitivo y Prevención de Demencia, Instituto Peruano de Neurociencias, Lima, Perú
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Perú
- Salud Mental, Universidad Privada Norbert Wiener, Lima, Perú
| | - Monica M Diaz
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Perú
| |
Collapse
|
18
|
Sapkota S, Ramirez J, Yhap V, Masellis M, Black SE. Brain atrophy trajectories predict differential functional performance in Alzheimer's disease: Moderations with apolipoprotein E and sex. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12244. [PMID: 34692981 PMCID: PMC8515221 DOI: 10.1002/dad2.12244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/09/2021] [Indexed: 12/30/2022]
Abstract
INTRODUCTION We examine whether distinct brain atrophy patterns (using brain parenchymal fraction [BPF]) differentially predict functional performance and decline in Alzheimer's disease (AD), and are independently moderated by (1) a key AD genetic risk marker (apolipoprotein E [APOE]), (2) sex, and (3) high-risk group (women APOE ɛ4 carriers). METHODS We used a 2-year longitudinal sample of AD patients (baseline N = 170; mean age = 71.3 [9.1] years) from the Sunnybrook Dementia Study. We applied latent class analysis, latent growth modeling, and path analysis. We aimed to replicate our findings (N = 184) in the Alzheimer's Disease Neuroimaging Initiative. RESULTS We observed that high brain atrophy class predicted lower functional performance and steeper decline. This association was moderated by APOE, sex, and high-risk group. Baseline findings as moderated by APOE and high-risk group were replicated. DISCUSSION Women APOE ɛ4 carriers may selectively be at a greater risk of functional impairment with higher brain atrophy.
Collapse
Affiliation(s)
- Shraddha Sapkota
- Hurvitz Brain Sciences Research ProgramSunnybrook Research InstituteSunnybrook Health Sciences CentreTorontoOntarioCanada
| | - Joel Ramirez
- Hurvitz Brain Sciences Research ProgramSunnybrook Research InstituteSunnybrook Health Sciences CentreTorontoOntarioCanada
| | - Vanessa Yhap
- Hurvitz Brain Sciences Research ProgramSunnybrook Research InstituteSunnybrook Health Sciences CentreTorontoOntarioCanada
| | - Mario Masellis
- Hurvitz Brain Sciences Research ProgramSunnybrook Research InstituteSunnybrook Health Sciences CentreTorontoOntarioCanada
- Department of Medicine (Neurology)University of TorontoTorontoOntarioCanada
| | - Sandra E. Black
- Hurvitz Brain Sciences Research ProgramSunnybrook Research InstituteSunnybrook Health Sciences CentreTorontoOntarioCanada
- Department of Medicine (Neurology)University of TorontoTorontoOntarioCanada
| | | |
Collapse
|
19
|
Waheed H. The neglected contributions of self-efficacy to older adults’ financial capacity. QUALITY IN AGEING AND OLDER ADULTS 2021. [DOI: 10.1108/qaoa-05-2021-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Purpose
An ageing population comes with its own set of challenges such as impaired financial capacity and resultant dependency on others to manage financial affairs. Dependency, in turn, as the evidence suggests, creates opportunities for financial exploitation of older adults. Related studies have primarily examined the clinical features and correlates of financial capacity or have attempted to develop its multidimensional measures. Both of which do little to resolve issues associated with impaired financial capacity. This paper aims to make a case for future researchers to assess older adults’ financial capacity from a non-clinical aspect.
Design/methodology/approach
Drawing on the notion of self-efficacy, as encapsulated within the social cognitive theory, this paper presents evidence from a host of different domains to demonstrate the potential contributions of self-efficacy to older adults’ financial capacity.
Findings
The contributions of self-efficacy in preserving older adults’ financial capacity appear to be much more profound than is currently acknowledged in the literature, thereby overlooking potentially promising and cost-effective interventions for autonomous ageing.
Originality/value
This paper presents a novel application of self-efficacy to autonomous ageing. Within this context, potential routes to the deployment of self-efficacy-based interventions are also discussed.
Collapse
|
20
|
Castagna A, Fabbo A, Manzo C, Lacava R, Ruberto C, Ruotolo G. A Retrospective Study on the Benefits of Combined Citicoline, Memantine, and Acetylcholinesterase Inhibitor Treatments in Older Patients Affected with Alzheimer's Disease. J Alzheimers Dis 2021; 79:1509-1515. [PMID: 33459645 DOI: 10.3233/jad-201211] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Background: Citicoline has been proven to have beneficial effects in patients with cognitive impairment. In previous studies, combined treatment with memantine and acetylcholinesterase inhibitors (AChEIs) maintained cognitive function in patients with Alzheimer's disease (AD) better than memantine or AChEIs alone. OBJECTIVE To evaluate the effectiveness and safety of a combination therapy of oral citicoline, memantine, and an AChEI in AD when compared with memantine and an AChEI without citicoline. METHODS This was a retrospective multi-centric case-control study, conducted in Italian Centers for Cognitive Impairment and Dementia. Overall, 170 patients were recruited (34.11%of men, mean age 76,81±4.93 years): 48.8%treated with memantine and donepezil; 48.2%with memantine and rivastigmine; 2.9%with memantine and galantamine. 89 patients (control-group) were treated with memantine and an AChEI, whereas 81 patients (case-group) were treated with oral citicoline 1000 mg/day added to memantine and an AChEI given orally. Cognitive functions, activities of daily living, instrumental activities of daily living, comorbidities, mood and behavioral disturbances were assessed at baseline, month 6, and month 12. RESULTS In the case group, MMSE score had a statistically significant increasing trend between T0 and T2 (14.88±2.95 versus 15.09±3.00; p = 0.040), whereas in the control group, MMSE score showed a statistically significant decrease trend (14.37±2.63 versus 14.03±2.92 p = 0.024). CONCLUSION In older patients with AD, a triple therapy with citicoline, memantine, and AChEI was more effective than memantine and AChEI without citicoline in maintaining the MMSE total score after 12 months.
Collapse
Affiliation(s)
- Alberto Castagna
- Azienda Sanitaria Provinciale Catanzaro, Primary Care Departiment, Center for Cognitive Disorders and Dementia, Catanzaro, Italy
| | - Andrea Fabbo
- Health Authority and Services of Modena (AUSL di Modena), Geriatric Service-Cognitive Disorders and Dementia Unit, Modena, Italy
| | - Ciro Manzo
- Azienda Sanitaria Locale Napoli 3 sud, Internal and Geriatric Medicine Department, Center for Cognitive Disorders and Dementia, Pomigliano d'Arco (Naples), Italy
| | - Roberto Lacava
- Azienda Sanitaria Provinciale Catanzaro, Primary Care Departiment, Center for Cognitive Disorders and Dementia, Catanzaro, Italy
| | - Carmen Ruberto
- Azienda Sanitaria Provinciale Catanzaro, Primary Care Departiment, Center for Cognitive Disorders and Dementia, Catanzaro, Italy
| | - Giovanni Ruotolo
- Azienda Ospedaliera Pugliese-Ciaccio, Geriatric Unit, Center for Cognitive Disorders and Dementia, Catanzaro, Italy
| |
Collapse
|
21
|
Goldeck D, Adriaensen W, Oettinger L, Vaes B, van Pottelbergh G, Degryse JM, Hamprecht K, Matheï C, Pawelec G. Cellular Immune Phenotypes and Worsening Scores of Frailty-Associated Parameters Over an 18-Month Period in the Very Old. J Gerontol A Biol Sci Med Sci 2021; 76:1356-1361. [PMID: 33780527 DOI: 10.1093/gerona/glab089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
Frailty has been related to inflammaging and certain immune parameters. In previous analyses of participants older than 80 years of age in the longitudinal BELFRAIL cohort study, the main focus was on T-cell phenotypes and the association with cytomegalovirus (CMV) serostatus and survival, finding that a CD4:CD8 ratio greater than 5 was associated with frailty, impaired activities of daily living (ADLs), and mortality (but only in women). Here, we phenotyped peripheral blood immune cells via multicolor flow cytometry and correlated these with the dynamics of changes in ADL, geriatric depression score, Mini-Mental State Examination, and Short Physical Performance Battery from baseline values over 18 months follow-up. We found that higher frequencies of B cells and late-differentiated CD8+ T cells at 18 months from baseline were associated with ADL impairment that had worsened over the preceding 18 months. There were no significant associations with monocyte, dendritic cell, or natural killer (NK) cell phenotypes. No associations with the Geriatric Depression Scale, the Mini-Mental State Examination, or the Short Physical Performance Battery were found. Thus, while these results do not establish causality, they suggest that certain adaptive immune, but not innate immune, parameters are associated with a worsened ADL in the very old.
Collapse
Affiliation(s)
| | - Wim Adriaensen
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Lilly Oettinger
- Department of Psychiatry and Psychotherapy, Section for Dementia Research, University of Tübingen, Germany
| | - Bert Vaes
- Department of Public Health and Primary Care, KU Leuven (KUL), Belgium
| | | | | | - Klaus Hamprecht
- Institute of Medical Virology and Epidemiology of Viral Disease, University Hospital, Eberhard-Karls University, Tübingen, Germany
| | - Catharina Matheï
- Department of Public Health and Primary Care, KU Leuven (KUL), Belgium
| | - Graham Pawelec
- Department of Immunology, University of Tübingen, Germany.,Health Sciences North Research Institute, Sudbury, Ontario, Canada
| |
Collapse
|
22
|
Arruda F, Rosselli M, Greig MT, Loewenstein DA, Lang M, Torres VL, Vélez-Uribe I, Conniff J, Barker WW, Curiel RE, Adjouadi M, Duara R. The Association Between Functional Assessment and Structural Brain Biomarkers in an Ethnically Diverse Sample With Normal Cognition, Mild Cognitive Impairment, or Dementia. Arch Clin Neuropsychol 2021; 36:51-61. [PMID: 32890393 DOI: 10.1093/arclin/acaa065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/13/2020] [Accepted: 07/27/2020] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To investigate the association between the functional activities questionnaire (FAQ) and brain biomarkers (bilateral hippocampal volume [HV], bilateral entorhinal volume [ERV], and entorhinal cortical thickness [ERT]) in cognitively normal (CN) individuals, mild cognitive impairment (MCI), or dementia. METHOD In total, 226 participants (137 females; mean age = 71.76, SD = 7.93; Hispanic Americans = 137; European Americans = 89) were assessed with a comprehensive clinical examination, a neuropsychological battery, a structural magnetic resonance imaging, and were classified as CN or diagnosed with MCI or dementia. Linear regression analyses examined the association between functional activities as measured by the FAQ on brain biomarkers, including HV, ERV, and ERT, controlling for age, education, global cognition, gender, and ethnicity. RESULTS The FAQ significantly predicted HV, ERV, and ERT for the entire sample. However, this association was not significant for ERV and ERT when excluding the dementia group. The FAQ score remained a significant predictor of HV for the non-dementia group. Age, education, gender, ethnicity, Montreal Cognitive Assessment score, and FAQ were also significant predictors of HV for the overall sample, suggesting that younger Hispanic females with fewer years of education, higher global mental status, and better functioning, were more likely to have larger HV. CONCLUSION FAQ scores were related to HV in older adults across clinical groups (CN, MCI, and dementia), but its association with the entorhinal cortex was driven by individuals with dementia. Demographic variables, including ethnicity, additionally influenced these associations.
Collapse
Affiliation(s)
- Fernanda Arruda
- Department of Psychology, Florida Atlantic University, Davie, FL, USA
| | - Mónica Rosselli
- Department of Psychology, Florida Atlantic University, Davie, FL, USA.,1Florida Alzheimer's Disease Research Center, Miami Beach, FL, USA
| | - Maria T Greig
- 1Florida Alzheimer's Disease Research Center, Miami Beach, FL, USA.,Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - David A Loewenstein
- 1Florida Alzheimer's Disease Research Center, Miami Beach, FL, USA.,Department of Psychiatry and Behavioral Sciences, Center for Cognitive Neuroscience and Aging, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Merike Lang
- Department of Psychology, Florida Atlantic University, Davie, FL, USA
| | - Valeria L Torres
- Department of Psychology, Florida Atlantic University, Davie, FL, USA
| | - Idaly Vélez-Uribe
- Department of Psychology, Florida Atlantic University, Davie, FL, USA
| | - Joshua Conniff
- Department of Psychology, Florida Atlantic University, Davie, FL, USA
| | - Warren W Barker
- 1Florida Alzheimer's Disease Research Center, Miami Beach, FL, USA.,Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Rosie E Curiel
- 1Florida Alzheimer's Disease Research Center, Miami Beach, FL, USA.,Department of Psychiatry and Behavioral Sciences, Center for Cognitive Neuroscience and Aging, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Malek Adjouadi
- 1Florida Alzheimer's Disease Research Center, Miami Beach, FL, USA.,Engineering Center, Florida International University, Miami, FL, USA
| | - Ranjan Duara
- 1Florida Alzheimer's Disease Research Center, Miami Beach, FL, USA.,Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL, USA
| |
Collapse
|
23
|
de Oliveira Silva F, Ferreira JV, Plácido J, Deslandes AC. Spatial navigation and dual-task performance in patients with Dementia that present partial dependence in instrumental activity of daily living. IBRO Rep 2020; 9:52-57. [PMID: 33336104 PMCID: PMC7733130 DOI: 10.1016/j.ibror.2020.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/27/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Instrumental activities of daily living (IADLs) ability impairments are clearly related to cognitive and motor decline, as well as to the progression of Dementia. However, more low-cost assessments are necessary to better understand the process of IADL in patients with Dementia. OBJECTIVE To compare cognitive, motor and cognitive-motor performance at different stages of dependence on IADL in patients with Dementia. METHODS Dementia patients (n = 53, age range: 63-94) and healthy older adults (n = 39, age range: 62-97) were included, and those with Dementia were separated into IADL 1 (n = 18), IADL 2 (n = 17), IADL 3 (n = 18). All groups performed cognitive (Trail making test A, semantic verbal fluency, and Stroop test), motor (sit to stand, aerobic steps, and 8-foot up-and-go), and cognitive-motor tests (dual-task, and spatial navigation). One-way ANOVA, Kruskal-Wallis, and Bonferroni post-hoc tests were used to compare groups. Also, an effect size (ES) has been applied to evaluate differences among the dementia groups while the healthy older adults were used as a reference group. RESULTS Only cognitive-motor and cognitive tests showed significant differences among groups (IADL 1 x IADL 2 x IADL 3). Compared with the healthy group, the ES analysis exposed that patients in different stages of IADL showed the worst performance on tests combining motor and cognitive demand, but not for motor and cognitive function separately. CONCLUSION Poor dual-task and spatial navigation abilities are present in partial dependence in IADL, and these tasks should be considered as a functionality screening tool in patients with Dementia.
Collapse
Affiliation(s)
| | | | - Jéssica Plácido
- Institute of Psychiatry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
24
|
Maresova P, Hruska J, Klimova B, Barakovic S, Krejcar O. Activities of Daily Living and Associated Costs in the Most Widespread Neurodegenerative Diseases: A Systematic Review. Clin Interv Aging 2020; 15:1841-1862. [PMID: 33061334 PMCID: PMC7538005 DOI: 10.2147/cia.s264688] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022] Open
Abstract
Nowadays, the population is rapidly ageing because of increasing life expectancy and decreasing birth rates. Thus, the purpose of this systematic review is to prepare a comprehensive overview which identifies the activities of daily living (ADLs) that are gradually reduced among patients with dementia, as well as explore the therapies applied in relation to dementia and how they effectively improve the quality of life (QoL) of patients and caregivers. Furthermore, we aim to summarise the ADL activities influenced by therapies and examine the treatment costs and care for patients so that recommendations for research and development (R&D) can be made to improve both the QoL of people with dementia and cost-saving measures. The research focuses on four selected neurodegenerative diseases: Alzheimer, Parkinson, vascular dementia, and amyotrophic lateral sclerosis. Therefore, the peer-reviewed English written articles from 2014 to 2019 were searched between September 1 and December 13, 2019. Twenty-seven papers were included in the analysis. The results show that essential assistance occurs in connection with activities: eating, drinking, dressing, bathing, personal hygiene, use of the toilet, and transport. By contrast, shopping or cleaning is not addressed as much. A lower ability to take care of oneself is connected with poor patient health and higher social care costs because the patient requires care from external sources, such as home aid or nurse visits. The challenge that remains is to shift new knowledge from scientific disciplines and connect it with the needs of patients to remove legitimate barriers and increase the acceptance of new solutions by popularisation. Additionally, regarding the burden on caregivers, it would be appropriate to promote this area of education and employment so that family members can use formal caregivers, ensuring them free time and much-needed rest.
Collapse
Affiliation(s)
- Petra Maresova
- Department of Economics, Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove 500 03, Czech Republic
| | - Jan Hruska
- Department of Economics, Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove 500 03, Czech Republic
| | - Blanka Klimova
- Department of Applied Linguistics, Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove 500 03, Czech Republic
| | - Sabina Barakovic
- Faculty of Transport and Communications, University of Sarajevo, Sarajevo 71000, Bosnia and Herzegovina
| | - Ondrej Krejcar
- Center for Basic and Applied Science, Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove 500 03, Czech Republic
| |
Collapse
|
25
|
Halawa OA, Gatchel JR, Amariglio RE, Rentz DM, Sperling RA, Johnson KA, Marshall GA. Inferior and medial temporal tau and cortical amyloid are associated with daily functional impairment in Alzheimer's disease. Alzheimers Res Ther 2019; 11:14. [PMID: 30704519 PMCID: PMC6357436 DOI: 10.1186/s13195-019-0471-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/17/2019] [Indexed: 01/30/2023]
Abstract
BACKGROUND A decline in instrumental activities of daily living (IADL) correlates with the progression from mild cognitive impairment (MCI) to Alzheimer's disease (AD) dementia and has been associated with frontal and parietal hypometabolism, lower cerebrospinal fluid amyloid β1-42, and inferior temporal cortical thinning. Identifying the underlying biomarkers of functional decline will allow for the early identification of individuals at risk of disease progression. OBJECTIVE To investigate the association between IADL impairment and in vivo regional cerebral tau and cortical amyloid deposition across clinically normal (CN) elderly, MCI, and AD dementia. METHODS Fifty-one CN elderly, 30 MCI, and 9 AD dementia participants of the Alzheimer's Disease Neuroimaging Initiative (ADNI) underwent assessment of regional tau deposition with flortaucipir (FTP) positron emission tomography (PET). An aggregate of cortical amyloid burden was assessed by florbetapir PET. IADL were assessed using the Functional Activities Questionnaire (FAQ). Tau regions with unadjusted correlations of p ≤ 0.006 (Bonferroni correction) with FAQ were used to evaluate the cross-sectional association between FAQ (dependent variable) and regional cerebral tau deposition, amyloid burden, and tau-amyloid interaction in separate general linear regression models with backward elimination. Covariates included age, American National Adult Reading Test (AMNART) intelligence quotient (IQ), and Rey Auditory Verbal Learning Test (RAVLT) total learning. RESULTS Unadjusted correlations between FAQ and tau in the entorhinal cortex (EC) and inferior temporal cortex (IT) survived Bonferroni correction. FAQ was associated with the tau-amyloid interaction, such that in participants with greater amyloid burden, greater IADL impairment was associated with greater regional tau (EC tau × amyloid: partial r (pr) = 0.47, p < 0.001; IT tau × amyloid: pr = 0.54, p < 0.001). Significant associations were found when these regression models were repeated in symptomatic participants alone but not among CN participants. CONCLUSIONS Greater medial and inferior temporal tau and cortical amyloid burden were associated with greater IADL impairment in AD. Further elucidation of the biomarkers underlying the functional decline will allow for the early identification of individual at risk of disease progression.
Collapse
Affiliation(s)
| | - Jennifer R Gatchel
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, 02114, USA
- Division of Geriatric Psychiatry, McLean Hospital, Belmont, MA, 02478, USA
| | - Rebecca E Amariglio
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, 60 Fenwood Road, 9016P, Boston, MA, 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Dorene M Rentz
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, 60 Fenwood Road, 9016P, Boston, MA, 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Reisa A Sperling
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, 60 Fenwood Road, 9016P, Boston, MA, 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Keith A Johnson
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, 60 Fenwood Road, 9016P, Boston, MA, 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Gad A Marshall
- Harvard Medical School, Boston, MA, 02115, USA.
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, 60 Fenwood Road, 9016P, Boston, MA, 02115, USA.
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|