51
|
Imbimbo BP, Ippati S, Watling M, Imbimbo C. Role of monomeric amyloid-β in cognitive performance in Alzheimer's disease: Insights from clinical trials with secretase inhibitors and monoclonal antibodies. Pharmacol Res 2023; 187:106631. [PMID: 36586644 DOI: 10.1016/j.phrs.2022.106631] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
According to the β-amyloid (Aβ) hypothesis of Alzheimer's disease (AD), brain Aβ accumulation is the primary cascade event leading to cognitive deficit and dementia. Numerous anti-Aβ drugs either inhibiting production or aggregation of Aβ or stimulating its clearance have failed to show clinical benefit in large scale AD trials, with β- and γ-secretase inhibitors consistently worsening cognitive and clinical decline. In June 2021, the FDA approved aducanumab, an anti-Aβ monoclonal antibody for early AD based on its ability to reduce brain amyloid plaques, while two other amyloid-clearing antibodies (lecanemab and donanemab) have recently produced encouraging cognitive and clinical results. We reviewed AD trials using PubMed, meeting abstracts and ClinicalTrials.gov and evaluated the effects of such drugs on cerebrospinal fluid (CSF) Aβ levels, correlating them with cognitive effects. We found that β-secretase and γ-secretase inhibitors produce detrimental cognitive effects by significantly reducing CSF Aβ levels. We speculate that monoclonal antibodies targeting Aβ protofibrils, fibrils or plaques may improve cognitive performance in early AD by increasing soluble Aβ levels through Aβ aggregate disassembly and/or stabilization of existing Aβ monomers.These findings suggest that the real culprit in AD may be decreased levels of soluble monomeric Aβ due to sequestration into brain Aβ aggregates and plaques.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy.
| | - Stefania Ippati
- San Raffaele Scientific Institute, San Raffaele Hospital, 20132 Milan, Italy
| | - Mark Watling
- CNS & Pain Department, TranScrip Ltd, Reading, UK
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
52
|
Eruysal E, Ravdin L, Zhang C, Kamel H, Iadecola C, Ishii M. Sexually Dimorphic Association of Circulating Plasminogen Activator Inhibitor-1 Levels and Body Mass Index with Cerebrospinal Fluid Biomarkers of Alzheimer's Pathology in Preclinical Alzheimer's Disease. J Alzheimers Dis 2023; 91:1073-1083. [PMID: 36565112 PMCID: PMC10518184 DOI: 10.3233/jad-220686] [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: 12/24/2022]
Abstract
BACKGROUND Plasminogen activator inhibitor-1 (PAI-1), an inhibitor of fibrinolysis that is associated with adiposity, has been implicated in Alzheimer's disease (AD) pathogenesis. However, whether circulating PAI-1 levels are altered during preclinical AD remains unclear. OBJECTIVE To measure plasma PAI-1 levels in cognitively normal cerebrospinal fluid (CSF) AD biomarker positive and biomarker negative participants and to examine the association of plasma PAI-1 levels with CSF AD biomarkers and Mini-Mental State Examination (MMSE) scores. METHODS In this cross-sectional study, plasma PAI-1 levels were measured in 155 cognitively normal (Clinical Dementia Rating, CDR 0) non-obese older adults. 29 men and 26 women were classified as preclinical AD by previously established CSF tau/Aβ42 criteria. All analyses were sex stratified due to reported sex differences in PAI-1 expression. RESULTS Plasma PAI-1 levels were associated with body mass index (BMI) but not age in men and women. In men, plasma PAI-1 levels and BMI were lower in preclinical AD compared to control. Plasma PAI-1 levels were positively associated with CSF amyloid-β42 (Aβ42) and CSF Aβ42/Aβ40 and negatively associated with CSF tau/Aβ42, while BMI was positively associated with CSF Aβ42 and negatively associated with CSF p-tau181 and CSF tau/Aβ42. In women, plasma PAI-1 levels and BMI were similar between preclinical AD and control and were not associated with CSF AD biomarkers. For men and women, plasma PAI-1 levels and BMI were not associated with MMSE scores. CONCLUSION These findings suggest that there are significant sex differences in the systemic metabolic changes seen in the preclinical stage of AD.
Collapse
Affiliation(s)
- Emily Eruysal
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Lisa Ravdin
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Cenai Zhang
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Hooman Kamel
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Makoto Ishii
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
53
|
Sidenkova A, Calabrese V, Tomasello M, Fritsch T. Subjective cognitive decline and cerebral-cognitive reserve in late age. TRANSLATIONAL MEDICINE OF AGING 2023; 7:137-147. [DOI: 10.1016/j.tma.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2024] Open
|
54
|
Long JM, Coble DW, Xiong C, Schindler SE, Perrin RJ, Gordon BA, Benzinger TLS, Grant E, Fagan AM, Harari O, Cruchaga C, Holtzman DM, Morris JC. Preclinical Alzheimer's disease biomarkers accurately predict cognitive and neuropathological outcomes. Brain 2022; 145:4506-4518. [PMID: 35867858 PMCID: PMC10200309 DOI: 10.1093/brain/awac250] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/30/2022] [Accepted: 07/20/2022] [Indexed: 01/24/2023] Open
Abstract
Alzheimer's disease biomarkers are widely accepted as surrogate markers of underlying neuropathological changes. However, few studies have evaluated whether preclinical Alzheimer's disease biomarkers predict Alzheimer's neuropathology at autopsy. We sought to determine whether amyloid PET imaging or CSF biomarkers accurately predict cognitive outcomes and Alzheimer's disease neuropathological findings. This study included 720 participants, 42-91 years of age, who were enrolled in longitudinal studies of memory and aging in the Washington University Knight Alzheimer Disease Research Center and were cognitively normal at baseline, underwent amyloid PET imaging and/or CSF collection within 1 year of baseline clinical assessment, and had subsequent clinical follow-up. Cognitive status was assessed longitudinally by Clinical Dementia Rating®. Biomarker status was assessed using predefined cut-offs for amyloid PET imaging or CSF p-tau181/amyloid-β42. Subsequently, 57 participants died and underwent neuropathologic examination. Alzheimer's disease neuropathological changes were assessed using standard criteria. We assessed the predictive value of Alzheimer's disease biomarker status on progression to cognitive impairment and for presence of Alzheimer's disease neuropathological changes. Among cognitively normal participants with positive biomarkers, 34.4% developed cognitive impairment (Clinical Dementia Rating > 0) as compared to 8.4% of those with negative biomarkers. Cox proportional hazards modelling indicated that preclinical Alzheimer's disease biomarker status, APOE ɛ4 carrier status, polygenic risk score and centred age influenced risk of developing cognitive impairment. Among autopsied participants, 90.9% of biomarker-positive participants and 8.6% of biomarker-negative participants had Alzheimer's disease neuropathological changes. Sensitivity was 87.0%, specificity 94.1%, positive predictive value 90.9% and negative predictive value 91.4% for detection of Alzheimer's disease neuropathological changes by preclinical biomarkers. Single CSF and amyloid PET baseline biomarkers were also predictive of Alzheimer's disease neuropathological changes, as well as Thal phase and Braak stage of pathology at autopsy. Biomarker-negative participants who developed cognitive impairment were more likely to exhibit non-Alzheimer's disease pathology at autopsy. The detection of preclinical Alzheimer's disease biomarkers is strongly predictive of future cognitive impairment and accurately predicts presence of Alzheimer's disease neuropathology at autopsy.
Collapse
Affiliation(s)
- Justin M Long
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Dean W Coble
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Chengjie Xiong
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Suzanne E Schindler
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Richard J Perrin
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Brian A Gordon
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Elizabeth Grant
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Anne M Fagan
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Oscar Harari
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Carlos Cruchaga
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - David M Holtzman
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - John C Morris
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| |
Collapse
|
55
|
Luo J, Agboola F, Grant E, Morris JC, Masters CL, Albert MS, Johnson SC, McDade EM, Fagan AM, Benzinger TLS, Hassenstab J, Bateman RJ, Perrin RJ, Wang G, Li Y, Gordon B, Cruchaga C, Day GS, Levin J, Vöglein J, Ikeuchi T, Suzuki K, Allegri RF, Xiong C. Accelerated longitudinal changes and ordering of Alzheimer disease biomarkers across the adult lifespan. Brain 2022; 145:4459-4473. [PMID: 35925685 PMCID: PMC10200301 DOI: 10.1093/brain/awac238] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 04/15/2022] [Accepted: 06/11/2022] [Indexed: 01/25/2023] Open
Abstract
The temporal evolutions and relative orderings of Alzheimer disease biomarkers, including CSF amyloid-β42 (Aβ42), Aβ40, total tau (Tau) and phosphorylated tau181 (pTau181), standardized uptake value ratio (SUVR) from the molecular imaging of cerebral fibrillar amyloid-β with PET using the 11C-Pittsburgh Compound-B (PiB), MRI-based hippocampal volume and cortical thickness and cognition have been hypothesized but not yet fully tested with longitudinal data for all major biomarker modalities among cognitively normal individuals across the adult lifespan starting from 18 years. By leveraging a large harmonized database from 8 biomarker studies with longitudinal data from 2609 participants in cognition, 873 in MRI biomarkers, 519 in PET PiB imaging and 475 in CSF biomarkers for a median follow-up of 5-6 years, we estimated the longitudinal trajectories of all major Alzheimer disease biomarkers as functions of baseline age that spanned from 18 to 103 years, located the baseline age window at which the longitudinal rates of change accelerated and further examined possible modifying effects of apolipoprotein E (APOE) genotype. We observed that participants 18-45 years at baseline exhibited learning effects on cognition and unexpected directions of change on CSF and PiB biomarkers. The earliest acceleration of longitudinal change occurred for CSF Aβ42 and Aβ42/Aβ40 ratio (with an increase) and for Tau, and pTau181 (with a decrease) at the next baseline age interval of 45-50 years, followed by an accelerated increase for PiB SUVR at the baseline age of 50-55 years and an accelerated decrease for hippocampal volume at the baseline age of 55-60 years and finally by an accelerated decline for cortical thickness and cognition at the baseline age of 65-70 years. Another acceleration in the rate of change occurred at the baseline age of 65-70 years for Aβ42/Aβ40 ratio, Tau, pTau181, PiB SUVR and hippocampal volume. Accelerated declines in hippocampal volume and cognition continued after 70 years. For participants 18-45 years at baseline, significant increases in Aβ42 and Aβ42/Aβ40 ratio and decreases in PiB SUVR occurred in APOE ɛ4 non-carriers but not carriers. After age 45 years, APOE ɛ4 carriers had greater magnitudes than non-carriers in the rates of change for all CSF biomarkers, PiB SUVR and cognition. Our results characterize the temporal evolutions and relative orderings of Alzheimer disease biomarkers across the adult lifespan and the modification effect of APOE ɛ4. These findings may better inform the design of prevention trials on Alzheimer disease.
Collapse
Affiliation(s)
- Jingqin Luo
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center Biostatistics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Folasade Agboola
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth Grant
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Departments of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer’s Institute and Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Veterans Memorial Hospital, Madison, WI, USA
| | - Eric M McDade
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M Fagan
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason Hassenstab
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard J Perrin
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Departments of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Guoqiao Wang
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yan Li
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian Gordon
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Kazushi Suzuki
- Unit for Early and Exploratory Clinical Development, The University of Tokyo, Tokyo, Japan
| | - Ricardo F Allegri
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires, Argentina
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
56
|
Xiong C, Luo J, Schindler SE, Fagan AM, Benzinger T, Hassenstab J, Balls-Berry JE, Agboola F, Grant E, Moulder KL, Morris JC. Racial differences in longitudinal Alzheimer's disease biomarkers among cognitively normal adults. Alzheimers Dement 2022; 18:2570-2581. [PMID: 35218143 PMCID: PMC9402805 DOI: 10.1002/alz.12608] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/28/2021] [Accepted: 01/01/2022] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Longitudinal changes in Alzheimer's disease (AD) biomarkers, including cerebrospinal fluid (CSF) analytes, amyloid uptakes from positron emission tomography (PET), structural outcomes from magnetic resonance imaging (MRI), and cognition, have not been compared between Blacks and Whites. METHODS A total of 179 Blacks and 1180 Whites who were cognitively normal at baseline and had longitudinal data from at least one biomarker modality were analyzed for the annual rates of change. RESULTS CSF amyloid beta (Aβ)42/Aβ40 declined more slowly (P = .0390), and amyloid (PET) accumulated more slowly (P = .0157), in Blacks than Whites. CSF Aβ42 changed in opposite directions over time between Blacks and Whites (P = .0039). The annual increase in CSF total tau and phosphorylated tau181 for Blacks was about half of that for Whites. DISCUSSION Longitudinal racial differences in amyloid biomarkers are observed. It will be important to comprehensively and prospectively examine the effects of apolipoprotein E genotype and sociocultural factors on these differences.
Collapse
Affiliation(s)
- Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Jingqin Luo
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Siteman Cancer Center Biostatistics Core, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Suzanne E. Schindler
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M. Fagan
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie Benzinger
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason Hassenstab
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joyce E. Balls-Berry
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Folasade Agboola
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth Grant
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Krista L. Moulder
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C. Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Departments of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
57
|
Pettigrew C, Soldan A, Wang J, Wang M, Greenberg B, Albert M, Moghekar A. Longitudinal CSF Alzheimer's disease biomarker changes from middle age to late adulthood. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12374. [PMID: 36415591 PMCID: PMC9673459 DOI: 10.1002/dad2.12374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/05/2022] [Accepted: 10/11/2022] [Indexed: 11/19/2022]
Abstract
Introduction We examined longitudinal cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarker changes among cognitively normal individuals with 10.7 years follow-up, on average. Methods Analyses included 278 participants (M age = 57.5 years); 94 have progressed from normal cognition to mild cognitive impairment (MCI). Amyloid beta (Aβ)42/Aβ40, phosphorylated tau181 (p-tau181), and total tau (t-tau) were measured using automated electrochemiluminescence assays. Results Apolipoprotein E (APOE) ε4 carriers had lower baseline Aβ42/Aβ40, but longitudinal Aβ42/Aβ40 decreases did not differ by APOE ε4 after accounting for Aβ42/Aβ40 positivity. Lower baseline Aβ42/Aβ40 was associated with greater increases in tau (more strongly in males), and APOE ε4 genotype was associated with greater tau increases after reaching Aβ42/Aβ40 positivity. Participants who progressed to MCI had more abnormal biomarker levels and greater tau increases prior to MCI symptom onset. Biomarkers were more abnormal among older adults, but unrelated to sex or education. Discussion Our results confirm accelerated biomarker changes during preclinical AD and highlight the important role of amyloid levels in tau accelerations.
Collapse
Affiliation(s)
- Corinne Pettigrew
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Anja Soldan
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jiangxia Wang
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Mei‐Cheng Wang
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Barry Greenberg
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Marilyn Albert
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Abhay Moghekar
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | | |
Collapse
|
58
|
Aschenbrenner AJ, Li Y, Henson RL, Volluz K, Hassenstab J, Verghese P, West T, Meyer MR, Kirmess KM, Fagan AM, Xiong C, Holtzman D, Morris JC, Bateman RJ, Schindler SE. Comparison of plasma and CSF biomarkers in predicting cognitive decline. Ann Clin Transl Neurol 2022; 9:1739-1751. [PMID: 36183195 PMCID: PMC9639639 DOI: 10.1002/acn3.51670] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/07/2022] [Indexed: 11/07/2022] Open
Abstract
OBJECTIVES Concentrations of amyloid-β peptides (Aβ42/Aβ40) and neurofilament light (NfL) can be measured in plasma or cerebrospinal fluid (CSF) and are associated with Alzheimer's disease brain pathology and cognitive impairment. This study directly compared plasma and CSF measures of Aβ42/Aβ40 and NfL as predictors of cognitive decline. METHODS Participants were 65 years or older and cognitively normal at baseline with at least one follow-up cognitive assessment. Analytes were measured with the following types of assays: plasma Aβ42/Aβ40, immunoprecipitation-mass spectrometry; plasma NfL, Simoa; CSF Aβ42/Aβ40, automated immunoassay; CSF NfL plate-based immunoassay. Mixed effects models evaluated the global cognitive composite score over a maximum of 6 years as predicted by the fluid biomarkers. RESULTS Analyses included 371 cognitively normal participants, aged 72.7 ± 5.2 years (mean ± standard deviation) with an average length of follow-up of 3.9 ± 1.6 years. Standardized concentrations of biomarkers were associated with annualized cognitive change: plasma Aβ42/Aβ40, 0.014 standard deviations (95% confidence intervals 0.002 to 0.026); CSF Aβ42/Aβ40, 0.020 (0.008 to 0.032); plasma Nfl, -0.018 (-0.030 to -0.005); and CSF NfL, -0.024 (-0.036 to -0.012). Power analyses estimated that 266 individuals in each treatment arm would be needed to detect a 50% slowing of decline if identified by abnormal plasma measures versus 229 for CSF measures. INTERPRETATION Both plasma and CSF measures of Aβ42/Aβ40 and NfL predicted cognitive decline. A clinical trial that enrolled individuals based on abnormal plasma Aβ42/Aβ40 and NfL levels would require only a marginally larger cohort than if CSF measures were used.
Collapse
Affiliation(s)
- Andrew J. Aschenbrenner
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | - Yan Li
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Division of BiostatisticsWashington University School of MedicineSt. LouisMOUSA
| | - Rachel L. Henson
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | - Katherine Volluz
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | - Jason Hassenstab
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| | | | | | | | | | - Anne M. Fagan
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Chengjie Xiong
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Division of BiostatisticsWashington University School of MedicineSt. LouisMOUSA
| | - David Holtzman
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - John C. Morris
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMOUSA
| | - Suzanne E. Schindler
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
- Knight Alzheimer Disease Research CenterWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
59
|
Schweiger A, Rodebaugh TL, Lenze EJ, Keenoy K, Hassenstab J, Kloeckner J, Gettinger TR, Nicol GE. Mindfulness Training for Depressed Older Adults Using Smartphone Technology: Protocol for a Fully Remote Precision Clinical Trial. JMIR Res Protoc 2022; 11:e39233. [PMID: 36301604 PMCID: PMC9650569 DOI: 10.2196/39233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Precision medicine, optimized interventions, and access to care are catchphrases for the future of behavioral treatments. Progress has been slow due to the dearth of clinical trials that optimize interventions' benefits, individually tailor interventions to meet individual needs and preferences, and lead to rapid implementation after effectiveness is demonstrated. Two innovations have emerged to meet these challenges: fully remote trials and precision clinical trials. OBJECTIVE This paper provides a detailed description of Mindful MyWay, a study designed to test online mindfulness training in older adults with depression. Consistent with the concept of fully remote trials using a smartphone app, the study requires no in-person contact and can be conducted with participants anywhere in the United States. Based upon the precision medicine framework, the study assesses participants using high-frequency assessments of symptoms, cognitive performance, and patient preferences to both understand the individualized nature of treatment response and help individually tailor the intervention. METHODS Mindful MyWay is an open-label early-phase clinical trial for individuals 65 years and older with current depression. A smartphone app was developed to help coordinate the study, deliver the intervention, and evaluate the acceptability of the intervention, as well as predictors and outcomes of it. The curriculum for the fully remote intervention parallels the mindfulness-based stress reduction curriculum, a protocolized group-based mindfulness training that is typically provided in person. After consent and screening, participants download The Healthy Mind Lab mobile health smartphone app from the Apple App Store, allowing them to complete brief smartphone-based assessments of depressive symptoms and cognitive performance 4 times each day for 4 weeks prior to and after completing the intervention. The intervention consists of an introduction video and 10 weekly mindfulness training sessions, with the expectation to practice mindfulness at home daily. The app collects participant preference data throughout the 10-week intervention period; these high-frequency assessments identify participants' individually dynamic preferences toward the goal of optimizing the intervention in future iterations. RESULTS Participant recruitment and data collection began in March 2019. Final end point assessments will be collected in May 2022. The paper describes lessons learned regarding the critical role of early-phase testing prior to moving to a randomized trial. CONCLUSIONS The Mindful MyWay study is an exemplar of innovative clinical trial designs that use smartphone technology in behavioral and neuropsychiatric conditions. These include fully remote studies that can recruit throughout the United States, including hard-to-access areas, and collect high-frequency data, which is ideal for idiographic assessment and individualized intervention optimization. Our findings will be used to modify our methods and inform future randomized controlled trials within a precision medicine framework. TRIAL REGISTRATION ClinicalTrials.gov NCT03922217; https://clinicaltrials.gov/ct2/show/NCT03922217. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/39233.
Collapse
Affiliation(s)
- Abigail Schweiger
- Healthy Mind Lab, Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
- School of Social Work, Saint Louis University, Saint Louis, MO, United States
| | - Thomas L Rodebaugh
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, Saint Louis, MO, United States
| | - Eric J Lenze
- Healthy Mind Lab, Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
- mHealth Research Core, Washington University School of Medicine, Saint Louis, MO, United States
| | - Katie Keenoy
- mHealth Research Core, Washington University School of Medicine, Saint Louis, MO, United States
- Trial Care Unit, Center for Clinical Studies, Washington University School of Medicine, Saint Louis, MO, United States
| | - Jason Hassenstab
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, Saint Louis, MO, United States
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Jeanne Kloeckner
- Healthy Mind Lab, Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
| | - Torie R Gettinger
- Healthy Mind Lab, Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
- School of Social Work, Saint Louis University, Saint Louis, MO, United States
| | - Ginger E Nicol
- Healthy Mind Lab, Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
- mHealth Research Core, Washington University School of Medicine, Saint Louis, MO, United States
- Division of Child and Adolescent Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
60
|
Greenberg BD, Pettigrew C, Soldan A, Wang J, Wang MC, Darrow JA, Albert MS, Moghekar A. CSF Alzheimer Disease Biomarkers: Time-Varying Relationships With MCI Symptom Onset and Associations With Age, Sex, and ApoE4. Neurology 2022; 99:e1640-e1650. [PMID: 36216518 PMCID: PMC9559947 DOI: 10.1212/wnl.0000000000200953] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 05/24/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND AND OBJECTIVES This study aimed to examine whether baseline CSF measures of Alzheimer disease (AD)-related pathology are associated with the time to onset of mild cognitive impairment (MCI) and whether these associations differ by age, sex, Apolipoprotein E (ApoE4) status, and proximal (≤7 years) vs distal (>7 years) time to symptom onset. METHODS Measures of amyloid (Aβ1-42 and Aβ1-40), phospho-tau (ptau181), and total tau (t-tau) were determined from CSF samples obtained at baseline from participants in an ongoing longitudinal project, known as the Biomarkers for Older Controls at Risk for Alzheimer Disease study (BIOCARD) study. The fully automated, Lumipulse G immunoassay was used to analyze the specimens. Cox regression models were used to examine the relationship of baseline biomarker levels with time to symptom onset of MCI and interactions with age, sex, and ApoE allelic status in subjects who progressed from normal cognition to MCI. RESULTS Analyses included 273 participants from the BIOCARD cohort, who were cognitively normal and predominantly middle-aged at baseline, and have been followed for an average of 16 years (max = 23.6). During follow-up, 94 progressed to MCI (median time to symptom onset = 6.9 years). In Cox regression models, elevated ptau181 and t-tau levels were associated with time to MCI symptom onset if it occurred within 7 years of baseline (HR 1.386 and 1.329; p = 0.009 and 0.017, respectively), while a lower Aβ42/Aβ40 ratio was associated with symptom onset if it occurred >7 years from baseline (HR 0.596, p = 0.003). There were also significant 3-way CSF × age × sex interactions for ptau181 and Aβ42/Aβ40, with follow-up analyses indicating that associations between these biomarkers and progression to MCI were stronger among men than among women, but this difference between sexes diminished with increasing age. DISCUSSION The lengthy follow-up of BIOCARD participants permitted an examination of time-varying associations between CSF AD biomarkers with MCI symptom onset and the influence of sex, baseline age, and ApoE4 genotype on these associations. These factors may inform clinical trial enrollment strategies, or trial duration and outcomes, which may use these measures as surrogate markers of treatment response.
Collapse
Affiliation(s)
- Barry D Greenberg
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD.
| | - Corinne Pettigrew
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Anja Soldan
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jiangxia Wang
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Mei-Cheng Wang
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jacqueline A Darrow
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Marilyn S Albert
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Abhay Moghekar
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
61
|
Janssen O, Jansen WJ, Vos SJ, Boada M, Parnetti L, Gabryelewicz T, Fladby T, Molinuevo JL, Villeneuve S, Hort J, Epelbaum S, Lleó A, Engelborghs S, van der Flier WM, Landau S, Popp J, Wallin A, Scheltens P, Rikkert MO, Snyder PJ, Rowe C, Chételat G, Ruíz A, Marquié M, Chipi E, Wolfsgruber S, Heneka M, Boecker H, Peters O, Jarholm J, Rami L, Tort‐Merino A, Binette AP, Poirier J, Rosa‐Neto P, Cerman J, Dubois B, Teichmann M, Alcolea D, Fortea J, Sánchez‐Saudinós MB, Ebenau J, Pocnet C, Eckerström M, Thompson L, Villemagne V, Buckley R, Burnham S, Delarue M, Freund‐Levi Y, Wallin ÅK, Ramakers I, Tsolaki M, Soininen H, Hampel H, Spiru L, Tijms B, Ossenkoppele R, Verhey FRJ, Jessen F, Visser PJ. Characteristics of subjective cognitive decline associated with amyloid positivity. Alzheimers Dement 2022; 18:1832-1845. [PMID: 34877782 PMCID: PMC9786747 DOI: 10.1002/alz.12512] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/21/2021] [Accepted: 09/22/2021] [Indexed: 01/28/2023]
Abstract
INTRODUCTION The evidence for characteristics of persons with subjective cognitive decline (SCD) associated with amyloid positivity is limited. METHODS In 1640 persons with SCD from 20 Amyloid Biomarker Study cohort, we investigated the associations of SCD-specific characteristics (informant confirmation, domain-specific complaints, concerns, feelings of worse performance) demographics, setting, apolipoprotein E gene (APOE) ε4 carriership, and neuropsychiatric symptoms with amyloid positivity. RESULTS Between cohorts, amyloid positivity in 70-year-olds varied from 10% to 76%. Only older age, clinical setting, and APOE ε4 carriership showed univariate associations with increased amyloid positivity. After adjusting for these, lower education was also associated with increased amyloid positivity. Only within a research setting, informant-confirmed complaints, memory complaints, attention/concentration complaints, and no depressive symptoms were associated with increased amyloid positivity. Feelings of worse performance were associated with less amyloid positivity at younger ages and more at older ages. DISCUSSION Next to age, setting, and APOE ε4 carriership, SCD-specific characteristics may facilitate the identification of amyloid-positive individuals.
Collapse
Affiliation(s)
- Olin Janssen
- Alzheimer Centre LimburgDepartment of Psychiatry and NeuropsychologySchool for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - Willemijn J. Jansen
- Alzheimer Centre LimburgDepartment of Psychiatry and NeuropsychologySchool for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - Stephanie J.B. Vos
- Alzheimer Centre LimburgDepartment of Psychiatry and NeuropsychologySchool for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - Merce Boada
- Fundació ACEInstitut Català de Neurociències AplicadesFacultat de MedicinaUniversitat International de Catalunya‐BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Lucilla Parnetti
- Section of NeurologyCenter for Memory Disturbances – Lab. of Clinical NeurochemistryDepartment of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Tomasz Gabryelewicz
- Department of Neurodegenerative DisordersMossakowski Medical Research CentrePolish Academy of SciencesWarsawPoland
| | - Tormod Fladby
- Department of NeurologyAkershus University HospitalLorenskogNorway
| | - José Luis Molinuevo
- Alzheimer's Disease and Other Cognitive Disorders UnitNeurology Service, Hospital Clínic of BarcelonaAugust Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
| | - Sylvia Villeneuve
- Centre for Studies on Prevention of Alzheimer's Disease (StOP‐AD) CentreMontrealQuebecCanada
| | - Jakub Hort
- Department of NeurologySecond Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic,International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
| | - Stéphane Epelbaum
- AP‐HPHôpital de la Pitié SalpêtrièreInstitute of Memory and Alzheimer's Disease (IM2A)Centre of excellence of neurodegenerative disease (CoEN)Department of NeurologyParisFrance,Inserm Sorbonne UniversitéInriaAramis project‐teamParis Brain Institute – Institut du Cerveau (ICM)ParisFrance
| | - Alberto Lleó
- Neurology DepartmentHospital de Sant PauBarcelonaSpain
| | | | - Wiesje M. van der Flier
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Susan Landau
- Helen Wills Neuroscience InstituteUniversity of CaliforniaBerkeley, CaliforniaUSA
| | - Julius Popp
- Department of Geriatric PsychiatryPsychiatric University Hospital, ZürichSwitzerland,Old Age PsychiatryUniversity Hospital of LausanneLausanneSwitzerland
| | - Anders Wallin
- CSIRO Health & BiosecurityParkvilleVictoriaAustralia,Institute of Neuroscience and PhysiologySahlgrenska Academy at University of GothenburgMölndalSweden
| | - Philip Scheltens
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Marcel Olde Rikkert
- Department of Geriatric MedicineRadboud Alzheimer CenterRadboud University Medical CenterNijmegenThe Netherlands
| | - Peter J. Snyder
- Institute of Clinical MedicineUniversity of OsloOsloNorway,KingstonThe University of Rhode IslandRhode IslandUSA
| | - Chris Rowe
- Department of Molecular Imaging & TherapyAustin HealthMelbourneAustralia
| | - Gaël Chételat
- Institut National de la Sant. et de la Recherche M.dicale (Inserm)CaenFrance
| | - Agustin Ruíz
- Fundació ACEInstitut Català de Neurociències AplicadesFacultat de MedicinaUniversitat International de Catalunya‐BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Marta Marquié
- Fundació ACEInstitut Català de Neurociències AplicadesFacultat de MedicinaUniversitat International de Catalunya‐BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Elena Chipi
- Section of NeurologyCenter for Memory Disturbances – Lab. of Clinical NeurochemistryDepartment of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Steffen Wolfsgruber
- German Center For Neurodegenerative Diseases/Clinical ResearchDeutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE)Zentrum für klinische Forschung/AGCologneGermany,Department of Neurodegenerative Diseases and PsychiatryUniversity Hospital BonnBonnGermany
| | - Michael Heneka
- Department of Neurodegenerative Diseases and PsychiatryUniversity Hospital BonnBonnGermany
| | - Henning Boecker
- Functional Neuroimaging GroupDepartment of RadiologyUniversity Hospital BonnBonnGermany
| | - Oliver Peters
- Klinik für Psychiatrie und PsychotherapieCharité Universitätsmedizin Berlin ‐ CBFBerlinDeutschland
| | - Jonas Jarholm
- Department of NeurologyAkershus University HospitalLorenskogNorway
| | - Lorena Rami
- Alzheimer's Disease and Other Cognitive Disorders UnitNeurology Service, Hospital Clínic of BarcelonaAugust Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
| | - Adrià Tort‐Merino
- Alzheimer's Disease and Other Cognitive Disorders UnitNeurology Service, Hospital Clínic of BarcelonaAugust Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
| | - Alexa Pichet Binette
- Centre for Studies on Prevention of Alzheimer's Disease (StOP‐AD) CentreMontrealQuebecCanada
| | - Judes Poirier
- Centre for Studies on Prevention of Alzheimer's Disease (StOP‐AD) CentreMontrealQuebecCanada
| | - Pedro Rosa‐Neto
- Centre for Studies on Prevention of Alzheimer's Disease (StOP‐AD) CentreMontrealQuebecCanada
| | - Jiri Cerman
- Department of NeurologySecond Faculty of MedicineCharles University and Motol University HospitalPragueCzech Republic,International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
| | - Bruno Dubois
- AP‐HPHôpital de la Pitié SalpêtrièreInstitute of Memory and Alzheimer's Disease (IM2A)Centre of excellence of neurodegenerative disease (CoEN)Department of NeurologyParisFrance
| | - Marc Teichmann
- AP‐HPHôpital de la Pitié SalpêtrièreInstitute of Memory and Alzheimer's Disease (IM2A)Centre of excellence of neurodegenerative disease (CoEN)Department of NeurologyParisFrance
| | | | - Juan Fortea
- Neurology DepartmentHospital de Sant PauBarcelonaSpain
| | | | - Jarith Ebenau
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Cornelia Pocnet
- Old Age PsychiatryUniversity Hospital of LausanneLausanneSwitzerland
| | - Marie Eckerström
- Institute of Neuroscience and PhysiologySahlgrenska Academy at University of GothenburgMölndalSweden
| | - Louisa Thompson
- Institute of Clinical MedicineUniversity of OsloOsloNorway,KingstonThe University of Rhode IslandRhode IslandUSA
| | - Victor Villemagne
- Department of Molecular Imaging & TherapyAustin HealthMelbourneAustralia,Department of PsychiatryUniversity of PittsburghPittsburghUSA
| | - Rachel Buckley
- Brigham and Women's Hospital and Department of Neurology Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Samantha Burnham
- Section of NeurologyCenter for Memory Disturbances – Lab. of Clinical NeurochemistryDepartment of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Marion Delarue
- Institut National de la Sant. et de la Recherche M.dicale (Inserm)CaenFrance
| | - Yvonne Freund‐Levi
- Department of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Åsa K. Wallin
- Department of Clinical Sciences MalmöClinical Memory Research UnitLund UniversityLundSweden
| | - Inez Ramakers
- Alzheimer Centre LimburgDepartment of Psychiatry and NeuropsychologySchool for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - Magda Tsolaki
- Memory and Dementia Center3rd Department of Neurology“G Papanicolau” General HospitalAristotle University of ThessalonikiThessalonikiGreece
| | - Hilkka Soininen
- Institute of Clinical MedicineNeurologyUniversity of Eastern FinlandKuopioFinland
| | - Harald Hampel
- GRC no 21, Alzheimer Precision Medicine (AMP)AP‐HPPitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
| | - Luiza Spiru
- Carol DAVILA University of Medicine and PharmacyBucharestRomania,Geriatrics‐ Gerontology and Old Age PsychiatryAlzheimer UnitAna Aslan International Foundation – Memory Center and Longevity MedicineBucharestRomania
| | | | | | | | - Betty Tijms
- Alzheimer Centre LimburgDepartment of Psychiatry and NeuropsychologySchool for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands,Clinical Memory Research UnitDepartment of Clinical SciencesMalmöLund UniversityLundSweden,Alzheimer's Disease and Other Cognitive Disorders UnitNeurology Service, Hospital Clínic of BarcelonaAugust Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
| | - Frans R. J. Verhey
- Alzheimer Centre LimburgDepartment of Psychiatry and NeuropsychologySchool for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - Frank Jessen
- Department of PsychiatryUniversity of CologneCologneGermany,German Center For Neurodegenerative Diseases/Clinical ResearchDeutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE)Zentrum für klinische Forschung/AGCologneGermany
| | - Pieter Jelle Visser
- Alzheimer Centre LimburgDepartment of Psychiatry and NeuropsychologySchool for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands,Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands,Department of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| |
Collapse
|
62
|
Deng Z, Jiang J, Wang J, Pan D, Zhu Y, Li H, Zhang X, Liu X, Xu Y, Li Y, Tang Y. Angiotensin Receptor Blockers Are Associated With a Lower Risk of Progression From Mild Cognitive Impairment to Dementia. Hypertension 2022; 79:2159-2169. [PMID: 35766029 DOI: 10.1161/hypertensionaha.122.19378] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Previous studies found that antihypertensive medications (AHMs) acting on the renin-angiotensin system had the potential to reduce the progression from mild cognitive impairment to dementia. However, it remains unclear whether this association differs between ACE (angiotensin-converting enzyme) inhibitors and angiotensin receptor blockers. METHODS We conducted a retrospective cohort study in the Alzheimer's Disease Neuroimaging Initiative among 403 participants with hypertension and mild cognitive impairment at baseline. Information on AHMs received during the follow-up period, including angiotensin receptor blockers, ACE inhibitors, beta-blockers, calcium channel blockers, and diuretics, were self-reported. Cox proportional hazards models adjusted for potential confounders were used in the time to event analysis with progression to dementia as outcome. RESULTS Of the 403 participants, the mean (SD) age was 74.0 (7.3) years, 152 (37.7%) were female, 158 (39.2%) progressed to dementia over a median follow-up time of 3.0 years. Angiotensin receptor blockers were associated with a lower risk of progression to dementia as compared to ACE inhibitors (adjusted hazard ratio=0.45 [95% CI, 0.25-0.81]; P=0.023), other classes of AHMs (beta-blockers, calcium channel blockers, diuretics; adjusted hazard ratio, 0.49 [95% CI, 0.27-0.89]; P=0.037), and none of AHMs (adjusted hazard ratio, 0.31 [95% CI, 0.16-0.58]; P=0.001). CONCLUSIONS In patients with hypertension and mild cognitive impairment, angiotensin receptor blockers were associated with a lower risk of progression to dementia compared with ACE inhibitors and other classes of AHMs. Our findings may have important implications for clinical practice but still warrant further investigations in larger prospective cohorts or clinical trials.
Collapse
Affiliation(s)
- Zhenhong Deng
- Department of Neurology (Z.D., J.J., J.W., D.P., Y.Z., H.L., X.Z.,X.L., Y.X., Y.L., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingru Jiang
- Department of Neurology (Z.D., J.J., J.W., D.P., Y.Z., H.L., X.Z.,X.L., Y.X., Y.L., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jia Wang
- Department of Neurology (Z.D., J.J., J.W., D.P., Y.Z., H.L., X.Z.,X.L., Y.X., Y.L., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dong Pan
- Department of Neurology (Z.D., J.J., J.W., D.P., Y.Z., H.L., X.Z.,X.L., Y.X., Y.L., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingying Zhu
- Department of Neurology (Z.D., J.J., J.W., D.P., Y.Z., H.L., X.Z.,X.L., Y.X., Y.L., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Division of Clinical Research Design (Y.Z., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Honghong Li
- Department of Neurology (Z.D., J.J., J.W., D.P., Y.Z., H.L., X.Z.,X.L., Y.X., Y.L., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoni Zhang
- Department of Neurology (Z.D., J.J., J.W., D.P., Y.Z., H.L., X.Z.,X.L., Y.X., Y.L., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohuan Liu
- Department of Neurology (Z.D., J.J., J.W., D.P., Y.Z., H.L., X.Z.,X.L., Y.X., Y.L., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongteng Xu
- Department of Neurology (Z.D., J.J., J.W., D.P., Y.Z., H.L., X.Z.,X.L., Y.X., Y.L., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Li
- Department of Neurology (Z.D., J.J., J.W., D.P., Y.Z., H.L., X.Z.,X.L., Y.X., Y.L., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Division of Clinical Research Design (Y.Z., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yamei Tang
- Department of Neurology (Z.D., J.J., J.W., D.P., Y.Z., H.L., X.Z.,X.L., Y.X., Y.L., Y.T.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China (Y.T.)
| | | |
Collapse
|
63
|
A genetically modified minipig model for Alzheimer’s disease with SORL1 haploinsufficiency. Cell Rep Med 2022; 3:100740. [PMID: 36099918 PMCID: PMC9512670 DOI: 10.1016/j.xcrm.2022.100740] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 04/20/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022]
Abstract
The established causal genes in Alzheimer’s disease (AD), APP, PSEN1, and PSEN2, are functionally characterized using biomarkers, capturing an in vivo profile reflecting the disease’s initial preclinical phase. Mutations in SORL1, encoding the endosome recycling receptor SORLA, are found in 2%–3% of individuals with early-onset AD, and SORL1 haploinsufficiency appears to be causal for AD. To test whether SORL1 can function as an AD causal gene, we use CRISPR-Cas9-based gene editing to develop a model of SORL1 haploinsufficiency in Göttingen minipigs, taking advantage of porcine models for biomarker investigations. SORL1 haploinsufficiency in young adult minipigs is found to phenocopy the preclinical in vivo profile of AD observed with APP, PSEN1, and PSEN2, resulting in elevated levels of β-amyloid (Aβ) and tau preceding amyloid plaque formation and neurodegeneration, as observed in humans. Our study provides functional support for the theory that SORL1 haploinsufficiency leads to endosome cytopathology with biofluid hallmarks of autosomal dominant AD. Minipig model of Alzheimer’s disease by CRISPR knockout of the causal gene SORL1 Young SORL1 het minipigs phenocopy a preclinical CSF biomarker profile of individuals with AD SORL1 haploinsufficiency causes enlarged endosomes similar to neuronal AD pathology A minipig model bridging the translational gap between AD mouse models and affected individuals
Collapse
|
64
|
Vakili O, Asili P, Babaei Z, Mirahmad M, Keshavarzmotamed A, Asemi Z, Mafi A. Circular RNAs in Alzheimer's Disease: A New Perspective of Diagnostic and Therapeutic Targets. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 22:CNSNDDT-EPUB-125997. [PMID: 36043720 DOI: 10.2174/1871527321666220829164211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/06/2022] [Accepted: 06/20/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Circular RNAs (circRNAs), as covalently closed single-stranded noncoding RNA molecules, have been recently identified to involve in several biological processes, principally through targeting microRNAs. Among various neurodegenerative diseases (NDs), accumulating evidence has proposed key roles for circRNAs in the pathogenesis of Alzheimer's disease (AD); although the exact relationship between these RNA molecules and AD progression is not clear, they have been believed to mostly act as miRNA sponges or gene transcription modulators through correlating with multiple proteins, involved in the accumulation of Amyloid β (Aβ) peptides, as well as tau protein, as AD's pathological hallmark. More interestingly, circRNAs have also been reported to play diagnostic and therapeutic roles during AD progression. OBJECTIVE Literature review indicated that circRNAs could essentially contribute to the onset and development of AD. Thus, in the current review, the circRNAs' biogenesis and functions are addressed at first, and then the interplay between particular circRNAs and AD is comprehensively discussed. Eventually, the diagnostic and therapeutic significance of these noncoding RNAs is highlighted in brief. RESULTS A large number of circRNAs are expressed in the brain. Thereby, these RNA molecules are noticed as potential regulators of neural functions in healthy circumstances, as well as neurological disorders. Moreover, circRNAs have also been reported to have potential diagnostic and therapeutic capacities in relation to AD, the most prevalent ND. CONCLUSION CircRNAs have been shown to act as sponges for miRNAs, thereby regulating the function of related miRNAs, including oxidative stress, reduction of neuroinflammation, and the formation and metabolism of Aβ, all of which developed in AD. CircRNAs have also been proposed as biomarkers that have potential diagnostic capacities in AD. Despite these characteristics, the use of circRNAs as therapeutic targets and promising diagnostic biomarkers will require further investigation and characterization of the function of these RNA molecules in AD.
Collapse
Affiliation(s)
- Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pooria Asili
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Babaei
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Mirahmad
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
65
|
Oberstein TJ, Schmidt MA, Florvaag A, Haas AL, Siegmann EM, Olm P, Utz J, Spitzer P, Doerfler A, Lewczuk P, Kornhuber J, Maler JM. Amyloid-β levels and cognitive trajectories in non-demented pTau181-positive subjects without amyloidopathy. Brain 2022; 145:4032-4041. [PMID: 35973034 DOI: 10.1093/brain/awac297] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/11/2022] [Accepted: 07/24/2022] [Indexed: 11/13/2022] Open
Abstract
Phosphorylated Tau181 (pTau181) in cerebrospinal fluid (CSF) and recently in plasma has been associated with Alzheimer's disease. In the absence of amyloidopathy, individuals with increased total Tau levels and/or temporal lobe atrophy experience no or only mild cognitive decline compared with biomarker-negative controls, leading to the proposal to categorize this constellation as Suspected non-Alzheimer disease pathophysiology (SNAP). We investigated whether the characteristics of SNAP also applied to individuals with increased CSF-pTau181 without amyloidopathy. In this long-term observational study, 285 non-demented individuals, including 76 individuals with subjective cognitive impairment and 209 individuals with mild cognitive impairment, were classified based on their CSF-levels of pTau181 (T), total Tau (N), Amyloid-beta-(Aβ)-42 and Aβ42/Aβ40 ratio (A) into A + T+N±, A + T-N±, A-T + N±, and A-T-N-. The longitudinal analysis included 154 subjects with a follow-up of more than 12 months who were followed to a median of 4.6 years (interquartile range = 4.3 years). We employed linear mixed models on psychometric tests and region of interest analysis of structural MRI data. Cognitive decline and hippocampal atrophy rate were significantly higher in A + T+N ± compared to A-T + N±, whereas there was no difference between A-T + N ± and A-T-N-. Furthermore, there was no significant difference between A-T + N ± and controls in dementia risk (Hazard ratio 0.3, 95% confidence interval [0.1, 1.9]). However, A-T + N ± and A-T-N- could be distinguished based on their Aβ42 and Aβ40 levels. Both Aβ40 and Aβ42 levels were significantly increased in A-T + N ± compared to controls. Long term follow-up of A-T + N ± individuals revealed no evidence that this biomarker constellation was associated with dementia or more severe hippocampal atrophy rates compared to controls. However, because of the positive association of pTau181 with Aβ in the A-T + N ± group, a link to the pathophysiology of Alzheimer´s disease cannot be excluded in this case. We propose to refer to these individuals in the SNAP group as "pTau and Aβ surge with subtle deterioration" (PASSED). The investigation of the circumstances of simultaneous elevation of pTau and Aβ might provide a deeper insight into the process under which Aβ becomes pathological.
Collapse
Affiliation(s)
- Timo Jan Oberstein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Manuel Alexander Schmidt
- Institute of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Florvaag
- Department of Radiology and Nuclear Medicine, Klinikum Nuremberg, Nuremberg, Germany
| | - Anna Lena Haas
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Eva Maria Siegmann
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Pauline Olm
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Janine Utz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Spitzer
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arnd Doerfler
- Institute of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Neurodegeneration Diagnostics, Medical University of Bialystok, University Hospital of Bialystok, Bialystok, Poland.,Department of Biochemical Diagnostics, University Hospital of Bialystok, Bialystok, Poland
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Juan Manuel Maler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
66
|
Kang DW, Wang SM, Um YH, Kim NY, Lee CU, Lim HK. Associations Between Sub-Threshold Amyloid-β Deposition, Cortical Volume, and Cognitive Function Modulated by APOE ɛ4 Carrier Status in Cognitively Normal Older Adults. J Alzheimers Dis 2022; 89:1003-1016. [PMID: 35964194 PMCID: PMC9535581 DOI: 10.3233/jad-220427] [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] [Indexed: 11/21/2022]
Abstract
Background: There has been renewed interest in the deteriorating effects of sub-threshold amyloid-β (Aβ) accumulation in Alzheimer’s disease (AD). Despite evidence suggesting a synergistic interaction between the APOE ɛ4 allele and Aβ deposition in neurodegeneration, few studies have investigated the modulatory role of this allele in sub-threshold Aβ deposition during the preclinical phase. Objective: We aimed to explore the differential effect of the APOE ɛ4 carrier status on the association between sub-threshold Aβ deposition, cortical volume, and cognitive performance in cognitively normal older adults (CN). Methods: A total of 112 CN with sub-threshold Aβ deposition was included in the study. Participants underwent structural magnetic resonance imaging, [18F] flutemetamol PET-CT, and a neuropsychological battery. Potential interactions between APOE ɛ4 carrier status, Aβ accumulation, and cognitive function for cortical volume were assessed with whole-brain voxel-wise analysis. Results: We found that greater cortical volume was observed with higher regional Aβ deposition in the APOE ɛ4 carriers, which could be attributed to an interaction between the APOE ɛ4 carrier status and regional Aβ deposition in the posterior cingulate cortex/precuneus. Finally, the APOE ɛ4 carrier status-neuropsychological test score interaction demonstrated a significant effect on the gray matter volume of the left middle occipital gyrus. Conclusion: There might be a compensatory response to initiating Aβ in APOE ɛ4 carriers during the earliest AD stage. Despite its exploratory nature, this study offers some insight into recent interests concerning probabilistic AD modeling, focusing on the modulating role of the APOE ɛ4 carrier status during the preclinical period.
Collapse
Affiliation(s)
- Dong Woo Kang
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sheng-Min Wang
- Department of Psychiatry, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoo Hyun Um
- Department of Psychiatry, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Nak Young Kim
- Department of Psychiatry, Keyo Hospital, Uiwang, Republic of Korea
| | - Chang Uk Lee
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
67
|
Chino-Vilca B, Concepción Rodríguez-Rojo I, Torres-Simón L, Cuesta P, Carnes Vendrell A, Piñol-Ripoll G, Huerto R, Tahan N, Maestú F. Sex specific EEG signatures associated with cerebrospinal fluid biomarkers in mild cognitive impairment. Clin Neurophysiol 2022; 142:190-198. [DOI: 10.1016/j.clinph.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 06/07/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022]
|
68
|
Lozupone M, Berardino G, Mollica A, Sardone R, Dibello V, Zupo R, Lampignano L, Castellana F, Bortone I, Stallone R, Daniele A, Altamura M, Bellomo A, Solfrizzi V, Panza F. ALZT-OP1: An experimental combination regimen for the treatment of Alzheimer's Disease. Expert Opin Investig Drugs 2022; 31:759-771. [PMID: 35758153 DOI: 10.1080/13543784.2022.2095261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION For Alzheimer's disease (AD) treatment, US FDA granted accelerated approval for aducanumab due to its amyloid-β (Aβ)-lowering effects, notwithstanding the reported poor correlation between amyloid plaque reduction and clinical change for this drug. The diversification of drug targets appears to be the future of the AD field and from this perspective, drugs modulating microglia dysfunction and combination treatment regimens offer some promise. AREAS COVERED The aim of the present article was to provide a comprehensive review of ALZT-OP1 (cromolyn sodium plus ibuprofen), an experimental combination treatment regimen for AD, discussing their mechanisms of action targeting Aβ and neuroinflammation, examining the role of microglia in AD and offering our own insights on the role of present and alternative approaches directed toward neuroinflammation. EXPERT OPINION Enrolling high-risk participants with elevated brain amyloid could help to slow cognitive decline in secondary prevention trials during AD preclinical stages. Long-term follow-up indicated that non-steroidal anti-inflammatory drugs use begun when the brain was still normal may benefit these patients, suggesting that the timing of therapy could be crucial. However, previous clinical failures and the present incomplete understanding of the Aβ pathophysiological role in AD put this novel experimental combination regimen at substantial risk of failure.
Collapse
Affiliation(s)
- Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Berardino
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia
| | - Anita Mollica
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia
| | - Rodolfo Sardone
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| | - Vittorio Dibello
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Roberta Zupo
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| | - Luisa Lampignano
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| | - Fabio Castellana
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| | - Ilaria Bortone
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| | - Roberta Stallone
- Neuroscience and Education, Human Resources Excellence in Research, University of Foggia, Foggia, Italy
| | - Antonio Daniele
- Department of Neuroscience, Catholic University of Sacred Heart, Rome, Italy.,Neurology Unit, IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Mario Altamura
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia
| | - Vincenzo Solfrizzi
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro", Bari, Italy
| | - Francesco Panza
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology and Research Hospital IRCCS "S. De Bellis" Castellana Grotte, Bari, Italy
| |
Collapse
|
69
|
Incremental diagnostic value of 18F-Fluetemetamol PET in differential diagnoses of Alzheimer's Disease-related neurodegenerative diseases from an unselected memory clinic cohort. Sci Rep 2022; 12:10385. [PMID: 35725910 PMCID: PMC9209498 DOI: 10.1038/s41598-022-14532-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/08/2022] [Indexed: 11/08/2022] Open
Abstract
To evaluate the incremental diagnostic value of 18F-Flutemetamol PET following MRI measurements on an unselected prospective cohort collected from a memory clinic. A total of 84 participants was included in this study. A stepwise study design was performed including initial analysis (based on clinical assessments), interim analysis (revision of initial analysis post-MRI) and final analysis (revision of interim analysis post-18F-Flutemetamol PET). At each time of evaluation, every participant was categorized into SCD, MCI or dementia syndromal group and further into AD-related, non-AD related or non-specific type etiological subgroup. Post 18F-Flutemetamol PET, the significant changes were seen in the syndromal MCI group (57%, p < 0.001) involving the following etiological subgroups: AD-related MCI (57%, p < 0.01) and non-specific MCI (100%, p < 0.0001); and syndromal dementia group (61%, p < 0.0001) consisting of non-specific dementia subgroup (100%, p < 0.0001). In the binary regression model, amyloid status significantly influenced the diagnostic results of interim analysis (p < 0.01). 18F-Flutemetamol PET can have incremental value following MRI measurements, particularly reflected in the change of diagnosis of individuals with unclear etiology and AD-related-suspected patients due to the role in complementing AD-related pathological information.
Collapse
|
70
|
Abstract
PURPOSE OF REVIEW Alzheimer disease (AD) is the most common cause of dementia in adults (mid to late life), highlighting the importance of understanding the risk factors, clinical manifestations, and recent developments in diagnostic testing and therapeutics. RECENT FINDINGS Advances in fluid (CSF and blood-based) and imaging biomarkers are allowing for a more precise and earlier diagnosis of AD (relative to non-AD dementias) across the disease spectrum and in patients with atypical clinical features. Specifically, tau- and amyloid-related AD pathologic changes can now be measured by CSF, plasma, and positron emission tomography (PET) with good precision. Additionally, a better understanding of risk factors for AD has highlighted the need for clinicians to address comorbidities to maximize prevention of cognitive decline in those at risk or to slow decline in patients who are symptomatic. Recent clinical trials of amyloid-lowering drugs have provided not only some optimism that amyloid reduction or prevention may be beneficial but also a recognition that addressing additional targets will be necessary for significant disease modification. SUMMARY Recent developments in fluid and imaging biomarkers have led to the improved understanding of AD as a chronic condition with a protracted presymptomatic phase followed by the clinical stage traditionally recognized by neurologists. As clinical trials of potential disease-modifying therapies continue, important developments in the understanding of the disease will improve clinical care now and lead to more effective therapies in the near future.
Collapse
|
71
|
Platero C. Categorical predictive and disease progression modeling in the early stage of Alzheimer's disease. J Neurosci Methods 2022; 374:109581. [PMID: 35346695 DOI: 10.1016/j.jneumeth.2022.109581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/02/2022] [Accepted: 03/21/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND A preclinical stage of Alzheimer's disease (AD) precedes the symptomatic phases of mild cognitive impairment (MCI) and dementia, which constitutes a window of opportunities for preventive therapies or delaying dementia onset. NEW METHOD We propose to use categorical predictive models based on survival analysis with longitudinal data which are capable of determining subsets of markers to classify cognitively unimpaired (CU) subjects who progress into MCI/dementia or not. Subsequently, the proposed combination of markers was used to construct disease progression models (DPMs), which reveal long-term pathological trajectories from short-term clinical data. The proposed methodology was applied to a population recruited by the ADNI. RESULTS A very small subset of standard MRI-based data, CSF markers and cognitive measures was used to predict CU-to-MCI/dementia progression. The longitudinal data of these selected markers were used to construct DPMs using the algorithms of growth models by alternating conditional expectation (GRACE) and the latent time joint mixed effects model (LTJMM). The results show that the natural history of the proposed cognitive decline classifies the subjects well according to the clinical groups and shows a moderate correlation between the conversion times and their estimates by the algorithms. COMPARISON WITH EXISTING METHODS Unlike the training of the DPM algorithms without preselection of the markers, here, it is proposed to construct and evaluate the DPMs using the subsets of markers defined by the categorical predictive models. CONCLUSIONS The estimates of the natural history of the proposed cognitive decline from GRACE were more robust than those using LTJMM. The transition from normal to cognitive decline is mostly associated with an increase in temporal atrophy, worsening of clinical scores and pTAU/Aβ. Furthermore, pTAU/Aβ, Everyday Cognition score and the normalized volume of the entorhinal cortex show alterations of more than 20% fifteen years before the onset of cognitive decline.
Collapse
Affiliation(s)
- Carlos Platero
- Health Science Technology Group, Technical University of Madrid, Ronda de Valencia 3, 28012 Madrid, Spain
| |
Collapse
|
72
|
Bhagavati S. Commentary: Diagnostic Accuracy of Blood-Based Biomarker Panels: A Systematic Review. Front Aging Neurosci 2022; 14:895398. [PMID: 35572139 PMCID: PMC9099371 DOI: 10.3389/fnagi.2022.895398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 04/13/2022] [Indexed: 11/30/2022] Open
|
73
|
Varesi A, Carrara A, Pires VG, Floris V, Pierella E, Savioli G, Prasad S, Esposito C, Ricevuti G, Chirumbolo S, Pascale A. Blood-Based Biomarkers for Alzheimer's Disease Diagnosis and Progression: An Overview. Cells 2022; 11:1367. [PMID: 35456047 PMCID: PMC9044750 DOI: 10.3390/cells11081367] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 01/10/2023] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disease characterized by amyloid-β (Aβ) plaque deposition and neurofibrillary tangle accumulation in the brain. Although several studies have been conducted to unravel the complex and interconnected pathophysiology of AD, clinical trial failure rates have been high, and no disease-modifying therapies are presently available. Fluid biomarker discovery for AD is a rapidly expanding field of research aimed at anticipating disease diagnosis and following disease progression over time. Currently, Aβ1-42, phosphorylated tau, and total tau levels in the cerebrospinal fluid are the best-studied fluid biomarkers for AD, but the need for novel, cheap, less-invasive, easily detectable, and more-accessible markers has recently led to the search for new blood-based molecules. However, despite considerable research activity, a comprehensive and up-to-date overview of the main blood-based biomarker candidates is still lacking. In this narrative review, we discuss the role of proteins, lipids, metabolites, oxidative-stress-related molecules, and cytokines as possible disease biomarkers. Furthermore, we highlight the potential of the emerging miRNAs and long non-coding RNAs (lncRNAs) as diagnostic tools, and we briefly present the role of vitamins and gut-microbiome-related molecules as novel candidates for AD detection and monitoring, thus offering new insights into the diagnosis and progression of this devastating disease.
Collapse
Affiliation(s)
- Angelica Varesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- Almo Collegio Borromeo, 27100 Pavia, Italy
| | - Adelaide Carrara
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (A.C.); (V.F.)
| | - Vitor Gomes Pires
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA;
| | - Valentina Floris
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (A.C.); (V.F.)
| | - Elisa Pierella
- School of Medicine, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Gabriele Savioli
- Emergency Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Sakshi Prasad
- Faculty of Medicine, National Pirogov Memorial Medical University, 21018 Vinnytsya, Ukraine;
| | - Ciro Esposito
- Unit of Nephrology and Dialysis, ICS Maugeri, University of Pavia, 27100 Pavia, Italy;
| | - Giovanni Ricevuti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37129 Verona, Italy;
| | - Alessia Pascale
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
74
|
Li TR, Yang Q, Hu X, Han Y. Biomarkers and Tools for Predicting Alzheimer's Disease in the Preclinical Stage. Curr Neuropharmacol 2022; 20:713-737. [PMID: 34030620 PMCID: PMC9878962 DOI: 10.2174/1570159x19666210524153901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is the only leading cause of death for which no disease-modifying therapy is currently available. Over the past decade, a string of disappointing clinical trial results has forced us to shift our focus to the preclinical stage of AD, which represents the most promising therapeutic window. However, the accurate diagnosis of preclinical AD requires the presence of brain β- amyloid deposition determined by cerebrospinal fluid or amyloid-positron emission tomography, significantly limiting routine screening and diagnosis in non-tertiary hospital settings. Thus, an easily accessible marker or tool with high sensitivity and specificity is highly needed. Recently, it has been discovered that individuals in the late stage of preclinical AD may not be truly "asymptomatic" in that they may have already developed subtle or subjective cognitive decline. In addition, advances in bloodderived biomarker studies have also allowed the detection of pathologic changes in preclinical AD. Exosomes, as cell-to-cell communication messengers, can reflect the functional changes of their source cell. Methodological advances have made it possible to extract brain-derived exosomes from peripheral blood, making exosomes an emerging biomarker carrier and liquid biopsy tool for preclinical AD. The eye and its associated structures have rich sensory-motor innervation. In this regard, studies have indicated that they may also provide reliable markers. Here, our report covers the current state of knowledge of neuropsychological and eye tests as screening tools for preclinical AD and assesses the value of blood and brain-derived exosomes as carriers of biomarkers in conjunction with the current diagnostic paradigm.
Collapse
Affiliation(s)
- Tao-Ran Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Qin Yang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Xiaochen Hu
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, 50924, Germany
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China;,Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China;,National Clinical Research Center for Geriatric Disorders, Beijing, 100053, China;,School of Biomedical Engineering, Hainan University, Haikou, 570228, China;,Address correspondence to this author at the Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China; Tel: +86 13621011941; E-mail:
| |
Collapse
|
75
|
Cerebrospinal fluid tau levels are associated with abnormal neuronal plasticity markers in Alzheimer's disease. Mol Neurodegener 2022; 17:27. [PMID: 35346299 PMCID: PMC8962234 DOI: 10.1186/s13024-022-00521-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/13/2022] [Indexed: 12/15/2022] Open
Abstract
Background Increased total tau (t-tau) in cerebrospinal fluid (CSF) is a key characteristic of Alzheimer’s disease (AD) and is considered to result from neurodegeneration. T-tau levels, however, can be increased in very early disease stages, when neurodegeneration is limited, and can be normal in advanced disease stages. This suggests that t-tau levels may be driven by other mechanisms as well. Because tau pathophysiology is emerging as treatment target for AD, we aimed to clarify molecular processes associated with CSF t-tau levels. Methods We performed a proteomic, genomic, and imaging study in 1380 individuals with AD, in the preclinical, prodromal, and mild dementia stage, and 380 controls from the Alzheimer’s Disease Neuroimaging Initiative and EMIF-AD Multimodality Biomarker Discovery study. Results We found that, relative to controls, AD individuals with increased t-tau had increased CSF concentrations of over 400 proteins enriched for neuronal plasticity processes. In contrast, AD individuals with normal t-tau had decreased levels of these plasticity proteins and showed increased concentrations of proteins indicative of blood–brain barrier and blood-CSF barrier dysfunction, relative to controls. The distinct proteomic profiles were already present in the preclinical AD stage and persisted in prodromal and dementia stages implying that they reflect disease traits rather than disease states. Dysregulated plasticity proteins were associated with SUZ12 and REST signaling, suggesting aberrant gene repression. GWAS analyses contrasting AD individuals with and without increased t-tau highlighted several genes involved in the regulation of gene expression. Targeted analyses of SNP rs9877502 in GMNC, associated with t-tau levels previously, correlated in individuals with AD with CSF concentrations of 591 plasticity associated proteins. The number of APOE-e4 alleles, however, was not associated with the concentration of plasticity related proteins. Conclusions CSF t-tau levels in AD are associated with altered levels of proteins involved in neuronal plasticity and blood–brain and blood-CSF barrier dysfunction. Future trials may need to stratify on CSF t-tau status, as AD individuals with increased t-tau and normal t-tau are likely to respond differently to treatment, given their opposite CSF proteomic profiles. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-022-00521-3.
Collapse
|
76
|
Saridin FN, Chew KA, Reilhac A, Giyanwali B, Villaraza SG, Tanaka T, Scheltens P, van der Flier WM, Chen CLH, Hilal S. Cerebrovascular disease in Suspected Non-Alzheimer's Pathophysiology and cognitive decline over time. Eur J Neurol 2022; 29:1922-1929. [PMID: 35340085 DOI: 10.1111/ene.15337] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/15/2022] [Accepted: 03/19/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND The underlying cause of cognitive decline in individuals who are positive for biomarkers of neurodegeneration (N) but negative for biomarkers of amyloid-beta (A), designated as Suspected Non-Alzheimer's Pathophysiology (SNAP), remains unclear. We evaluate whether cerebrovascular disease (CeVD) is more prevalent in those with SNAP compared to A-N- and A+N+ individuals and whether CeVD is associated with cognitive decline over time in SNAP patients. METHODS A total of 216 individuals from a prospective memory clinic cohort [mean (SD) age, 72.7(7.3) years, 100 women (56.5%)] were included and were diagnosed as no cognitive impairment (NCI), cognitive impairment no dementia (CIND), Alzheimer's dementia (AD) or Vascular dementia (VaD). All individuals underwent clinical evaluation and neuropsychological assessment annually for up to 5 years. [11 C]-PiB or [18 F]-Flutafuranol-PET imaging was performed to ascertain amyloid-beta status. MRI was performed to assess neurodegeneration as measured by medial temporal atrophy≥2, as well as significant CeVD (sCeVD) burden, defined by cortical infarct count≥1, Fazekas-score≥2, lacune count≥2 or cerebral microbleed count≥2. RESULTS Of the 216 individuals, 50(23.1%) A-N+ were (SNAP), 93(43.1%) A-N-, 36(16.7%) A+N- and 37(17.1%) A+N+. A+N+ individuals were significantly older, while A+N+ and SNAP individuals were more likely to have dementia. The SNAP group had a higher prevalence of sCeVD (90.0%) compared to A-N-. Moreover, SNAP individuals with sCeVD had significantly steeper decline in global cognition compared to A-N- over 5 years (P=0.042). CONCLUSIONS These findings suggest that CeVD is a contributing factor to cognitive decline in SNAP. Therefore, SNAP-individuals should be carefully assessed and treated for CeVD.
Collapse
Affiliation(s)
- Francis Nicole Saridin
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Memory Aging & Cognition Centre, National University Health System, Singapore
| | - Kimberly Ann Chew
- Memory Aging & Cognition Centre, National University Health System, Singapore
| | - Anthonin Reilhac
- Clinical Imaging Research Centre, National University of Singapore, Singapore
| | - Bibek Giyanwali
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Tomotaka Tanaka
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Phillip Scheltens
- Department of Neurology & Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Wiesje M van der Flier
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, Netherlands
| | - Christopher Li Hsian Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Memory Aging & Cognition Centre, National University Health System, Singapore.,Department of Psychological Medicine, National University Hospital, Singapore
| | - Saima Hilal
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Memory Aging & Cognition Centre, National University Health System, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| |
Collapse
|
77
|
Kagerer SM, Schroeder C, van Bergen JMG, Schreiner SJ, Meyer R, Steininger SC, Vionnet L, Gietl AF, Treyer V, Buck A, Pruessmann KP, Hock C, Unschuld PG. Low Subicular Volume as an Indicator of Dementia-Risk Susceptibility in Old Age. Front Aging Neurosci 2022; 14:811146. [PMID: 35309894 PMCID: PMC8926841 DOI: 10.3389/fnagi.2022.811146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Hippocampal atrophy is an established Alzheimer’s Disease (AD) biomarker. Volume loss in specific subregions as measurable with ultra-high field magnetic resonance imaging (MRI) may reflect earliest pathological alterations. Methods Data from positron emission tomography (PET) for estimation of cortical amyloid β (Aβ) and high-resolution 7 Tesla T1 MRI for assessment of hippocampal subfield volumes were analyzed in 61 non-demented elderly individuals who were divided into risk-categories as defined by high levels of cortical Aβ and low performance in standardized episodic memory tasks. Results High cortical Aβ and low episodic memory interactively predicted subicular volume [F(3,57) = 5.90, p = 0.018]. The combination of high cortical Aβ and low episodic memory was associated with significantly lower subicular volumes, when compared to participants with high episodic memory (p = 0.004). Discussion Our results suggest that low subicular volume is linked to established indicators of AD risk, such as increased cortical Aβ and low episodic memory. Our data support subicular volume as a marker of dementia-risk susceptibility in old-aged non-demented persons.
Collapse
Affiliation(s)
- Sonja M. Kagerer
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Psychogeriatric Medicine, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Clemens Schroeder
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | | | - Simon J. Schreiner
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Rafael Meyer
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Stefanie C. Steininger
- Psychogeriatric Medicine, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Laetitia Vionnet
- Institute for Biomedical Engineering, University of Zurich and ETH Zürich, Zurich, Switzerland
| | - Anton F. Gietl
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Psychogeriatric Medicine, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Valerie Treyer
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alfred Buck
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Klaas P. Pruessmann
- Institute for Biomedical Engineering, University of Zurich and ETH Zürich, Zurich, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Neurimmune, Schlieren, Switzerland
| | - Paul G. Unschuld
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Psychogeriatric Medicine, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, University of Zurich and ETH Zürich, Zurich, Switzerland
- Geriatric Psychiatry, Department of Psychiatry, University Hospitals of Geneva, University of Geneva, Geneva, Switzerland
- *Correspondence: Paul G. Unschuld,
| |
Collapse
|
78
|
Pelkmans W, Vromen EM, Dicks E, Scheltens P, Teunissen CE, Barkhof F, van der Flier WM, Tijms BM. Grey matter network markers identify individuals with prodromal Alzheimer's disease who will show rapid clinical decline. Brain Commun 2022; 4:fcac026. [PMID: 35310828 PMCID: PMC8924646 DOI: 10.1093/braincomms/fcac026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/22/2021] [Accepted: 02/07/2022] [Indexed: 11/25/2022] Open
Abstract
Individuals with prodromal Alzheimer's disease show considerable variability in rates of cognitive decline, which hampers the ability to detect potential treatment effects in clinical trials. Prognostic markers to select those individuals who will decline rapidly within a trial time frame are needed. Brain network measures based on grey matter covariance patterns have been associated with future cognitive decline in Alzheimer's disease. In this longitudinal cohort study, we investigated whether cut-offs for grey matter networks could be derived to detect fast disease progression at an individual level. We further tested whether detection was improved by adding other biomarkers known to be associated with future cognitive decline [i.e. CSF tau phosphorylated at threonine 181 (p-tau181) levels and hippocampal volume]. We selected individuals with mild cognitive impairment and abnormal CSF amyloid β1-42 levels from the Amsterdam Dementia Cohort and the Alzheimer's Disease Neuroimaging Initiative, when they had available baseline structural MRI and clinical follow-up. The outcome was progression to dementia within 2 years. We determined prognostic cut-offs for grey matter network properties (gamma, lambda and small-world coefficient) using time-dependent receiver operating characteristic analysis in the Amsterdam Dementia Cohort. We tested the generalization of cut-offs in the Alzheimer's Disease Neuroimaging Initiative, using logistic regression analysis and classification statistics. We further tested whether combining these with CSF p-tau181 and hippocampal volume improved the detection of fast decliners. We observed that within 2 years, 24.6% (Amsterdam Dementia Cohort, n = 244) and 34.0% (Alzheimer's Disease Neuroimaging Initiative, n = 247) of prodromal Alzheimer's disease patients progressed to dementia. Using the grey matter network cut-offs for progression, we could detect fast progressors with 65% accuracy in the Alzheimer's Disease Neuroimaging Initiative. Combining grey matter network measures with CSF p-tau and hippocampal volume resulted in the best model fit for classification of rapid decliners, increasing detecting accuracy to 72%. These data suggest that single-subject grey matter connectivity networks indicative of a more random network organization can contribute to identifying prodromal Alzheimer's disease individuals who will show rapid disease progression. Moreover, we found that combined with p-tau and hippocampal volume this resulted in the highest accuracy. This could facilitate clinical trials by increasing chances to detect effects on clinical outcome measures.
Collapse
Affiliation(s)
- Wiesje Pelkmans
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ellen M. Vromen
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ellen Dicks
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, UCL, London, UK
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Epidemiology & Biostatistics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Betty M. Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | | |
Collapse
|
79
|
Lobo JD, Moore DJ, Bondi MW, Soontornniyomkij V, Soontornniyomkij B, Gouaux B, Achim CL, Ellis RJ, Sundermann EE. CSF markers of AD-related pathology relate specifically to memory impairment in older people with HIV: a pilot study. J Neurovirol 2022; 28:162-167. [PMID: 35103880 PMCID: PMC9081235 DOI: 10.1007/s13365-021-01048-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/07/2021] [Accepted: 12/18/2021] [Indexed: 02/03/2023]
Abstract
Given the co-occurrence of memory impairment in HIV-associated neurocognitive disorders (HAND) and amnestic mild cognitive impairment/Alzheimer's disease (aMCI/AD), biomarkers are needed that can disentangle these conditions among people with HIV (PWH). We assessed whether cerebrospinal fluid (CSF) markers of AD could help in this effort by determining their relationship to learning and memory deficits versus cognitive deficits more characteristic of HAND than aMCI/AD (processing speed and complex visual/motor coordination) among 31 older PWH. CSF amyloid-β42 phosphorylated-tau, amyloid-β40/amyloid-β42 and phosphorylated-tau/amyloid-β42 ratio related to learning/memory performance but not HAND-related deficits, suggesting that these biomarkers may have utility in disentangling aMCI/AD from HAND.
Collapse
Affiliation(s)
- Judith D Lobo
- Department of Psychiatry, University of California, 220 Dickinson St, #B, San Diego, CA, 92103, USA.
| | - David J Moore
- Department of Psychiatry, University of California, 220 Dickinson St, #B, San Diego, CA, 92103, USA
| | - Mark W Bondi
- Department of Psychiatry, University of California, 220 Dickinson St, #B, San Diego, CA, 92103, USA
- Psychology Service, VA San Diego Healthcare System, San Diego, USA
| | | | | | - Ben Gouaux
- Department of Psychiatry, University of California, 220 Dickinson St, #B, San Diego, CA, 92103, USA
| | - Cristian L Achim
- Department of Psychiatry, University of California, 220 Dickinson St, #B, San Diego, CA, 92103, USA
- Department of Pathology, University of California, San Diego, USA
| | - Ronald J Ellis
- Department of Neurosciences, University of California, San Diego, USA
| | - Erin E Sundermann
- Department of Psychiatry, University of California, 220 Dickinson St, #B, San Diego, CA, 92103, USA
| |
Collapse
|
80
|
Ashton NJ, Benedet AL, Pascoal TA, Karikari TK, Lantero-Rodriguez J, Brum WS, Mathotaarachchi S, Therriault J, Savard M, Chamoun M, Stoops E, Francois C, Vanmechelen E, Gauthier S, Zimmer ER, Zetterberg H, Blennow K, Rosa-Neto P. Cerebrospinal fluid p-tau231 as an early indicator of emerging pathology in Alzheimer's disease. EBioMedicine 2022; 76:103836. [PMID: 35158308 PMCID: PMC8850760 DOI: 10.1016/j.ebiom.2022.103836] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Phosphorylated tau (p-tau) epitopes in cerebrospinal fluid (CSF) are accurate biomarkers for a pathological and clinical diagnosis of Alzheimer's disease (AD) and are seen to be increased in preclinical stage of the disease. However, it is unknown if these increases transpire earlier, prior to amyloid-beta (Aβ) positivity as determined by position emission tomography (PET), and if an ordinal sequence of p-tau epitopes occurs at this incipient phase METHODS: We measured CSF concentrations of p-tau181, p-tau217 and p-tau231 in 171 participants across the AD continuum who had undergone Aβ ([18F]AZD4694) and tau ([18F]MK6240) position emission tomography (PET) and clinical assessment FINDINGS: All CSF p-tau biomarkers were accurate predictors of cognitive impairment but CSF p-tau217 demonstrated the largest fold-changes in AD patients in comparison to non-AD dementias and cognitively unimpaired individuals. CSF p-tau231 and p-tau217 predicted Aβ and tau to a similar degree but p-tau231 attained abnormal levels first. P-tau231 was sensitive to the earliest changes of Aβ in the medial orbitofrontal, precuneus and posterior cingulate before global Aβ PET positivity was reached INTERPRETATION: We demonstrate that CSF p-tau231 increases early in development of AD pathology and is a principal candidate for detecting incipient Aβ pathology for therapeutic trial application FUNDING: Canadian Institutes of Health Research (CIHR), Canadian Consortium of Neurodegeneration and Aging, Weston Brain Institute, Brain Canada Foundation, the Fonds de Recherche du Québec.
Collapse
Affiliation(s)
- Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada; Department of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, USA
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Department of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, USA
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Erik Stoops
- ADx NeuroSciences, Technologiepark 94, Ghent 9052, Belgium
| | - Cindy Francois
- ADx NeuroSciences, Technologiepark 94, Ghent 9052, Belgium
| | | | - Serge Gauthier
- McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Canada; Department of Neurology and Neurosurgery, Director of the McGill University Research Centre for Studies in Aging, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Pharmacology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, Director of the McGill University Research Centre for Studies in Aging, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Montreal Neurological Institute, Montreal, QC, Canada.
| |
Collapse
|
81
|
Ferrer I. Alzheimer's disease is an inherent, natural part of human brain aging: an integrated perspective. FREE NEUROPATHOLOGY 2022; 3:17. [PMID: 37284149 PMCID: PMC10209894 DOI: 10.17879/freeneuropathology-2022-3806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/21/2022] [Indexed: 06/08/2023]
Abstract
Alzheimer disease is one of the most challenging demons in our society due to its very high prevalence and its clinical manifestations which cause deterioration of cognition, intelligence, and emotions - the very capacities that distinguish Homo sapiens from other animal species. Besides the personal, social, and economical costs, late stages of AD are vivid experiences for the family, relatives, friends, and general observers of the progressive ruin of an individual who turns into a being with lower mental and physical capacities than less evolved species. A human brain with healthy cognition, conscience, and emotions can succeed in dealing with most difficulties that life may pose. Without these capacities, the same person probably cannot. Due, in part, to this emotional impact, the absorbing study of AD has generated, over the years, a fascinating and complex story of theories, hypotheses, controversies, fashion swings, and passionate clashes, together with tremendous efforts and achievements geared to improve understanding of the pathogenesis and treatment of the disorder. Familal AD is rare and linked to altered genetic information associated with three genes. Sporadic AD (sAD) is much more common and multifactorial. A major point of clinical discussion has been, and still is, establishing the differences between brain aging and sAD. This is not a trivial question, as the neuropathological and molecular characteristics of normal brain aging and the first appearance of early stages of sAD-related pathology are not easily distinguishable in most individuals. Another important point is confidence in assigning responsibility for the beginning of sAD to a few triggering molecules, without considering the wide number of alterations that converge in the pathogenesis of aging and sAD. Genetic risk factors covering multiple molecular signals are increasing in number. In the same line, molecular pathways are altered at early stages of sAD pathology, currently grouped under the aegis of normal brain aging, only to increase massively at advanced stages of the process. Sporadic AD is here considered an inherent, natural part of human brain aging, which is prevalent in all humans, and variably present or not in a few individuals in other species. The progression of the process has devastating effects in a relatively low percentage of human beings eventually evolving to dementia. The continuum of brain aging and sAD implies the search for a different approach in the study of human brain aging at the first stages of the biological process, and advances in the use of new technologies aimed at slowing down the molecular defects underlying human brain aging and sAD at the outset, and transfering information and tasks to AI and coordinated devices.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Emeritus Researcher of the Bellvitge Institute of Biomedical Research (IDIBELL); Biomedical Research Network of Neurodegenerative Diseases (CIBERNED); Institute of Neurosciences, University of Barcelona; Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
82
|
Frisoni GB, Altomare D, Thal DR, Ribaldi F, van der Kant R, Ossenkoppele R, Blennow K, Cummings J, van Duijn C, Nilsson PM, Dietrich PY, Scheltens P, Dubois B. The probabilistic model of Alzheimer disease: the amyloid hypothesis revised. Nat Rev Neurosci 2022; 23:53-66. [PMID: 34815562 PMCID: PMC8840505 DOI: 10.1038/s41583-021-00533-w] [Citation(s) in RCA: 242] [Impact Index Per Article: 80.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 01/03/2023]
Abstract
The current conceptualization of Alzheimer disease (AD) is driven by the amyloid hypothesis, in which a deterministic chain of events leads from amyloid deposition and then tau deposition to neurodegeneration and progressive cognitive impairment. This model fits autosomal dominant AD but is less applicable to sporadic AD. Owing to emerging information regarding the complex biology of AD and the challenges of developing amyloid-targeting drugs, the amyloid hypothesis needs to be reconsidered. Here we propose a probabilistic model of AD in which three variants of AD (autosomal dominant AD, APOE ε4-related sporadic AD and APOE ε4-unrelated sporadic AD) feature decreasing penetrance and decreasing weight of the amyloid pathophysiological cascade, and increasing weight of stochastic factors (environmental exposures and lower-risk genes). Together, these variants account for a large share of the neuropathological and clinical variability observed in people with AD. The implementation of this model in research might lead to a better understanding of disease pathophysiology, a revision of the current clinical taxonomy and accelerated development of strategies to prevent and treat AD.
Collapse
Affiliation(s)
- Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland.
| | - Daniele Altomare
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology, and Leuven Brain Institute, University of Leuven, Leuven, Belgium
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Centro S. Giovanni di Dio Fatebenefratelli, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Rik van der Kant
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Kaj Blennow
- Cinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences; University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Cornelia van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | | | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Life Science Partners, Amsterdam, Netherlands
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d'Alzheimer, IM2A, Groupe Hospitalier Pitié-Salpêtrière, Sorbonne Université, Paris, France
- Institut du Cerveau et de la Moelle Épinière, UMR-S975, INSERM, Paris, France
| |
Collapse
|
83
|
Sun Y, Wang M, Zhao Y, Hu K, Liu Y, Liu B. A Pathway-Specific Polygenic Risk Score is Associated with Tau Pathology and Cognitive Decline. J Alzheimers Dis 2021; 85:1745-1754. [DOI: 10.3233/jad-215163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: Tauopathy is a primary neuropathological hallmark of Alzheimer’s disease with a strong relationship to cognitive impairment. In the brain, tau aggregation is associated with the regulation of tau kinases and the binding ability of tau to microtubules. Objective: To explore the potential for using specific polygenic risk scores (PRSs), combining the genetic influences involved in tau-protein kinases and the tau-protein binding pathway, as predictors of tau pathology and cognitive decline in non-demented individuals. Methods: We computed a pathway-specific PRS using summary statistics from previous large-scale genome-wide association studies of dementia. We examined whether PRS is related to tau uptake in positron emission tomography (PET), tau levels, and the rate of tau level changes in cerebrospinal fluid (CSF). We further assessed whether PRS is associated with memory impairment mediated by CSF tau levels. Results: A higher PRS was related to elevated CSF tau levels and tau-PET uptake at baseline, as well as greater rates of change in CSF tau levels. Moreover, PRS was associated with memory impairment, mediated by increased CSF tau levels. The association between PRS and tau pathology was significant when APOE was excluded, even among females. However, the effect of PRS on cognitive decline appeared to be driven by the inclusion of APOE. Conclusion: The influence of genetic risk in a specific tau-related biological pathway may make an individual more susceptible to tau pathology, resulting in cognitive dysfunction in an early preclinical phase of the disease.
Collapse
Affiliation(s)
- Yuqing Sun
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Meng Wang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Yuxin Zhao
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Ke Hu
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Bing Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | | |
Collapse
|
84
|
Alzheimer disease in African American individuals: increased incidence or not enough data? Nat Rev Neurol 2021; 18:56-62. [PMID: 34873310 PMCID: PMC8647782 DOI: 10.1038/s41582-021-00589-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2021] [Indexed: 11/13/2022]
Abstract
Research on racial differences in Alzheimer disease (AD) dementia has increased in recent years. Older African American individuals bear a disproportionate burden of AD and cognitive impairment compared with non-Latino white individuals. Tremendous progress has been made over the past two decades in our understanding of the neurobiological substrates of AD. However, owing to well-documented challenges of study participant recruitment and a persistent lack of biological data in the African American population, knowledge of the drivers of these racial disparities has lagged behind. Therapeutic targets and effective interventions for AD are increasingly sought, but without a better understanding of the disease in African American individuals, any identified treatments and/or cures will evade this rapidly growing at-risk population. In this Perspective, I introduce three key obstacles to progress in understanding racial differences in AD: uncertainty about diagnostic criteria, disparate cross-sectional and longitudinal findings; and a dearth of neuropathological data. I also highlight evidence-informed strategies to move the field forward. In this Perspective, Barnes introduces three key obstacles to progress in our understanding of racial differences in Alzheimer disease and highlights evidence-informed strategies that can move the field forward.
Collapse
|
85
|
Andersen E, Casteigne B, Chapman WD, Creed A, Foster F, Lapins A, Shatz R, Sawyer RP. Diagnostic biomarkers in Alzheimer’s disease. Biomark Neuropsychiatry 2021. [DOI: 10.1016/j.bionps.2021.100041] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
86
|
Chan MY, Han L, Carreno CA, Zhang Z, Rodriguez RM, LaRose M, Hassenstab J, Wig GS. Long-term prognosis and educational determinants of brain network decline in older adult individuals. NATURE AGING 2021; 1:1053-1067. [PMID: 35382259 PMCID: PMC8979545 DOI: 10.1038/s43587-021-00125-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/07/2021] [Indexed: 12/21/2022]
Abstract
Older adults with lower education are at greater risk for dementia. It is unclear which brain changes lead to these outcomes. Longitudinal imaging-based measures of brain structure and function were examined in adult individuals (baseline age, 45-86 years; two to five visits per participant over 1-9 years). College degree completion differentiates individual-based and neighborhood-based measures of socioeconomic status and disadvantage. Older adults (~65 years and over) without a college degree exhibit a pattern of declining large-scale functional brain network organization (resting-state system segregation) that is less evident in their college-educated peers. Declining brain system segregation predicts impending changes in dementia severity, measured up to 10 years past the last scan date. The prognostic value of brain network change is independent of Alzheimer's disease (AD)-related genetic risk (APOE status), the presence of AD-associated pathology (cerebrospinal fluid phosphorylated tau, cortical amyloid) and cortical thinning. These results demonstrate that the trajectory of an individual's brain network organization varies in relation to their educational attainment and, more broadly, is a unique indicator of individual brain health during older age.
Collapse
Affiliation(s)
- Micaela Y. Chan
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Liang Han
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Claudia A. Carreno
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Ziwei Zhang
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Rebekah M. Rodriguez
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Megan LaRose
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gagan S. Wig
- Center for Vital Longevity and School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
87
|
Autobiographical Memory Fluency Reductions in Cognitively Unimpaired Middle-Aged and Older Adults at Increased Risk for Alzheimer's Disease Dementia. J Int Neuropsychol Soc 2021; 27:905-915. [PMID: 33509324 PMCID: PMC8319219 DOI: 10.1017/s1355617720001319] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Recent research has revealed that cognitively unimpaired older adults who are at higher risk for developing Alzheimer's disease (AD) dementia often exhibit subtle cognitive alterations in their neuropsychological profiles. Emerging evidence suggests that autobiographical memory, which is memory for personal events and knowledge, may be sensitive to early AD-related cognitive alterations. In the present study, we investigated whether the rapid generation of autobiographical memory category exemplars, a retrieval process that taxes the neural network that is vulnerable to early AD, is compromised in cognitively unimpaired middle-aged and older carriers of the e4 allele of the apolipoprotein E gene (APOE4), which increases risk for AD dementia. METHODS In addition to standard neuropsychological tests, we administered a fluency task that requires generating exemplars for two types of autobiographical memory, namely episodic memories and personal semantics, to a group of cognitively unimpaired middle-aged and older adults (n = 45) enriched with APOE4 carriers (n = 20). RESULTS While no APOE4 deficits were found on standard neuropsychological tests, episodic and personal semantic exemplar generation was reduced in the APOE4 group. DISCUSSION Autobiographical memory aberrations associated with a higher risk for AD are evident in fluency and affect both episodic memory and personal semantics.
Collapse
|
88
|
Bollinger RM, Keleman A, Thompson R, Westerhaus E, Fagan AM, Benzinger TL, Schindler SE, Xiong C, Balota D, Morris JC, Ances BM, Stark SL. Falls: a marker of preclinical Alzheimer disease: a cohort study protocol. BMJ Open 2021; 11:e050820. [PMID: 34526343 PMCID: PMC8444237 DOI: 10.1136/bmjopen-2021-050820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION Progression to symptomatic Alzheimer disease (AD) occurs slowly over a series of preclinical stages. Declining functional mobility may be an early indicator of loss of brain network integration and may lead to an increased risk of experiencing falls. It is unknown whether measures of functional mobility and falls are preclinical markers of AD. The purpose of this study is to examine (1) the relationship between falls and functional mobility with AD biomarkers to determine when falls occur within the temporal progression to symptomatic Alzheimer disease, and (2) the attentional compared with perceptual/motor systems that underlie falls and functional mobility changes seen with AD. METHODS AND ANALYSIS This longitudinal cohort study will be conducted at the Knight Alzheimer Disease Research Center. Approximately 350 cognitively normal participants (with and without preclinical AD) will complete an in-home visit every year for 4 years. During each yearly assessment, functional mobility will be assessed using the Performance Oriented Mobility Assessment, Timed Up and Go, and Timed Up and Go dual task. Data regarding falls (including number and severity) will be collected monthly by self-report and confirmed through interviews. This study will leverage ongoing neuropsychological assessments and neuroimaging (including molecular imaging using positron emission tomography and MRI) performed by the Knight Alzheimer Disease Research Center. Relationships between falls and biomarkers of amyloid, tau and neurodegeneration will be evaluated. ETHICS AND DISSEMINATION This study was approved by the Washington University in St. Louis Institutional Review Board (reference number 201807135). Written informed consent will be obtained in the home prior to the collection of any study data. Results will be published in peer-reviewed publications and presented at national and international conferences. TRIAL REGISTRATION NUMBER NCT04949529; Pre-results.
Collapse
Affiliation(s)
- Rebecca M Bollinger
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Audrey Keleman
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Regina Thompson
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Elizabeth Westerhaus
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tammie Ls Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David Balota
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, Missouri, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Beau M Ances
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Susan L Stark
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
89
|
Hassenstab J, Nicosia J, LaRose M, Aschenbrenner AJ, Gordon BA, Benzinger TLS, Xiong C, Morris JC. Is comprehensiveness critical? Comparing short and long format cognitive assessments in preclinical Alzheimer disease. Alzheimers Res Ther 2021; 13:153. [PMID: 34517889 PMCID: PMC8436865 DOI: 10.1186/s13195-021-00894-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/24/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Comprehensive testing of cognitive functioning is standard practice in studies of Alzheimer disease (AD). Short-form tests like the Montreal Cognitive Assessment (MoCA) use a "sampling" of measures, administering key items in a shortened format to efficiently assess cognition while reducing time requirements, participant burden, and administrative costs. We compared the MoCA to a commonly used long-form cognitive battery in predicting AD symptom onset and sensitivity to AD neuroimaging biomarkers. METHODS Survival, area under the receiver operating characteristic (ROC) curve (AUC), and multiple regression analyses compared the MoCA and long-form measures in predicting time to symptom onset in cognitively normal older adults (n = 6230) from the National Alzheimer's Coordinating Center (NACC) cohort who had, on average, 2.3 ± 1.2 annual assessments. Multiple regression models in a separate sample (n = 416) from the Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC) compared the sensitivity of the MoCA and long-form measures to neuroimaging biomarkers including amyloid PET, tau PET, and cortical thickness. RESULTS Hazard ratios suggested that both the MoCA and the long-form measures are similarly and modestly efficacious in predicting symptomatic conversion, although model comparison analyses indicated that the long-form measures slightly outperformed the MoCA (HRs > 1.57). AUC analyses indicated no difference between the measures in predicting conversion (DeLong's test, Z = 1.48, p = 0.13). Sensitivity to AD neuroimaging biomarkers was similar for the two measures though there were only modest associations with tau PET (rs = - 0.13, ps < 0.02) and cortical thickness in cognitively normal participants (rs = 0.15-0.16, ps < 0.007). CONCLUSIONS Both test formats showed weak associations with symptom onset, AUC analyses indicated low diagnostic accuracy, and biomarker correlations were modest in cognitively normal participants. Alternative assessment approaches are needed to improve how clinicians and researchers monitor cognitive changes and disease progression prior to symptom onset.
Collapse
Affiliation(s)
- Jason Hassenstab
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA.
| | - Jessica Nicosia
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Megan LaRose
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
90
|
Howlett J, Hill SM, Ritchie CW, Tom BDM. Disease Modelling of Cognitive Outcomes and Biomarkers in the European Prevention of Alzheimer's Dementia Longitudinal Cohort. Front Big Data 2021; 4:676168. [PMID: 34490422 PMCID: PMC8417903 DOI: 10.3389/fdata.2021.676168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/30/2021] [Indexed: 12/04/2022] Open
Abstract
A key challenge for the secondary prevention of Alzheimer’s dementia is the need to identify individuals early on in the disease process through sensitive cognitive tests and biomarkers. The European Prevention of Alzheimer’s Dementia (EPAD) consortium recruited participants into a longitudinal cohort study with the aim of building a readiness cohort for a proof-of-concept clinical trial and also to generate a rich longitudinal data-set for disease modelling. Data have been collected on a wide range of measurements including cognitive outcomes, neuroimaging, cerebrospinal fluid biomarkers, genetics and other clinical and environmental risk factors, and are available for 1,828 eligible participants at baseline, 1,567 at 6 months, 1,188 at one-year follow-up, 383 at 2 years, and 89 participants at three-year follow-up visit. We novelly apply state-of-the-art longitudinal modelling and risk stratification approaches to these data in order to characterise disease progression and biological heterogeneity within the cohort. Specifically, we use longitudinal class-specific mixed effects models to characterise the different clinical disease trajectories and a semi-supervised Bayesian clustering approach to explore whether participants can be stratified into homogeneous subgroups that have different patterns of cognitive functioning evolution, while also having subgroup-specific profiles in terms of baseline biomarkers and longitudinal rate of change in biomarkers.
Collapse
Affiliation(s)
- James Howlett
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | - Steven M Hill
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | - Craig W Ritchie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Brian D M Tom
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
91
|
Yang YW, Hsu KC, Wei CY, Tzeng RC, Chiu PY. Operational Determination of Subjective Cognitive Decline, Mild Cognitive Impairment, and Dementia Using Sum of Boxes of the Clinical Dementia Rating Scale. Front Aging Neurosci 2021; 13:705782. [PMID: 34557083 PMCID: PMC8455062 DOI: 10.3389/fnagi.2021.705782] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/10/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives: The Clinical Dementia Rating (CDR) Scale is the gold standard for the staging of dementia due to Alzheimer's disease (AD). However, the application of CDR for the staging of subjective cognitive decline (SCD) and mild cognitive impairment (MCI) in AD remains controversial. This study aimed to use the sum of boxes of the CDR (CDR-SB) plus an SCD single questionnaire to operationally determine the different stages of cognitive impairment (CI) due to AD and non-AD. Methods: This was a two-phase study, and we retrospectively analyzed the Show Chwan Dementia registry database using the data selected from 2015 to 2020. Individuals with normal cognition (NC), SCD, MCI, and mild dementia (MD) due to AD or non-AD with a CDR < 2 were included in the analysis. Results: A total of 6,946 individuals were studied, including 875, 1,009, 1,585, and 3,447 with NC, SCD, MCI, and MD, respectively. The cutoff scores of CDR-SB for NC/SCD, SCD/MCI, and MCI/dementia were 0/0.5, 0.5/1.0, and 2.5/3.0, respectively. The receiver operating characteristic (ROC) analysis showed that the area under the curve (AUC) values of the test groups were 0.85, 0.90, and 0.92 for discriminating NC from SCD, SCD from MCI, and MCI from dementia, respectively. Compared with the Cognitive Abilities Screening Instrument or the Montreal Cognitive Assessment, the use of CDR-SB is less influenced by age and education. Conclusion: Our study showed that the operational determination of SCD, MCI, and dementia using the CDR-SB is practical and can be applied in clinical settings and research on CI or dementia.
Collapse
Affiliation(s)
- Yu-Wan Yang
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | - Kai-Cheng Hsu
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan
- Department of Medicine, China Medical University, Taichung, Taiwan
- Artificial Intelligence Center for Medical Diagnosis, China Medical University Hospital, Taichung, Taiwan
| | - Cheng-Yu Wei
- Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, Taipei, Taiwan
| | - Ray-Chang Tzeng
- Department of Neurology, Tainan Municipal Hospital, Tainan, Taiwan
| | - Pai-Yi Chiu
- Department of Neurology, Show Chwan Memorial Hospital, Changhua, Taiwan
- Department of Nursing, College of Nursing and Health Sciences, Da-Yeh University, Changhua, Taiwan
| |
Collapse
|
92
|
Alawode DOT, Heslegrave AJ, Ashton NJ, Karikari TK, Simrén J, Montoliu‐Gaya L, Pannee J, O´Connor A, Weston PSJ, Lantero‐Rodriguez J, Keshavan A, Snellman A, Gobom J, Paterson RW, Schott JM, Blennow K, Fox NC, Zetterberg H. Transitioning from cerebrospinal fluid to blood tests to facilitate diagnosis and disease monitoring in Alzheimer's disease. J Intern Med 2021; 290:583-601. [PMID: 34021943 PMCID: PMC8416781 DOI: 10.1111/joim.13332] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/18/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is increasingly prevalent worldwide, and disease-modifying treatments may soon be at hand; hence, now, more than ever, there is a need to develop techniques that allow earlier and more secure diagnosis. Current biomarker-based guidelines for AD diagnosis, which have replaced the historical symptom-based guidelines, rely heavily on neuroimaging and cerebrospinal fluid (CSF) sampling. While these have greatly improved the diagnostic accuracy of AD pathophysiology, they are less practical for application in primary care, population-based and epidemiological settings, or where resources are limited. In contrast, blood is a more accessible and cost-effective source of biomarkers in AD. In this review paper, using the recently proposed amyloid, tau and neurodegeneration [AT(N)] criteria as a framework towards a biological definition of AD, we discuss recent advances in biofluid-based biomarkers, with a particular emphasis on those with potential to be translated into blood-based biomarkers. We provide an overview of the research conducted both in CSF and in blood to draw conclusions on biomarkers that show promise. Given the evidence collated in this review, plasma neurofilament light chain (N) and phosphorylated tau (p-tau; T) show particular potential for translation into clinical practice. However, p-tau requires more comparisons to be conducted between its various epitopes before conclusions can be made as to which one most robustly differentiates AD from non-AD dementias. Plasma amyloid beta (A) would prove invaluable as an early screening modality, but it requires very precise tests and robust pre-analytical protocols.
Collapse
Affiliation(s)
- D. O. T. Alawode
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - A. J. Heslegrave
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - N. J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Department of Old Age PsychiatryInstitute of Psychiatry, Psychology & NeuroscienceKing’s College LondonLondonUK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS FoundationLondonUK
| | - T. K. Karikari
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Simrén
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - L. Montoliu‐Gaya
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Pannee
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - A. O´Connor
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - P. S. J. Weston
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. Lantero‐Rodriguez
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - A. Keshavan
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - A. Snellman
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Turku PET CentreUniversity of TurkuTurkuFinland
| | - J. Gobom
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - R. W. Paterson
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. M. Schott
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - K. Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - N. C. Fox
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - H. Zetterberg
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| |
Collapse
|
93
|
Fasae KD, Abolaji AO, Faloye TR, Odunsi AY, Oyetayo BO, Enya JI, Rotimi JA, Akinyemi RO, Whitworth AJ, Aschner M. Metallobiology and therapeutic chelation of biometals (copper, zinc and iron) in Alzheimer's disease: Limitations, and current and future perspectives. J Trace Elem Med Biol 2021; 67:126779. [PMID: 34034029 DOI: 10.1016/j.jtemb.2021.126779] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 04/03/2021] [Accepted: 05/10/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent cause of cognitive impairment and dementia worldwide. The pathobiology of the disease has been studied in the form of several hypotheses, ranging from oxidative stress, amyloid-beta (Aβ) aggregation, accumulation of tau forming neurofibrillary tangles (NFT) through metal dysregulation and homeostasis, dysfunction of the cholinergic system, and to inflammatory and autophagic mechanism. However, none of these hypotheses has led to confirmed diagnostics or approved cure for the disease. OBJECTIVE This review is aimed as a basic and an encyclopedic short course into metals in AD and discusses the advances in chelation strategies and developments adopted in the treatment of the disease. Since there is accumulating evidence of the role of both biometal dyshomeostasis (iron (Fe), copper (Cu), and zinc (Zn)) and metal-amyloid interactions that lead to the pathogenesis of AD, this review focuses on unraveling therapeutic chelation strategies that have been considered in the treatment of the disease, aiming to sequester free and protein-bound metal ions and reducing cerebral metal burden. Promising compounds possessing chemically modified moieties evolving as multi-target ligands used as anti-AD drug candidates are also covered. RESULTS AND CONCLUSION Several multidirectional and multifaceted studies on metal chelation therapeutics show the need for improved synthesis, screening, and analysis of compounds to be able to effectively present chelating anti-AD drugs. Most drug candidates studied have limitations in their physicochemical properties; some enhance redistribution of metal ions, while others indirectly activate signaling pathways in AD. The metal chelation process in vivo still needs to be established and the design of potential anti-AD compounds that bi-functionally sequester metal ions as well as inhibit the Aβ aggregation by competing with the metal ions and reducing metal-induced oxidative damage and neurotoxicity may signal a bright end in chelation-based therapeutics of AD.
Collapse
Affiliation(s)
- Kehinde D Fasae
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Amos O Abolaji
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria.
| | - Tolulope R Faloye
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Atinuke Y Odunsi
- Department of Biochemistry, Molecular Drug Metabolism and Toxicology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Bolaji O Oyetayo
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, College of Medicine, University of Ibadan, Nigeria
| | - Joseph I Enya
- Department of Anatomy, University of Ilorin, Kwara State, Nigeria
| | - Joshua A Rotimi
- Department of Biochemistry and Molecular Biology, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Rufus O Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | | | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
94
|
Liss JL, Seleri Assunção S, Cummings J, Atri A, Geldmacher DS, Candela SF, Devanand DP, Fillit HM, Susman J, Mintzer J, Bittner T, Brunton SA, Kerwin DR, Jackson WC, Small GW, Grossberg GT, Clevenger CK, Cotter V, Stefanacci R, Wise‐Brown A, Sabbagh MN. Practical recommendations for timely, accurate diagnosis of symptomatic Alzheimer's disease (MCI and dementia) in primary care: a review and synthesis. J Intern Med 2021; 290:310-334. [PMID: 33458891 PMCID: PMC8359937 DOI: 10.1111/joim.13244] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/10/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023]
Abstract
The critical role of primary care clinicians (PCCs) in Alzheimer's disease (AD) prevention, diagnosis and management must evolve as new treatment paradigms and disease-modifying therapies (DMTs) emerge. Our understanding of AD has grown substantially: no longer conceptualized as a late-in-life syndrome of cognitive and functional impairments, we now recognize that AD pathology builds silently for decades before cognitive impairment is detectable. Clinically, AD first manifests subtly as mild cognitive impairment (MCI) due to AD before progressing to dementia. Emerging optimism for improved outcomes in AD stems from a focus on preventive interventions in midlife and timely, biomarker-confirmed diagnosis at early signs of cognitive deficits (i.e. MCI due to AD and mild AD dementia). A timely AD diagnosis is particularly important for optimizing patient care and enabling the appropriate use of anticipated DMTs. An accelerating challenge for PCCs and AD specialists will be to respond to innovations in diagnostics and therapy for AD in a system that is not currently well positioned to do so. To overcome these challenges, PCCs and AD specialists must collaborate closely to navigate and optimize dynamically evolving AD care in the face of new opportunities. In the spirit of this collaboration, we summarize here some prominent and influential models that inform our current understanding of AD. We also advocate for timely and accurate (i.e. biomarker-defined) diagnosis of early AD. In doing so, we consider evolving issues related to prevention, detecting emerging cognitive impairment and the role of biomarkers in the clinic.
Collapse
Affiliation(s)
| | - S. Seleri Assunção
- US Medical Affairs – Neuroscience, Genentech, A Member of the Roche GroupSouth San FranciscoCAUSA
| | - J. Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health SciencesUniversity of NevadaLas VegasNVUSA
- Lou Ruvo Center for Brain Health – Cleveland Clinic NevadaLas VegasNVUSA
| | - A. Atri
- Banner Sun Health Research InstituteSun CityAZUSA
- Center for Brain/Mind MedicineDepartment of NeurologyBrigham and Women’s HospitalBostonMAUSA
- Harvard Medical SchoolBostonMAUSA
| | - D. S. Geldmacher
- Department of NeurologyUniversity of Alabama at BirminghamBirminghamALUSA
| | - S. F. Candela
- Health & Wellness Partners, LLCUpper Saddle RiverNJUSA
| | - D. P. Devanand
- Division of Geriatric PsychiatryNew York State Psychiatric Institute and Columbia University Irving Medical CenterNew YorkNYUSA
| | - H. M. Fillit
- Departments of Geriatric Medicine, Medicine, and NeuroscienceIcahn School of Medicine and Mt. SinaiNew YorkNYUSA
- Alzheimer’s Drug Discovery FoundationNew YorkNYUSA
| | - J. Susman
- Department of Family and Community MedicineNortheast Ohio Medical UniversityRootstownOHUSA
| | - J. Mintzer
- Roper St Francis HealthcareCharlestonSCUSA
- Ralph H. Johnson VA Medical CenterCharlestonSCUSA
| | | | - S. A. Brunton
- Department of Family MedicineTouro UniversityVallejoCAUSA
| | - D. R. Kerwin
- Kerwin Medical CenterDallasTXUSA
- Department of Neurology and NeurotherapeuticsUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - W. C. Jackson
- Departments of Family Medicine and PsychiatryUniversity of Tennessee College of MedicineMemphisTNUSA
| | - G. W. Small
- Division of Geriatric PsychiatryUCLA Longevity CenterSemel Institute for Neuroscience & Human BehaviorUniversity of California – Los AngelesLos AngelesCAUSA
| | - G. T. Grossberg
- Division of Geriatric PsychiatrySt Louis University School of MedicineSt LouisMOUSA
| | - C. K. Clevenger
- Department of NeurologyNell Hodgson Woodruff School of NursingEmory UniversityAtlantaGAUSA
| | - V. Cotter
- Johns Hopkins School of NursingBaltimoreMDUSA
| | - R. Stefanacci
- Jefferson College of Population HealthThomas Jefferson UniversityPhiladelphiaPAUSA
| | - A. Wise‐Brown
- US Medical Affairs – Neuroscience, Genentech, A Member of the Roche GroupSouth San FranciscoCAUSA
| | - M. N. Sabbagh
- Lou Ruvo Center for Brain Health – Cleveland Clinic NevadaLas VegasNVUSA
| |
Collapse
|
95
|
Lv S, Zhou X, Li Y, Zhang S, Wang Y, Jia S, Niu X, Wang L, Peng D. The Association Between Plasma α-Synuclein (α-syn) Protein, Urinary Alzheimer-Associated Neuronal Thread Protein (AD7c-NTP), and Apolipoprotein Epsilon 4 (ApoE ε4) Alleles and Cognitive Decline in 60 Patients with Alzheimer's Disease Compared with 28 Age-Matched Normal Individuals. Med Sci Monit 2021; 27:e932998. [PMID: 34312362 PMCID: PMC8325392 DOI: 10.12659/msm.932998] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Accumulating evidence has shown that α-synuclein (α-syn) pathology is involved in the pathophysiology of Alzheimer’s disease (AD). This study aimed to investigate the association between the levels of plasma α-syn protein, urinary Alzheimer-associated neuronal thread protein (AD7c-NTP), apolipoprotein epsilon 4 (ApoE ɛ4) alleles and cognitive decline in 60 AD patients compared with 28 age-matched normal controls (NCs) at a single center. Material/Methods All participants underwent α-syn, apolipoprotein E (ApoE), AD7c-NTP, cholesterol (CHO), high-density lipoprotein (HDL), low-density lipoprotein (LDL) and triglycerides (TGs) analyses, neuropsychological scale assessments and neuroimaging analysis. Moreover, urine and peripheral blood samples were collected from all participants. The levels of plasma α-syn and AD7c-NTP were assayed using an enzyme-linked immunosorbent assay (ELISA) kit. Other test results were obtained from China-Japan Friendship Hospital. Results We found that plasma α-syn levels were significantly different between AD patients and NCs (p=0.045). α-Syn levels were also associated with AD7c-NTP (r=0.231, p=0.03) but not ApoE ɛ4 (Z=−0.147, p=0.883) levels. Neither α-syn [CHO (p=0.432), HDL (p=0.484), LDL (p=0.733) or TGs (p=0.253)] nor AD7c-NTP [CHO (p=0.867), HDL (p=0.13), LDL (p=0.57) or TGs (p=0.678)] had a relationship with lipids. Conclusions This study showed that the levels of plasma α-syn protein and urinary AD7c-NTP were significantly increased in AD patients compared with NCs, but not with ApoE alleles or serum lipid levels.
Collapse
Affiliation(s)
- Shuang Lv
- Department of Neurology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China (mainland).,Department of Neurology, China-Japan Friendship Hospital, Beijing, China (mainland)
| | - Xiao Zhou
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China (mainland).,Department of Neurology, Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China (mainland)
| | - Yiming Li
- Department of Cardiovascular, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China (mainland)
| | - Shujuan Zhang
- Department of Neurology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China (mainland).,Department of Neurology, China-Japan Friendship Hospital, Beijing, China (mainland)
| | - Yu Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China (mainland)
| | - Shuhong Jia
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China (mainland)
| | - Xiaoqian Niu
- Department of Neurology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China (mainland).,Department of Neurology, China-Japan Friendship Hospital, Beijing, China (mainland)
| | - Lei Wang
- Department of Neurology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China (mainland).,Department of Neurology, China-Japan Friendship Hospital, Beijing, China (mainland)
| | - Dantao Peng
- Department of Neurology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China (mainland).,Department of Neurology, China-Japan Friendship Hospital, Beijing, China (mainland).,Department of Neurology, Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China (mainland)
| |
Collapse
|
96
|
Williams ME, Elman JA, McEvoy LK, Andreassen OA, Dale AM, Eglit GML, Eyler LT, Fennema-Notestine C, Franz CE, Gillespie NA, Hagler DJ, Hatton SN, Hauger RL, Jak AJ, Logue MW, Lyons MJ, McKenzie RE, Neale MC, Panizzon MS, Puckett OK, Reynolds CA, Sanderson-Cimino M, Toomey R, Tu XM, Whitsel N, Xian H, Kremen WS. 12-year prediction of mild cognitive impairment aided by Alzheimer's brain signatures at mean age 56. Brain Commun 2021; 3:fcab167. [PMID: 34396116 PMCID: PMC8361427 DOI: 10.1093/braincomms/fcab167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/26/2021] [Accepted: 05/10/2021] [Indexed: 01/22/2023] Open
Abstract
Neuroimaging signatures based on composite scores of cortical thickness and hippocampal volume predict progression from mild cognitive impairment to Alzheimer's disease. However, little is known about the ability of these signatures among cognitively normal adults to predict progression to mild cognitive impairment. Towards that end, a signature sensitive to microstructural changes that may predate macrostructural atrophy should be useful. We hypothesized that: (i) a validated MRI-derived Alzheimer's disease signature based on cortical thickness and hippocampal volume in cognitively normal middle-aged adults would predict progression to mild cognitive impairment; and (ii) a novel grey matter mean diffusivity signature would be a better predictor than the thickness/volume signature. This cohort study was part of the Vietnam Era Twin Study of Aging. Concurrent analyses compared cognitively normal and mild cognitive impairment groups at each of three study waves (ns = 246-367). Predictive analyses included 169 cognitively normal men at baseline (age = 56.1, range = 51-60). Our previously published thickness/volume signature derived from independent data, a novel mean diffusivity signature using the same regions and weights as the thickness/volume signature, age, and an Alzheimer's disease polygenic risk score were used to predict incident mild cognitive impairment an average of 12 years after baseline (follow-up age = 67.2, range = 61-71). Additional analyses adjusted for predicted brain age difference scores (chronological age minus predicted brain age) to determine if signatures were Alzheimer-related and not simply ageing-related. In concurrent analyses, individuals with mild cognitive impairment had higher (worse) mean diffusivity signature scores than cognitively normal participants, but thickness/volume signature scores did not differ between groups. In predictive analyses, age and polygenic risk score yielded an area under the curve of 0.74 (sensitivity = 80.00%; specificity = 65.10%). Prediction was significantly improved with addition of the mean diffusivity signature (area under the curve = 0.83; sensitivity = 85.00%; specificity = 77.85%; P = 0.007), but not with addition of the thickness/volume signature. A model including both signatures did not improve prediction over a model with only the mean diffusivity signature. Results held up after adjusting for predicted brain age difference scores. The novel mean diffusivity signature was limited by being yoked to the thickness/volume signature weightings. An independently derived mean diffusivity signature may thus provide even stronger prediction. The young age of the sample at baseline is particularly notable. Given that the brain signatures were examined when participants were only in their 50 s, our results suggest a promising step towards improving very early identification of Alzheimer's disease risk and the potential value of mean diffusivity and/or multimodal brain signatures.
Collapse
Affiliation(s)
- McKenna E Williams
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Joint Doctoral Program in Clinical Psychology, San Diego State University/University of California, San Diego, CA 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Jeremy A Elman
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Linda K McEvoy
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Ole A Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo 0316, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo 0372, Norway
| | - Anders M Dale
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
- Department of Neuroscience, University of California San Diego, La Jolla, CA 92093, USA
| | - Graham M L Eglit
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Lisa T Eyler
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Desert Pacific Mental Illness Research Education and Clinical Center, VA San Diego Healthcare System, CA 92093, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Carol E Franz
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Nathan A Gillespie
- Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Donald J Hagler
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Sean N Hatton
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Department of Neuroscience, University of California San Diego, La Jolla, CA 92093, USA
| | - Richard L Hauger
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center of Excellence for Stress and Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA 92093, USA
| | - Amy J Jak
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- VA San Diego Healthcare System, San Diego, CA 92093, USA
| | - Mark W Logue
- National Center for PTSD: Behavioral Science Division, VA Boston Healthcare System, Boston, MA 02130, USA
- Department of Psychiatry and the Biomedical Genetics Section, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Michael J Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, MA 02212, USA
| | - Ruth E McKenzie
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
- School of Education and Social Policy, Merrimack College, North Andover, MA 01845, USA
| | - Michael C Neale
- Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Matthew S Panizzon
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Olivia K Puckett
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Chandra A Reynolds
- Department of Psychology, University of California Riverside, Riverside, CA 92521, USA
| | - Mark Sanderson-Cimino
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Joint Doctoral Program in Clinical Psychology, San Diego State University/University of California, San Diego, CA 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Rosemary Toomey
- Department of Psychological and Brain Sciences, Boston University, Boston, MA 02212, USA
| | - Xin M Tu
- Family Medicine and Public Health, University of California San Diego, La Jolla, CA 92093, USA
| | - Nathan Whitsel
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Hong Xian
- Department of Biostatistics, St. Louis University, St. Louis, MO 63103, USA
| | - William S Kremen
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center of Excellence for Stress and Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA 92093, USA
| |
Collapse
|
97
|
McDade E, Llibre-Guerra JJ, Holtzman DM, Morris JC, Bateman RJ. The informed road map to prevention of Alzheimer Disease: A call to arms. Mol Neurodegener 2021; 16:49. [PMID: 34289882 PMCID: PMC8293489 DOI: 10.1186/s13024-021-00467-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/10/2021] [Indexed: 12/31/2022] Open
Abstract
Alzheimer disease (AD) prevention trials hold the promise to delay or prevent cognitive decline and dementia onset by intervening before significant neuronal damage occurs. In recent years, the first AD prevention trials have launched and are yielding important findings on the biology of targeting asymptomatic AD pathology. However, there are limitations that impact the design of these prevention trials, including the translation of animal models that recapitulate key stages and multiple pathological aspects of the human disease, missing target validation in asymptomatic disease, uncertain causality of the association of pathophysiologic changes with cognitive and clinical symptoms, and limited biomarker validation for novel targets. The field is accelerating advancements in key areas including the development of highly specific and quantitative biomarker measures for AD pathology, increasing our understanding of the course and relationship of amyloid and tau pathology in asymptomatic through symptomatic stages, and the development of powerful interventions that can slow or reverse AD amyloid pathology. We review the current status of prevention trials and propose key areas of needed research as a call to basic and translational scientists to accelerate AD prevention. Specifically, we review (1) sporadic and dominantly inherited primary and secondary AD prevention trials, (2) proposed targets, mechanisms, and drugs including the amyloid, tau, and inflammatory pathways and combination treatments, (3) the need for more appropriate prevention animal models and experiments, and (4) biomarkers and outcome measures needed to design human asymptomatic prevention trials. We conclude with actions needed to effectively move prevention targets and trials forward.
Collapse
Affiliation(s)
- Eric McDade
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Avenue, Campus Box, St Louis, MO 8111 USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Dominantly Inherited Alzheimer’s Network Trials Unit, St. Louis, MO 63110 USA
| | - Jorge J. Llibre-Guerra
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Avenue, Campus Box, St Louis, MO 8111 USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Dominantly Inherited Alzheimer’s Network Trials Unit, St. Louis, MO 63110 USA
| | - David M. Holtzman
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Avenue, Campus Box, St Louis, MO 8111 USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Dominantly Inherited Alzheimer’s Network Trials Unit, St. Louis, MO 63110 USA
| | - John C. Morris
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Avenue, Campus Box, St Louis, MO 8111 USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Dominantly Inherited Alzheimer’s Network Trials Unit, St. Louis, MO 63110 USA
| | - Randall J. Bateman
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Avenue, Campus Box, St Louis, MO 8111 USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Dominantly Inherited Alzheimer’s Network Trials Unit, St. Louis, MO 63110 USA
| |
Collapse
|
98
|
Pan L, Ou YN, Tan L, Tan L, Yu JT. Cerebrospinal fluid heart fatty acid‐binding protein as a predictive biomarker of neurodegeneration in Alzheimer’s disease. BRAIN SCIENCE ADVANCES 2021. [DOI: 10.26599/bsa.2021.9050003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Objective This study aims to investigate whether the heart fatty acid‐binding protein (HFABP) in the cerebrospinal fluid (CSF) was a potential predictive biomarker for Alzheimer’s disease (AD). Methods We evaluated the associations of CSF HFABP levels with core biomarkers, cognition, and brain structure in a sample population ( n = 302) from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database. Multiple linear regression and mixed‐effects models were employed in the analyses. AD progression was assessed using the Kaplan–Meier survival analysis. Results CSF HFABP was higher in patients with mild cognitive impairment and AD than the normal controls ( p < 0.001) and was particularly higher in those with amyloid‐β (Aβ) pathologic features. CSF HFABP was associated with higher baseline CSF t‐tau ( p < 0.001), CSF p‐tau ( p < 0.001), and CSF t‐tau/Aβ42 and CSF p‐tau/Aβ42 ( p < 0.01). Moreover, CSF HFABP was found to play predictive roles in hippocampal atrophy ( p < 0.01), cognitive decline ( p < 0.05), and the risk of AD ( p < 0.001). Conclusion Our findings suggest that CSF HFABP can be a predictive biomarker of AD.
Collapse
Affiliation(s)
- Lu Pan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong, China
| | - Lin Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian 116044, Liaoning, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| |
Collapse
|
99
|
Vecchio I, Sorrentino L, Paoletti A, Marra R, Arbitrio M. The State of The Art on Acetylcholinesterase Inhibitors in the Treatment of Alzheimer's Disease. J Cent Nerv Syst Dis 2021; 13:11795735211029113. [PMID: 34285627 PMCID: PMC8267037 DOI: 10.1177/11795735211029113] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/10/2021] [Indexed: 12/03/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic disabling disease that affects the central nervous system. The main consequences of AD include the decline of cognitive functions and language disorders. One of the causes leading to AD is the decrease of neurotransmitter acetylcholine (ACh) levels in the brain, in part due to a higher activity of acetylcholinesterase (AChE), the enzyme responsible for its degradation. Many acetylcholinesterase inhibitors (AChEIs), both natural and synthetic, have been developed and used through the years to counteract the progression of the disease. The first of such drugs approved for a therapeutic use was tacrine, that binds through a reversible bond to the enzyme. However, tacrine has since been withdrawn because of its adverse effects. Currently, donepezil and galantamine are very promising AChEIs with clinical benefits. Moreover, rivastigmine is considered a pseudo-irreversible compound with anti-AChE action, providing similar effects at the clinical level. The purpose of this review is to provide an overview of what has been published over the last decade on the effectiveness of AChEIs in AD, analysing the most relevant issues under the clinical and methodological profiles and the consequent possible welfare effects for the whole world. Furthermore, novel drugs and possible therapeutic approaches are also discussed.
Collapse
Affiliation(s)
- Immacolata Vecchio
- Institute for Biomedical Research and Innovation of the National Research Council, Catanzaro, Italy
| | | | - Annamaria Paoletti
- Institute for Biomedical Research and Innovation of the National Research Council, Catanzaro, Italy
| | - Rosario Marra
- Institute for Biomedical Research and Innovation of the National Research Council, Catanzaro, Italy
| | - Mariamena Arbitrio
- Institute for Biomedical Research and Innovation of the National Research Council, Catanzaro, Italy
| |
Collapse
|
100
|
Gao L, Li P, Gaba A, Musiek E, Ju YS, Hu K. Fractal motor activity regulation and sex differences in preclinical Alzheimer's disease pathology. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12211. [PMID: 34189248 PMCID: PMC8220856 DOI: 10.1002/dad2.12211] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Degradation in fractal motor activity regulation (FMAR), a measure of multiscale self-similarity of motor control, occurs in aging and accelerates with clinical progression to Alzheimer's disease (AD). Whether FMAR changes occur during the pre-symptomatic phase of the disease in women and men remains unknown. METHODS FMAR was assessed in cognitively normal participants (n = 178) who underwent 7 to 14 days of home actigraphy. Preclinical AD pathology was determined by amyloid imaging-Pittsburgh compound B (PiB) and cerebrospinal fluid (CSF) phosphorylated-tau181 (p-tau) to amyloid beta 42 (Aβ42) ratio. RESULTS Degradation in daytime FMAR was overall significantly associated with preclinical amyloid plaque pathology via PiB+ imaging (beta coefficient β = 0.217, standard error [SE] = 0.101, P = .034) and increasing CSF tau181-Aβ42 ratio (β = 0.220, SE = 0.084, P = .009). In subset analysis by sex, the effect sizes were significant in women for PiB+ (β = 0.279, SE = 0.112, P = .015) and CSF (β = 0.245, SE = 0.094, P = .011) but not in men (both Ps > .05). These associations remained after inclusion of daily activity level, apolipoprotein E ε4 carrier status, and rest/activity patterns. DISCUSSION Changes in daytime FMAR from actigraphy appear to be present in women early in preclinical AD. This may be a combination of earlier pathology changes in females reflected in daytime FMAR, and a relatively underpowered male group. Further studies are warranted to test FMAR as an early noncognitive physiological biomarker that precedes the onset of cognitive symptoms.
Collapse
Affiliation(s)
- Lei Gao
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Medical Biodynamics ProgramBrigham and Women's HospitalBostonMassachusettsUSA
- Division of Sleep MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Peng Li
- Medical Biodynamics ProgramBrigham and Women's HospitalBostonMassachusettsUSA
- Division of Sleep MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Arlen Gaba
- Medical Biodynamics ProgramBrigham and Women's HospitalBostonMassachusettsUSA
| | - Erik Musiek
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMissouriUSA
| | - Yo‐El S. Ju
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMissouriUSA
| | - Kun Hu
- Medical Biodynamics ProgramBrigham and Women's HospitalBostonMassachusettsUSA
- Division of Sleep MedicineHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|