151
|
Platero C, Tohka J, Strange B. Estimating Dementia Onset: AT(N) Profiles and Predictive Modeling in Mild Cognitive Impairment Patients. Curr Alzheimer Res 2024; 20:778-790. [PMID: 38425106 DOI: 10.2174/0115672050295317240223162312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Mild Cognitive Impairment (MCI) usually precedes the symptomatic phase of dementia and constitutes a window of opportunities for preventive therapies. OBJECTIVES The objective of this study was to predict the time an MCI patient has left to reach dementia and obtain the most likely natural history in the progression of MCI towards dementia. METHODS This study was conducted on 633 MCI patients and 145 subjects with dementia through 4726 visits over 15 years from Alzheimer Disease Neuroimaging Initiative (ADNI) cohort. A combination of data from AT(N) profiles at baseline and longitudinal predictive modeling was applied. A data-driven approach was proposed for categorical diagnosis prediction and timeline estimation of cognitive decline progression, which combined supervised and unsupervised learning techniques. RESULTS A reduced vector of only neuropsychological measures was selected for training the models. At baseline, this approach had high performance in detecting subjects at high risk of converting from MCI to dementia in the coming years. Furthermore, a Disease Progression Model (DPM) was built and also verified using three metrics. As a result of the DPM focused on the studied population, it was inferred that amyloid pathology (A+) appears about 7 years before dementia, and tau pathology (T+) and neurodegeneration (N+) occur almost simultaneously, between 3 and 4 years before dementia. In addition, MCI-A+ subjects were shown to progress more rapidly to dementia compared to MCI-A- subjects. CONCLUSION Based on proposed natural histories and cross-sectional and longitudinal analysis of AD markers, the results indicated that only a single cerebrospinal fluid sample is necessary during the prodromal phase of AD. Prediction from MCI into dementia and its timeline can be achieved exclusively through neuropsychological measures.
Collapse
Affiliation(s)
- Carlos Platero
- Health Science Technology Group, Technical University of Madrid, 28012 Madrid, Spain
| | - Jussi Tohka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Bryan Strange
- Laboratory for Clinical Neuroscience, CTB, Technical University of Madrid, IdISSC, Madrid, Spain
- Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| |
Collapse
|
152
|
Satake Y, Kanemoto H, Taomoto D, Suehiro T, Koizumi F, Sato S, Wada T, Matsunaga K, Shimosegawa E, Gotoh S, Mori K, Morihara T, Yoshiyama K, Ikeda M. Characteristics of very late-onset schizophrenia-like psychosis classified with the biomarkers for Alzheimer's disease: a retrospective cross-sectional study. Int Psychogeriatr 2024; 36:64-77. [PMID: 36714996 DOI: 10.1017/s1041610222001132] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OBJECTIVES We aimed to investigate the association between very late-onset schizophrenia-like psychosis (VLOSLP), a schizophrenia spectrum disorder with an onset of ≥60 years, and Alzheimer's disease (AD) using biomarkers. DESIGN Retrospective cross-sectional study. SETTING Neuropsychology clinic of Osaka University Hospital in Japan. PARTICIPANTS Thirty-three participants were classified into three groups: eight AD biomarker-negative VLOSLP (VLOSLP-AD), nine AD biomarker-positive VLOSLP (VLOSLP+AD), and sixteen amnestic mild cognitive impairment due to AD without psychosis (aMCI-P+AD) participants. MEASUREMENTS Phosphorylated tau levels in the cerebrospinal fluid and 18F-Florbetapir positron emission tomography results were used as AD biomarkers. Several scales (e.g. the Mini-Mental State Examination (MMSE), Wechsler Memory Scale-Revised (WMS-R) Logical Memory (LM) I and II, and Neuropsychiatric Inventory (NPI)-plus) were conducted to assess clinical characteristics. RESULTS Those in both VLOSLP-AD and +AD groups scored higher than those in aMCI-P+AD in WMS-R LM I. On the other hand, VLOSLP+AD participants scored in between the other two groups in the WMS-R LM II, with only VLOSLP-AD participants scoring significantly higher than aMCI-P+AD participants. There were no significant differences in sex distribution and MMSE scores among the three groups or in the subtype of psychotic symptoms between VLOSLP-AD and +AD participants. Four VLOSLP-AD and five VLOSLP+AD participants harbored partition delusions. Delusion of theft was shown in two VLOSLP-AD patients and five VLOSLP+AD patients. CONCLUSION Some VLOSLP patients had AD pathology. Clinical characteristics were different between AD biomarker-positive and AD biomarker-negative VLOSLP, which may be helpful for detecting AD pathology in VLOSLP patients.
Collapse
Affiliation(s)
- Yuto Satake
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hideki Kanemoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Daiki Taomoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takashi Suehiro
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Fuyuki Koizumi
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shunsuke Sato
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tamiki Wada
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Keiko Matsunaga
- Department of Molecular Imaging in Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Eku Shimosegawa
- Department of Molecular Imaging in Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shiho Gotoh
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kohji Mori
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takashi Morihara
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Psychiatry, Toyonaka Municipal Hospital, Toyonaka, Japan
| | - Kenji Yoshiyama
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
153
|
Hale MR, Langhough R, Du L, Hermann BP, Van Hulle CA, Carboni M, Kollmorgen G, Basche KE, Bruno D, Sanson-Miles L, Jonaitis EM, Chin NA, Okonkwo OC, Bendlin BB, Carlsson CM, Zetterberg H, Blennow K, Betthauser TJ, Johnson SC, Mueller KD. Associations between recall of proper names in story recall and CSF amyloid and tau in adults without cognitive impairment. Neurobiol Aging 2024; 133:87-98. [PMID: 37925995 PMCID: PMC10842469 DOI: 10.1016/j.neurobiolaging.2023.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 11/07/2023]
Abstract
Neuropsychological measures sensitive to decline in the preclinical phase of Alzheimer's disease are needed. We previously demonstrated that higher amyloid-beta (Aβ) assessed by positron emission tomography in adults without cognitive impairment was associated with recall of fewer proper names in Logical Memory story recall. The current study investigated the association between proper names and cerebrospinal fluid biomarkers (Aβ42/40, phosphorylated tau181 [pTau181], neurofilament light) in 223 participants from the Wisconsin Registry for Alzheimer's Prevention. We assessed associations between biomarkers and delayed Logical Memory total score and proper names using binary logistic regressions. Sensitivity analyses used multinomial logistic regression and stratified biomarker groups. Lower Logical Memory total score and proper names scores from the most recent visit were associated with biomarker positivity. Relatedly, there was a 27% decreased risk of being classified Aβ42/40+/pTau181+ for each additional proper name recalled. A linear mixed effects model found that longitudinal change in proper names recall was predicted by biomarker status. These results demonstrate a novel relationship between proper names and Alzheimer's disease-cerebrospinal fluid pathology.
Collapse
Affiliation(s)
- Madeline R Hale
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, USA
| | - Rebecca Langhough
- Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Lianlian Du
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Bruce P Hermann
- Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Carol A Van Hulle
- Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
| | | | | | - Kristin E Basche
- Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Davide Bruno
- School of Psychology, Liverpool John Moores University, Liverpool, UK
| | - Leah Sanson-Miles
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, USA
| | - Erin M Jonaitis
- Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Nathaniel A Chin
- Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA; VA Geriatric Research, Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Ozioma C Okonkwo
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Barbara B Bendlin
- Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA; VA Geriatric Research, Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Cynthia M Carlsson
- Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
| | - Henrik Zetterberg
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; 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, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - 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
| | - Tobey J Betthauser
- Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
| | - Sterling C Johnson
- Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA; VA Geriatric Research, Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Kimberly D Mueller
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, USA; Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
154
|
Hanert A, Schönfeld R, Weber FD, Nowak A, Döhring J, Philippen S, Granert O, Burgalossi A, Born J, Berg D, Göder R, Häussermann P, Bartsch T. Reduced overnight memory consolidation and associated alterations in sleep spindles and slow oscillations in early Alzheimer's disease. Neurobiol Dis 2024; 190:106378. [PMID: 38103701 DOI: 10.1016/j.nbd.2023.106378] [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: 08/01/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Spatial navigation critically underlies hippocampal-entorhinal circuit function that is early affected in Alzheimer's disease (AD). There is growing evidence that AD pathophysiology dynamically interacts with the sleep/wake cycle impairing hippocampal memory. To elucidate sleep-dependent consolidation in a cohort of symptomatic AD patients (n = 12, 71.25 ± 2.16 years), we tested hippocampal place learning by means of a virtual reality task and verbal memory by a word-pair association task before and after a night of sleep. Our results show an impaired overnight memory retention in AD compared with controls in the verbal task, together with a significant reduction of sleep spindle activity (i.e., lower amplitude of fast sleep spindles, p = 0.016) and increased duration of the slow oscillation (SO; p = 0.019). Higher spindle density, faster down-to-upstate transitions within SOs, and the time delay between SOs and nested spindles predicted better memory performance in healthy controls but not in AD patients. Our results show that mnemonic processing and memory consolidation in AD is slightly impaired as reflected by dysfunctional oscillatory dynamics and spindle-SO coupling during NonREM sleep. In this translational study based on experimental paradigms in animals and extending previous work in healthy aging and preclinical disease stages, our results in symptomatic AD further deepen the understanding of the memory decline within a bidirectional relationship of sleep and AD pathology.
Collapse
Affiliation(s)
- Annika Hanert
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Robby Schönfeld
- Institute of Psychology, Division of Clinical Psychology, Martin-Luther-University Halle-Wittenberg, 06099 Halle (Saale), Germany
| | - Frederik D Weber
- Institute for Medical Psychology and Behavioral Neurobiology, University of Tübingen, 72074 Tübingen, Germany; Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 EN Nijmegen, the Netherlands; Department of Sleep and Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Alexander Nowak
- Department of Psychiatry and Psychotherapy, Sleep Laboratory, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Juliane Döhring
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany; Institute for General Medicine, University Hospital of Schleswig-Holstein, 24105 Kiel, Germany
| | - Sarah Philippen
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Oliver Granert
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Andrea Burgalossi
- Institute of Neurobiology, Werner-Reichardt Center for Integrative Neuroscience, University of Tübingen, 72074 Tübingen, Germany
| | - Jan Born
- Institute for Medical Psychology and Behavioral Neurobiology, University of Tübingen, 72074 Tübingen, Germany
| | - Daniela Berg
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Robert Göder
- Department of Psychiatry and Psychotherapy, Sleep Laboratory, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Peter Häussermann
- Department of Geriatric Psychiatry, LVR Klinik Köln, Academic Teaching Hospital, University of Cologne, Köln, Germany
| | - Thorsten Bartsch
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany.
| |
Collapse
|
155
|
Lin X, Feng T, Cui E, Li Y, Qin Z, Zhao X. A rat model established by simulating genetic-environmental interactions recapitulates human Alzheimer's disease pathology. Brain Res 2024; 1822:148663. [PMID: 37918702 DOI: 10.1016/j.brainres.2023.148663] [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: 08/02/2023] [Revised: 10/16/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND In humans, Alzheimer's disease (AD) is typically sporadic in nature, and its pathology is usually influenced by extensive factors. The study established a rat model based on the genetic-environmental interaction. METHODS A rat model was established by transduction of an adeno-associated virus combined with acrolein treatment. Rats were assigned to the normal control (NC), acrolein group, AAV (-) group, AAV-APP group, and AAV-APP/acrolein group. The success of model construction was verified in multiple ways, including by assessing cognitive function, examining microstructural changes in the brain in vivo, and performing immunohistochemistry. The contribution of genetic (APP mutation) and environmental (acrolein) factors to AD-like phenotypes in the model was explored by factorial analysis. RESULTS 1) The AAV-APP/acrolein group showed a decline in cognitive function, as indicated by a reduced gray matter volume in key cognition-related brain areas, lower FA values in the hippocampus and internal olfactory cortex, and Aβ deposition in the cortex and hippocampus. 2) The AAV-APP group also showed a decline in cognitive function, although the group exhibited atypical brain atrophy in the gray matter and insignificant Aβ deposition. 3) The acrolein group did not show any significant changes in Aβ levels, gray matter volume, or cognitive function. 4) The genetic factor (APP mutation) explained 39.74% of the AD-like phenotypes in the model factors, and the environmental factor (acrolein exposure) explained 33.3%. CONCLUSIONS The genetic-environmental interaction rat model exhibited a phenotype that resembled the features of human AD and will be useful for research on AD.
Collapse
Affiliation(s)
- Xiaomei Lin
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China
| | - Tianyuyi Feng
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China
| | - Erheng Cui
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China
| | - Yunfei Li
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China
| | - Zhang Qin
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China
| | - Xiaohu Zhao
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China.
| |
Collapse
|
156
|
Sánchez-Soblechero A, López-García S, Lage C, Fernández-Matarrubia M, Irure J, López-Hoyos M, Jiménez-Bonilla J, Quirce R, de Arcocha-Torres M, Cuenca-Vera O, Martín-Arroyo J, Martínez-Dubarbie F, Pozueta A, García-Martínez M, Infante J, Sánchez-Juan P, Rodríguez-Rodríguez E. Where Should I Draw the Line: PET-Driven, Data-Driven, or Manufacturer Cut-Off? J Alzheimers Dis 2024; 98:957-967. [PMID: 38489172 DOI: 10.3233/jad-230678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Background The optimal cut-off for Alzheimer's disease (AD) CSF biomarkers remains controversial. Objective To analyze the performance of cut-off points standardized by three methods: one that optimized the agreement between 11C-Pittsburgh compound B PET (a-PET) and CSF biomarkers (Aβ1-42, pTau, tTau, and Aβ1-42/Aβ1-40 ratio) in our population, called PET-driven; an unbiased cut-off using data from a healthy research cohort, called data-driven, and that provided by the manufacturer. We also compare changes in ATN classification. Methods CSF biomarkers measured by the LUMIPULSE G600II platform and qualitative visualization of amyloid positron emission tomography (a-PET) were performed in all the patients. We established a cut-off for each single biomarker and Aβ1-42/Aβ1-40 ratio that optimized their agreement with a-PET using ROC curves. Sensitivity, Specificity, and Overall Percent of Agreement are assessed using a-PET or clinical diagnosis as gold standard for every cut-off. Also, we established a data-driven cut-off from our cognitively unimpaired cohort. We then analyzed changes in ATN classification. Results One hundred and ten patients were recruited. Sixty-six (60%) were a-PET positive. PET-driven cut-offs were: pTau > 57, tTau > 362.62, Aβ1-42/Aβ1-40 < 0.069. For a single biomarker, pTau showed the highest accuracy (AUC 0.926). New PET-driven cut-offs classified patients similarly to manufacturer cut-offs (only two patients changed). However, 20 patients (18%) changed when data-driven cut-offs were used. Conclusions We established our sample's best CSF biomarkers cut-offs using a-PET as the gold standard. These cut-offs categorize better symptomatic subjects than data-driven in ATN classification, but they are very similar to the manufacturer's.
Collapse
Affiliation(s)
| | - Sara López-García
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Carmen Lage
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Marta Fernández-Matarrubia
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Irure
- Immunology Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain
| | - Marcos López-Hoyos
- Immunology Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain
| | - Julio Jiménez-Bonilla
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Nuclear Medicine Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
| | - Remedios Quirce
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Nuclear Medicine Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
| | - María de Arcocha-Torres
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Nuclear Medicine Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
| | - Oriana Cuenca-Vera
- Nuclear Medicine Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
| | - Juan Martín-Arroyo
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
| | - Francisco Martínez-Dubarbie
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ana Pozueta
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - María García-Martínez
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jon Infante
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain
| | - Pascual Sánchez-Juan
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Alzheimer's Centre Reina Sofia-CIEN Foundation-ISCIII, Madrid, Spain
| | - Eloy Rodríguez-Rodríguez
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain
| |
Collapse
|
157
|
Baril A, Picard C, Labonté A, Sanchez E, Duclos C, Mohammediyan B, Ashton NJ, Zetterberg H, Blennow K, Breitner JCS, Villeneuve S, Poirier J. Day-to-day sleep variability with Alzheimer's biomarkers in at-risk elderly. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12521. [PMID: 38371359 PMCID: PMC10870017 DOI: 10.1002/dad2.12521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 02/20/2024]
Abstract
INTRODUCTION Measuring day-to-day sleep variability might reveal unstable sleep-wake cycles reflecting neurodegenerative processes. We evaluated the association between Alzheimer's disease (AD) fluid biomarkers with day-to-day sleep variability. METHODS In the PREVENT-AD cohort, 203 dementia-free participants (age: 68.3 ± 5.4; 78 males) with a parental history of sporadic AD were tested with actigraphy and fluid biomarkers. Day-to-day variability (standard deviations over a week) was assessed for sleep midpoint, duration, efficiency, and nighttime activity count. RESULTS Lower cerebrospinal fluid (CSF) ApoE, higher CSF p-tau181/amyloid-β (Aβ)42, and higher plasma p-tau231/Aβ42 were associated with higher variability of sleep midpoint, sleep duration, and/or activity count. The associations between fluid biomarkers with greater sleep duration variability were especially observed in those that carried the APOE4 allele, mild cognitive impairment converters, or those with gray matter atrophy. DISCUSSION Day-to-day sleep variability were associated with biomarkers of AD in at-risk individuals, suggesting that unstable sleep promotes neurodegeneration or, conversely, that AD neuropathology disrupts sleep-wake cycles.
Collapse
Affiliation(s)
- Andrée‐Ann Baril
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Cynthia Picard
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Anne Labonté
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Erlan Sanchez
- Sunnybrook Research InstituteUniversity of TorontoTorontoOntarioCanada
| | - Catherine Duclos
- Hôpital du Sacré‐Coeur de MontréalCIUSSS‐NIMMontréalQuébecCanada
- Department of Anesthesiology and Pain MedicineUniversité de MontréalMontréalQuébecCanada
| | - Béry Mohammediyan
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- King's College LondonInstitute of PsychiatryPsychology and Neuroscience Maurice Wohl Institute Clinical Neuroscience InstituteLondonUK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS FoundationLondonUK
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayHong KongChina
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - John C. S. Breitner
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Sylvia Villeneuve
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | - Judes Poirier
- Douglas Mental Health University InstituteMcGill UniversityMontrealQuébecCanada
| | | |
Collapse
|
158
|
Puig‐Pijoan A, Jimenez‐Balado J, Fernández‐Lebrero A, García‐Escobar G, Navalpotro‐Gómez I, Contador J, Manero‐Borràs R, Puente‐Periz V, Suárez A, Muñoz FJ, Grau‐Rivera O, Suárez‐Calvet M, de la Torre R, Roquer J, Ois A. Risk of cognitive decline progression is associated to increased blood-brain-barrier permeability: A longitudinal study in a memory unit clinical cohort. Alzheimers Dement 2024; 20:538-548. [PMID: 37727082 PMCID: PMC10916969 DOI: 10.1002/alz.13433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/21/2023]
Abstract
INTRODUCTION This study examined the relationship between blood-brain-barrier permeability (BBBp), measured by cerebrospinal fluid/serum albumin ratio (QAlb), and cognitive decline progression in a clinical cohort. METHODS This prospective observational study included 334 participants from the BIODEGMAR cohort. Cognitive decline progression was defined as an increase in Global Deterioration Scale and/or Clinical Dementia Rating scores. Associations between BBBp, demographics, and clinical factors were explored. RESULTS Male sex, diabetes mellitus, and cerebrovascular burden were associated with increased log-QAlb. Vascular cognitive impairment patients had the highest log-QAlb levels. Among the 273 participants with valid follow-up data, 154 (56.4%) showed cognitive decline progression. An 8% increase in the hazard of clinical worsening was observed for each 10% increase in log-QAlb. DISCUSSION These results suggest that increased BBBp in individuals with cognitive decline may contribute to clinical worsening, pointing to potential targeted therapies. QAlb could be a useful biomarker for identifying patients with a worse prognosis.
Collapse
|
159
|
Aye S, Handels R, Winblad B, Jönsson L. Optimising Alzheimer's Disease Diagnosis and Treatment: Assessing Cost-Utility of Integrating Blood Biomarkers in Clinical Practice for Disease-Modifying Treatment. J Prev Alzheimers Dis 2024; 11:928-942. [PMID: 39044504 PMCID: PMC11266371 DOI: 10.14283/jpad.2024.67] [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: 12/15/2023] [Accepted: 02/28/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Recent developments in blood biomarkers (BBM) have shown promising results in diagnosing amyloid pathology in Alzheimer's Disease (AD). However, information on how these BBMs can best be used in clinical settings to optimise clinical decision-making and long-term health outcomes for individuals with AD is still lacking. OBJECTIVES We aim to assess the potential value of BBM in AD diagnosis within the context of disease-modifying treatment (DMT). DESIGN We developed a decision analytic model to evaluate the long-term health outcomes using BBM in AD diagnosis. We compared standard of care (SOC) diagnosis workflow to the integration of BBM as a (1) referral decision tool in primary health center (PHC) and (2) triaging tool for invasive CSF examination in specialist memory clinic (MC). We combined a decision tree and a Markov model to simulate the patient's diagnostic journey, treatment decisions following diagnosis and long-term health outcomes. Input parameters for the model were identified from published literature and registry data analysis. We conducted a cost-utility analysis from the societal perspective using a one-year cycle length and a 30-year (lifetime) horizon. MEASUREMENTS We reported the simulated outcomes in the percentage of correct diagnosis, costs (in 2022 Euros), quality-adjusted life year (QALY), and incremental cost-effectiveness ratios (ICER) associated with each diagnosis strategy. RESULTS Compared to SOC, integrating BBM in PHC increased patient referrals by 8% and true positive AD diagnoses by 10.4%. The lifetime costs for individuals diagnosed with AD were € 249,685 and €250,287, and QALYs were 9.5 and 9.52 in SOC and PHC pathways, respectively. The cost increments were €603, and QALYs gained were 0.01, resulting in an ICER of €48,296. Using BBM in MC reduced the exposure to invasive CSF procedures and costs but also reduced true positive AD diagnoses and QALYs. CONCLUSIONS Using BBM at PHC to make referral decisions might increase initial diagnostic costs but can prevent high costs associated with disease progression, providing a cost-effective DMT is available, whereas using BBM in MC could reduce the initial evaluation cost but incur high costs associated with disease progression.
Collapse
Affiliation(s)
- S Aye
- Sandar Aye, Karolinska Institutet, BioClinicum J9:20, Akademiska stråket 171 64 Solna, Sweden, Phone: +46 704347761,
| | | | | | | |
Collapse
|
160
|
Nallapu BT, Petersen KK, Lipton RB, Davatzikos C, Ezzati A. Plasma Biomarkers as Predictors of Progression to Dementia in Individuals with Mild Cognitive Impairment. J Alzheimers Dis 2024; 98:231-246. [PMID: 38393899 PMCID: PMC11044769 DOI: 10.3233/jad-230620] [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: 02/25/2024]
Abstract
Background Blood-based biomarkers (BBMs) are of growing interest in the field of Alzheimer's disease (AD) and related dementias. Objective This study aimed to assess the ability of plasma biomarkers to 1) predict disease progression from mild cognitive impairment (MCI) to dementia and 2) improve the predictive ability of magnetic resonance imaging (MRI) and cerebrospinal fluid (CSF) measures when combined. Methods We used data from the Alzheimer's Disease Neuroimaging Initiative. Machine learning models were trained using the data from participants who remained cognitively stable (CN-s) and with Dementia diagnosis at 2-year follow-up visit. The models were used to predict progression to dementia in MCI individuals. We assessed the performance of models with plasma biomarkers against those with CSF and MRI measures, and also in combination with them. Results Our models with plasma biomarkers classified CN-s individuals from AD with an AUC of 0.75±0.03 and could predict conversion to dementia in MCI individuals with an AUC of 0.64±0.03 (17.1% BP, base prevalence). Models with plasma biomarkers performed better when combined with CSF and MRI measures (CN versus AD: AUC of 0.89±0.02; MCI-to-AD: AUC of 0.76±0.03, 21.5% BP). Conclusions Our results highlight the potential of plasma biomarkers in predicting conversion to dementia in MCI individuals. While plasma biomarkers could improve the predictive ability of CSF and MRI measures when combined, they also show the potential to predict non-progression to AD when considered alone. The predictive ability of plasma biomarkers is crucially linked to reducing the costly and effortful collection of CSF and MRI measures.
Collapse
Affiliation(s)
- Bhargav T. Nallapu
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - Kellen K. Petersen
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - Richard B. Lipton
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - Christos Davatzikos
- Radiology Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ali Ezzati
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | | |
Collapse
|
161
|
Jethwa A, Stöckl L. Optimized Pre-analytical Handling Protocol for Blood-Based Biomarkers of Alzheimer's Disease. Methods Mol Biol 2024; 2785:67-73. [PMID: 38427188 DOI: 10.1007/978-1-0716-3774-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The therapeutic management of patients with Alzheimer's disease (AD) has been hindered by poor diagnostic accuracy. As such, there is an unmet clinical need for tools that can detect and diagnose the disease in its early stages. Compared with cerebrospinal fluid (CSF)-based biomarkers or positron emission tomography (PET), the use of reliable blood-based biomarkers could offer an accessible and minimally invasive method of streamlining diagnosis in the clinical setting. However, the influence of pre-analytical processing and sample handling parameters on the accurate measurement of protein biomarkers is well established, especially for AD CSF-based biomarkers. In this chapter, we provide recommendations for an optimal sample handling protocol for the analysis of blood-based biomarkers specifically for amyloid pathology in AD.
Collapse
|
162
|
Wang RT, Sun Z, Tan CC, Tan L, Xu W. Dynamic Features of Body Mass Index in Late Life Predict Cognitive Trajectories and Alzheimer's Disease: A Longitudinal Study. J Alzheimers Dis 2024; 100:1365-1378. [PMID: 39031359 DOI: 10.3233/jad-240292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2024]
Abstract
Background The causal relationships of late-life body mass index (BMI) with Alzheimer's disease (AD) remains debated. Objective We aimed to assess the associations of dynamic BMI features (ΔBMIs) with cognitive trajectories, AD biomarkers, and incident AD risk. Methods We analyzed an 8-year cohort of 542 non-demented individuals who were aged ≥65 years at baseline and had BMI measurements over the first 4 years. ΔBMIs were defined as changing extent (change ≤ or >5%), variability (standard deviation), and trajectories over the first 4 years measured using latent class trajectory modeling. Linear mixed-effect models were utilized to examine the influence of ΔBMIs on changing rates of AD pathology biomarkers, hippocampus volume, and cognitive functions. Cox proportional hazards models were used to test the associations with AD risk. Stratified analyzes were conducted by the baseline BMI group and age. Results Over the 4-year period, compared to those with stable BMI, individuals who experienced BMI decreases demonstrated accelerated declined memory function (p = 0.006) and amyloid-β deposition (p = 0.034) while BMI increases were associated with accelerated hippocampal atrophy (p = 0.036). Three BMI dynamic features, including stable BMI, low BMI variability, and persistently high BMI, were associated with lower risk of incident AD (p < 0.005). The associations were validated over the 8-year period after excluding incident AD over the first 4 years. No stratified effects were revealed by the BMI group and age. Conclusions High and stable BMI in late life could predict better cognitive trajectory and lower risk of AD.
Collapse
Affiliation(s)
- Ruo-Tong Wang
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zhen Sun
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
163
|
Memon A, Moore JA, Kang C, Ismail Z, Forkert ND. Visual Functions Are Associated with Biomarker Changes in Alzheimer's Disease. J Alzheimers Dis 2024; 99:623-637. [PMID: 38669529 DOI: 10.3233/jad-231084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Background While various biomarkers of Alzheimer's disease (AD) have been associated with general cognitive function, their association to visual-perceptive function across the AD spectrum warrant more attention due to its significant impact on quality of life. Thus, this study explores how AD biomarkers are associated with decline in this cognitive domain. Objective To explore associations between various fluid and imaging biomarkers and visual-based cognitive assessments in participants across the AD spectrum. Methods Data from participants (N = 1,460) in the Alzheimer's Disease Neuroimaging Initiative were analyzed, including fluid and imaging biomarkers. Along with the Mini-Mental State Examination (MMSE), three specific visual-based cognitive tests were investigated: Trail Making Test (TMT) A and TMT B, and the Boston Naming Test (BNT). Locally estimated scatterplot smoothing curves and Pearson correlation coefficients were used to examine associations. Results MMSE showed the strongest correlations with most biomarkers, followed by TMT-B. The p-tau181/Aβ1-42 ratio, along with the volume of the hippocampus and entorhinal cortex, had the strongest associations among the biomarkers. Conclusions Several biomarkers are associated with visual processing across the disease spectrum, emphasizing their potential in assessing disease severity and contributing to progression models of visual function and cognition.
Collapse
Affiliation(s)
- Ashar Memon
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jasmine A Moore
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Program, University of Calgary, Calgary, AB, Canada
| | - Chris Kang
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Zahinoor Ismail
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Departments of Clinical Neurosciences, Psychiatry, Community Health Sciences, and Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Nils D Forkert
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Departments of Clinical Neurosciences, Psychiatry, Community Health Sciences, and Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Departments of Clinical Neurosciences, Psychiatry, Community Health Sciences, and Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
164
|
Zhu Y, Wu Y, Lv X, Wu J, Shen C, Tang Q, Wang G. The relationship between APOE genotype, CSF Tau and cognition across the Alzheimer's disease spectrum, moderation and mediation role of insula network connectivity. CNS Neurosci Ther 2024; 30:e14401. [PMID: 37577852 PMCID: PMC10805399 DOI: 10.1111/cns.14401] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/07/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023] Open
Abstract
AIMS To investigate whether insula network connectivity modulates the relationship between apolipoprotein E (APOE) ε4 genotype, cerebrospinal fluid (CSF) biomarkers (Aβ, Tau, and pTau) and cognition across Alzheimer's disease (AD) spectrum. METHODS Forty-six cognitive normal (CN), 35 subjective memory complaint (SMC), 41 mild cognitive impairment (MCI), and 32 AD subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) were obtained. Multivariable linear regression analyses were conducted to investigate the main effects and interaction of the APOE genotype and disease status on the insula functional connectivity (IFC) network. Mediation and moderation analysis were performed to investigate whether IFC strengths regulate the association between APOE genotype, CSF biomarkers and cognition. Additionally, the support vector machine (SVM) model integrating APOE genotype, CSF biomarkers, and neuroimaging biomarkers (insula volumes and altered regional IFCs) was used to classify the AD spectrum. RESULTS The interactive effect of the APOE genotype and disease on the insula network was found in the left medial superior frontal gyrus (SFGmed.L), right anterior medial prefrontal cortex (aMPFC.R), and bilateral thalamus (THA.B). The functional connectivities (FCs) in the left insula (LIns) connecting with the left posterior middle temporal gyrus (pMTG.L), SFGmed.L, and right lingual gyrus (LING.R) were correlated with cognition. LIns-SFGmed.L and LIns-pMTG.L FCs could moderate the effects of Tau on cognition. Furthermore, LIns-SFGmed.L FC may suppress the association between APOE genotype and cognition. More importantly, the integrated biomarkers from the SVM model yielded strong powers for classifying the AD spectrum. CONCLUSIONS Insula functional connectivity regulated the association between APOE genotype, CSF Tau and cognition and provided stage-dependent biomarkers for early differentiation of the AD spectrum. The present study used a cross-sectional design. Follow-up studies are needed to validate the relationship.
Collapse
Affiliation(s)
- Yao Zhu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Yan Wu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Xinyi Lv
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Jiaonan Wu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Chunzi Shen
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Qiqiang Tang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Guoping Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | | |
Collapse
|
165
|
Seligmann B, Camiolo S, Hernandez M, Yeakley JM, Sahagian G, McComb J. Molecular Gene Expression Testing to Identify Alzheimer's Disease with High Accuracy from Fingerstick Blood. J Alzheimers Dis 2024; 101:813-822. [PMID: 39269833 PMCID: PMC11492108 DOI: 10.3233/jad-240174] [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: 09/15/2024]
Abstract
Background There is no molecular test for Alzheimer's disease (AD) using self-collected samples, nor is there a definitive molecular test for AD. We demonstrate an accurate and potentially definitive TempO-Seq® gene expression test for AD using fingerstick blood spotted and dried on filter paper, a sample that can be collected in any doctor's office or can be self-collected. Objective Demonstrate the feasibility of developing an accurate test for the classification of persons with AD from a minimally invasive sample of fingerstick blood spotted on filter paper which can be obtained in any doctor's office or self-collected to address health disparities. Methods Fingerstick blood samples from patients clinically diagnosed with AD, Parkinson's disease (PD), or asymptomatic controls were spotted onto filter paper in the doctor's office, dried, and shipped to BioSpyder for testing. Three independent patient cohorts were used for training/retraining and testing/retesting AD and PD classification algorithms. Results After initially identifying a 770 gene classification signature, a minimum set of 68 genes was identified providing classification test areas under the ROC curve of 0.9 for classifying patients as having AD, and 0.94 for classifying patients as having PD. Conclusions These data demonstrate the potential to develop a screening and/or definitive, minimally invasive, molecular diagnostic test for AD and PD using dried fingerstick blood spot samples that are collected in a doctor's office or clinic, or self-collected, and thus, can address health disparities. Whether the test can classify patients with AD earlier then possible with cognitive testing remains to be determined.
Collapse
Affiliation(s)
| | | | | | | | | | - Joel McComb
- BioSpyder Technologies, Inc., Carlsbad, CA, USA
| |
Collapse
|
166
|
Cartas-Cejudo P, Cortés A, Lachén-Montes M, Anaya-Cubero E, Peral E, Ausín K, Díaz-Peña R, Fernández-Irigoyen J, Santamaría E. Mapping the human brain proteome: opportunities, challenges, and clinical potential. Expert Rev Proteomics 2024; 21:55-63. [PMID: 38299555 DOI: 10.1080/14789450.2024.2313073] [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: 09/25/2023] [Accepted: 01/24/2024] [Indexed: 02/02/2024]
Abstract
INTRODUCTION Due to the segmented functions and complexity of the human brain, the characterization of molecular profiles within specific areas such as brain structures and biofluids is essential to unveil the molecular basis for structure specialization as well as the molecular imbalance associated with neurodegenerative and psychiatric diseases. AREAS COVERED Much of our knowledge about brain functionality derives from neurophysiological, anatomical, and transcriptomic approaches. More recently, laser capture and imaging proteomics, technological and computational developments in LC-MS/MS, as well as antibody/aptamer-based platforms have allowed the generation of novel cellular, spatial, and posttranslational dimensions as well as innovative facets in biomarker validation and druggable target identification. EXPERT OPINION Proteomics is a powerful toolbox to functionally characterize, quantify, and localize the extensive protein catalog of the human brain across physiological and pathological states. Brain function depends on multi-dimensional protein homeostasis, and its elucidation will help us to characterize biological pathways that are essential to properly maintain cognitive functions. In addition, comprehensive human brain pathological proteomes may be the basis in computational drug-repositioning methods as a strategy for unveiling potential new therapies in neurodegenerative and psychiatric disorders.
Collapse
Affiliation(s)
- Paz Cartas-Cejudo
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Adriana Cortés
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Mercedes Lachén-Montes
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Elena Anaya-Cubero
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Erika Peral
- Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Karina Ausín
- Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Ramón Díaz-Peña
- Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
167
|
Frank B, Walsh M, Hurley L, Groh J, Blennow K, Zetterberg H, Tripodis Y, Budson AE, O'Connor MK, Martin B, Weller J, McKee A, Qiu W, Stein TD, Stern RA, Mez J, Henson R, Long J, Aschenbrenner AJ, Babulal GM, Morris JC, Schindler S, Alosco ML. Cognition Mediates the Association Between Cerebrospinal Fluid Biomarkers of Amyloid and P-Tau and Neuropsychiatric Symptoms. J Alzheimers Dis 2024; 100:1055-1073. [PMID: 38995786 PMCID: PMC11805585 DOI: 10.3233/jad-240125] [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: 07/14/2024]
Abstract
Background Neuropsychiatric symptoms (NPS) can be an early manifestation of Alzheimer's disease (AD). However, the associations among NPS, cognition, and AD biomarkers across the disease spectrum are unclear. Objective We analyzed cross-sectional mediation pathways between cerebrospinal fluid (CSF) biomarkers of AD (Aβ1-42, p-tau181), cognitive function, and NPS. Methods Primary models included 781 participants from the National Alzheimer's Coordinating Center (NACC) data set who had CSF analyzed for AD biomarkers using Lumipulse. NPS were assessed with the Neuropsychiatric Inventory Questionnaire (NPI-Q). We assessed cognition with the harmonized MMSE/MoCA, as well as neuropsychological tests sensitive to AD pathology: story recall, naming, animal fluency, and Trails B. The Clinical Dementia Rating (CDR®) scale assessed dementia severity. Mediation models were estimated with Kemeny metric covariance in a structural equation model framework, controlling for age, education, sex, and APOEɛ4. Results The sample was older adults (M = 73.85, SD = 6.68; 49.9% male, 390; 27.9% dementia, 218) who were predominantly white (n = 688, 88.1%). Higher p-tau181/Aβ1-42 ratio predicted higher NPI-Q, which was partially mediated by the MMSE/MoCA and, in a second model, story recall. No other pathway was statistically significant. Both the MMSE/MoCA and NPI-Q independently mediated the association between p-tau181/Aβ1-42 ratio and CDR global impairment. With dementia excluded, p-tau181/Aβ1-42 ratio was no longer associated with the NPI-Q. Conclusions NPS may be secondary to cognitive impairment and AD pathology through direct and indirect pathways. NPS independently predict dementia severity in AD. However, AD pathology likely plays less of a role in NPS in samples without dementia.
Collapse
Affiliation(s)
- Brandon Frank
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Michael Walsh
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Landon Hurley
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Boston, MA, USA
| | - Jenna Groh
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Yorghos Tripodis
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Andrew E Budson
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Maureen K O'Connor
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, U.S. Department of Veteran Affairs, Bedford, MA, USA
| | - Brett Martin
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Jason Weller
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Ann McKee
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Wendy Qiu
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Robert A Stern
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
- Department of Neurosurgery, Boston University School of Medicine, Boston, MA, USA
| | - Jesse Mez
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Framingham Heart Study, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Rachel Henson
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - Justin Long
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - Ganesh M Babulal
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - Suzanne Schindler
- Knight Alzheimer Disease Research Center (ADRC), Washington University, St. Louis, MO, USA
| | - Michael L Alosco
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
168
|
Guha D, Misra V, Chettimada S, Yin J, Gabuzda D. CSF Extracellular Vesicle Aβ42 and Tau/Aβ42 Ratio Are Associated with Cognitive Impairment in Older People with HIV. Viruses 2023; 16:72. [PMID: 38257772 PMCID: PMC10818296 DOI: 10.3390/v16010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 01/24/2024] Open
Abstract
HIV-associated neurocognitive disorders (HAND) remain prevalent despite viral suppression on antiretroviral therapy (ART). Older people with HIV (PWH) are also at risk for amnestic mild cognitive impairment (aMCI) and Alzheimer's disease (AD). β-amyloid (Aβ) and Tau biomarkers are associated with aMCI/AD, but their relationship to HAND is unclear. Given the role of extracellular vesicles (EVs) in age-related neurological disorders, we investigated soluble and EV-associated Aβ42, total Tau, NFL, GFAP, ICAM-1, VCAM-1, and CRP in relation to cognitive impairment in PWH. Plasma and CSF EVs were isolated from 184 participants (98 PWH on ART and 86 HIV- controls). Biomarkers were measured using Meso Scale Discovery assays. The median age of PWH was 53 years, and 52% were diagnosed with mild forms of HAND. PWH had increased plasma NFL (p = 0.04) and CSF Aβ42 (p = 0.0003) compared with HIV- controls but no significant difference in Tau or EV-associated forms of these markers. CSF EV Aβ42 was decreased (p = 0.0002) and CSF EV Tau/Aβ42 ratio was increased (p = 0.001) in PWH with HAND vs. no HAND, while soluble forms of these markers showed no significant differences. Decreased CSF EV Aβ42 (p < 0.0001) and an increased CSF EV Tau/Aβ42 ratio (p = 0.0003) were associated with lower neurocognitive T scores in age-adjusted models; an optimal model included both CSF EV Aβ42 and plasma NFL. Levels of soluble, but not EV-associated, ICAM-1, VCAM-1, and CRP were increased in PWH with HAND vs. no HAND (p < 0.05). These findings suggest that decreased Aβ42 and an increased Tau/Aβ42 ratio in CSF EVs are associated with cognitive impairment in older PWH, and these EV-associated biomarkers may help to distinguish aMCI/AD from HIV-related cognitive disorders in future studies.
Collapse
Affiliation(s)
- Debjani Guha
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Vikas Misra
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sukrutha Chettimada
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jun Yin
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Dana Gabuzda
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
169
|
Yang Z, Wen J, Erus G, Govindarajan ST, Melhem R, Mamourian E, Cui Y, Srinivasan D, Abdulkadir A, Parmpi P, Wittfeld K, Grabe HJ, Bülow R, Frenzel S, Tosun D, Bilgel M, An Y, Yi D, Marcus DS, LaMontagne P, Benzinger TL, Heckbert SR, Austin TR, Waldstein SR, Evans MK, Zonderman AB, Launer LJ, Sotiras A, Espeland MA, Masters CL, Maruff P, Fripp J, Toga A, O’Bryant S, Chakravarty MM, Villeneuve S, Johnson SC, Morris JC, Albert MS, Yaffe K, Völzke H, Ferrucci L, Bryan NR, Shinohara RT, Fan Y, Habes M, Lalousis PA, Koutsouleris N, Wolk DA, Resnick SM, Shou H, Nasrallah IM, Davatzikos C. Five dominant dimensions of brain aging are identified via deep learning: associations with clinical, lifestyle, and genetic measures. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.29.23300642. [PMID: 38234857 PMCID: PMC10793523 DOI: 10.1101/2023.12.29.23300642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Brain aging is a complex process influenced by various lifestyle, environmental, and genetic factors, as well as by age-related and often co-existing pathologies. MRI and, more recently, AI methods have been instrumental in understanding the neuroanatomical changes that occur during aging in large and diverse populations. However, the multiplicity and mutual overlap of both pathologic processes and affected brain regions make it difficult to precisely characterize the underlying neurodegenerative profile of an individual from an MRI scan. Herein, we leverage a state-of-the art deep representation learning method, Surreal-GAN, and present both methodological advances and extensive experimental results that allow us to elucidate the heterogeneity of brain aging in a large and diverse cohort of 49,482 individuals from 11 studies. Five dominant patterns of neurodegeneration were identified and quantified for each individual by their respective (herein referred to as) R-indices. Significant associations between R-indices and distinct biomedical, lifestyle, and genetic factors provide insights into the etiology of observed variances. Furthermore, baseline R-indices showed predictive value for disease progression and mortality. These five R-indices contribute to MRI-based precision diagnostics, prognostication, and may inform stratification into clinical trials.
Collapse
Affiliation(s)
- Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja T. Govindarajan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Paraskevi Parmpi
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University of Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Daniel S. Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan R. Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Thomas R. Austin
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Shari R. Waldstein
- Department of Psychology, University of Maryland, Baltimore County, Catonsville, MD, USA
| | - Michele K. Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Alan B. Zonderman
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Lenore J. Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, USA
| | - Aristeidis Sotiras
- Department of Radiology and Institute of Informatics, Washington University in St. Luis, St. Luis, MO63110, USA
| | - Mark A. Espeland
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Colin L. Masters
- Florey Institute, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Paul Maruff
- Florey Institute, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, Queensland, Australia
| | - Arthur Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Sid O’Bryant
- Institute for Translational Research University of North Texas Health Science Center Fort Worth Texas USA
| | - Mallar M. Chakravarty
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - John C. Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Marilyn S. Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Yaffe
- Departments of Neurology, Psychiatry and Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, 3001 S. Hanover Street, Baltimore, MD, USA
| | - Nick R. Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Russell T. Shinohara
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Biggs Alzheimer’s Institute, University of Texas San Antonio Health Science Center, USA
| | - Paris Alexandros Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Nikolaos Koutsouleris
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- Section for Precision Psychiatry, Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - David A. Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M. Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
170
|
Dyer AH, Dolphin H, O'Connor A, Morrison L, Sedgwick G, McFeely A, Killeen E, Gallagher C, Davey N, Connolly E, Lyons S, Young C, Gaffney C, Ennis R, McHale C, Joseph J, Knight G, Kelly E, O'Farrelly C, Bourke NM, Fallon A, O'Dowd S, Kennelly SP. Protocol for the Tallaght University Hospital Institute for Memory and Cognition-Biobank for Research in Ageing and Neurodegeneration. BMJ Open 2023; 13:e077772. [PMID: 38070888 PMCID: PMC10729202 DOI: 10.1136/bmjopen-2023-077772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
INTRODUCTION Alzheimer's disease and other dementias affect >50 million individuals globally and are characterised by broad clinical and biological heterogeneity. Cohort and biobank studies have played a critical role in advancing the understanding of disease pathophysiology and in identifying novel diagnostic and treatment approaches. However, further discovery and validation cohorts are required to clarify the real-world utility of new biomarkers, facilitate research into the development of novel therapies and advance our understanding of the clinical heterogeneity and pathobiology of neurodegenerative diseases. METHODS AND ANALYSIS The Tallaght University Hospital Institute for Memory and Cognition Biobank for Research in Ageing and Neurodegeneration (TIMC-BRAiN) will recruit 1000 individuals over 5 years. Participants, who are undergoing diagnostic workup in the TIMC Memory Assessment and Support Service (TIMC-MASS), will opt to donate clinical data and biological samples to a biobank. All participants will complete a detailed clinical, neuropsychological and dementia severity assessment (including Addenbrooke's Cognitive Assessment, Repeatable Battery for Assessment of Neuropsychological Status, Clinical Dementia Rating Scale). Participants undergoing venepuncture/lumbar puncture as part of the clinical workup will be offered the opportunity to donate additional blood (serum/plasma/whole blood) and cerebrospinal fluid samples for longitudinal storage in the TIMC-BRAiN biobank. Participants are followed at 18-month intervals for repeat clinical and cognitive assessments. Anonymised clinical data and biological samples will be stored securely in a central repository and used to facilitate future studies concerned with advancing the diagnosis and treatment of neurodegenerative diseases. ETHICS AND DISSEMINATION Ethical approval has been granted by the St. James's Hospital/Tallaght University Hospital Joint Research Ethics Committee (Project ID: 2159), which operates in compliance with the European Communities (Clinical Trials on Medicinal Products for Human Use) Regulations 2004 and ICH Good Clinical Practice Guidelines. Findings using TIMC-BRAiN will be published in a timely and open-access fashion.
Collapse
Affiliation(s)
- Adam H Dyer
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Helena Dolphin
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | | | - Laura Morrison
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Gavin Sedgwick
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Aoife McFeely
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Emily Killeen
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Conal Gallagher
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Naomi Davey
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Eimear Connolly
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Shane Lyons
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland
| | - Conor Young
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Christine Gaffney
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland
| | - Ruth Ennis
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Cathy McHale
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Jasmine Joseph
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Graham Knight
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | - Emmet Kelly
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
| | | | - Nollaig M Bourke
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Aoife Fallon
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Sean O'Dowd
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland
- Academic Unit of Neurology, Trinity College Dublin, Dublin, Ireland
| | - Sean P Kennelly
- Institute of Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
171
|
Mitsunaga S, Fujito N, Nakaoka H, Imazeki R, Nagata E, Inoue I. Detection of APP gene recombinant in human blood plasma. Sci Rep 2023; 13:21703. [PMID: 38066066 PMCID: PMC10709617 DOI: 10.1038/s41598-023-48993-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
The pathogenesis of Alzheimer's disease (AD) is believed to involve the accumulation of amyloid-β in the brain, which is produced by the sequential cleavage of amyloid precursor protein (APP) by β-secretase and γ-secretase. Recently, analysis of genomic DNA and mRNA from postmortem brain neurons has revealed intra-exonic recombinants of APP (gencDNA), which have been implicated in the accumulation of amyloid-β. In this study, we computationally analyzed publicly available sequence data (SRA) using probe sequences we constructed to screen APP gencDNAs. APP gencDNAs were detected in SRAs constructed from both genomic DNA and RNA obtained from the postmortem brain and in the SRA constructed from plasma cell-free mRNA (cf-mRNA). The SRA constructed from plasma cf-mRNA showed a significant difference in the number of APP gencDNA reads between SAD and NCI: the p-value from the Mann-Whitney U test was 5.14 × 10-6. The transcripts were also found in circulating nucleic acids (CNA) from our plasma samples with NGS analysis. These data indicate that transcripts of APP gencDNA can be detected in blood plasma and suggest the possibility of using them as blood biomarkers for Alzheimer's disease.
Collapse
Affiliation(s)
- Shigeki Mitsunaga
- Laboratory of Human Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan.
| | - Naoko Fujito
- Laboratory of Human Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan
- Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, 411-8540, Japan
| | - Hirofumi Nakaoka
- Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Ryoko Imazeki
- Department of Neurology, Tokai University School of Medicine, Isehara, Japan
| | - Eiichiro Nagata
- Department of Neurology, Tokai University School of Medicine, Isehara, Japan
| | - Ituro Inoue
- Laboratory of Human Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan.
| |
Collapse
|
172
|
Gao Y, Su D, Xue Z, Ji L, Wang S. Association Between Serum Neurofilament Light Chain and Cognitive Performance Among Older Adults in the United States: A Cross-Sectional Study. Neurol Ther 2023; 12:2147-2160. [PMID: 37845473 PMCID: PMC10630257 DOI: 10.1007/s40120-023-00555-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 09/29/2023] [Indexed: 10/18/2023] Open
Abstract
INTRODUCTION Serum neurofilament light chain (sNfL) is an emerging biomarker of neuronal damage in several neurological disorders. Its association with cognitive function in the general US population aged 60 years and above is unknown. The aim of this study was to investigate the correlation between sNfL and cognitive function in the general US population aged 60 and above. METHODS The data were obtained from the 2013-2014 National Health and Nutrition Examination Survey (NHANES), which include 506 individuals aged 60 or older who met our search criteria. In our study, sNfL levels were divided into two groups based on dichotomization (19.0 pg/mL). After adjusting for multiple covariates, it was found that the high sNfL group (≥ 19.0 pg/mL) had lower cognitive performance than the low sNfL group (< 19.0 pg/mL). This relationship was also stable in subgroup analysis. CONCLUSION In this sample of an American elderly population, higher sNfL levels are correlated with lower cognitive performance. Our findings suggest that sNfL may become a potential screening tool for early prediction and confirmation of cognitive damage.
Collapse
Affiliation(s)
- Yuanyuan Gao
- Affiliated Hospital Six of Nantong University, Yancheng Third People's Hospital, No. 75 Juchang Road, Yancheng, 224000, Jiangsu, China
- Yancheng Third People's Hospital, Yancheng, 224000, Jiangsu, China
| | - Dan Su
- Affiliated Hospital Six of Nantong University, Yancheng Third People's Hospital, No. 75 Juchang Road, Yancheng, 224000, Jiangsu, China
- Yancheng Third People's Hospital, Yancheng, 224000, Jiangsu, China
| | - Zhouya Xue
- The First People's Hospital of Yancheng, Yancheng, 224000, Jiangsu, China
| | - Lin Ji
- Affiliated Hospital Six of Nantong University, Yancheng Third People's Hospital, No. 75 Juchang Road, Yancheng, 224000, Jiangsu, China
- Yancheng Third People's Hospital, Yancheng, 224000, Jiangsu, China
| | - Shu Wang
- Affiliated Hospital Six of Nantong University, Yancheng Third People's Hospital, No. 75 Juchang Road, Yancheng, 224000, Jiangsu, China.
- Yancheng Third People's Hospital, Yancheng, 224000, Jiangsu, China.
| |
Collapse
|
173
|
Grari O, Elmoujtahide D, Sebbar E, Choukri M. The Biochemistry Behind Cognitive Decline: Biomarkers of Alzheimer's Disease. EJIFCC 2023; 34:276-283. [PMID: 38303754 PMCID: PMC10828533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia. Pathologically, the disease is marked by neurofibrillary tangles (NFT), which are aberrant accumulations of the tau protein that develop inside neurons, and extracellular plaque deposits of the amyloid β peptide (Aβ). These pathological lesions are present in the brain before the beginning of clinical manifestations. However, despite advancements in the comprehension of AD pathophysiology, timely and accurate clinical diagnosis remains challenging. Therefore, developing biomarkers capable of detecting AD during the preclinical phase holds enormous promise for precise diagnosis since detecting the disease early is crucial because it enables interventions when treatments may be more effective. This article intends to provide a comprehensive review of AD biomarkers, discussing their significance, classification, and recent developments in the field.
Collapse
Affiliation(s)
- O. Grari
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| | - D. Elmoujtahide
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| | - E. Sebbar
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| | - M. Choukri
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| |
Collapse
|
174
|
Lan G, Du J, Chen X, Wang Q, Han Y, Guo T. Association of APOE-ε4 and GAP-43-related presynaptic loss with β-amyloid, tau, neurodegeneration, and cognitive decline. Neurobiol Aging 2023; 132:209-219. [PMID: 37852045 DOI: 10.1016/j.neurobiolaging.2023.09.012] [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: 02/08/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023]
Abstract
Apolipoprotein E-ε4 (APOE-ε4) carriers had elevated cerebrospinal fluid (CSF) presynaptic protein growth-associated protein-43 (GAP-43), but the underlying mechanism is not fully understood. We investigated how the APOE-ε4 genotype affects the baseline and longitudinal changes in CSF GAP-43 and their associations with β-amyloid positron emission tomography (Aβ PET), CSF phosphorylated tau 181 (p-Tau181), neurodegeneration, and cognitive decline. Compared to APOE-ε4 non-carriers, APOE-ε4 carriers had higher baseline levels and faster rates of increases in Aβ PET, CSF p-Tau181, and CSF GAP-43. Both higher baseline levels and faster rates of increase in CSF GAP-43 were associated with greater baseline Aβ PET and CSF p-Tau181, which fully mediated the APOE-ε4 effect on CSF GAP-43 elevations. Independent of Aβ PET and CSF p-Tau181, APOE-ε4 carriage was associated with exacerbated GAP-43-related longitudinal hippocampal atrophy and cognitive decline, especially in Aβ+ participants (GAP-43 × time × APOE-ε4). These findings suggest that the APOE-ε4 effect on GAP-43-related presynaptic dysfunction is mediated by primary Alzheimer's pathologies and independently correlates to hippocampal atrophy and cognitive decline in the future.
Collapse
Affiliation(s)
- Guoyu Lan
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jing Du
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Xuhui Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qingyong Wang
- Department of Neurology, University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen, China
| | - Ying Han
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China; Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, China.
| |
Collapse
|
175
|
Qiang Q, Skudder-Hill L, Toyota T, Huang Z, Wei W, Adachi H. CSF 14-3-3β is associated with progressive cognitive decline in Alzheimer's disease. Brain Commun 2023; 5:fcad312. [PMID: 38035365 PMCID: PMC10684297 DOI: 10.1093/braincomms/fcad312] [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: 07/12/2023] [Revised: 09/22/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
Alzheimer's disease is a neurodegenerative disorder characterized pathologically by amyloid-beta plaques, tau tangles and neuronal loss. In clinical practice, the 14-3-3 isoform beta (β) is a biomarker that aids in the diagnosis of sporadic Creutzfeldt-Jakob disease. Recently, a proteomics study found increased CSF 14-3-3β levels in Alzheimer's disease patients, suggesting a potential link between CSF 14-3-3β and Alzheimer's disease. Our present study aimed to further investigate the role of CSF 14-3-3β in Alzheimer's disease by analysing the data of 719 participants with available CSF 14-3-3β measurements from the Alzheimer's Disease Neuroimaging Initiative. Higher CSF 14-3-3β levels were observed in the mild cognitive impairment group compared to the cognitively normal group, with the highest CSF 14-3-3β levels in the Alzheimer's disease dementia group. This study also found significant associations between CSF 14-3-3β levels and CSF biomarkers of p-tau, t-tau, pTau/Aβ42 ratios and GAP-43, as well as other Alzheimer's disease biomarkers such as Aβ-PET. An early increase in CSF 14-3-3β levels was observed prior to Aβ-PET-positive status, and CSF 14-3-3β levels continued to rise after crossing the Aβ-PET positivity threshold before reaching a plateau. The diagnostic accuracy of CSF 14-3-3β (area under the receiver operating characteristic curve = 0.819) was moderate compared to other established Alzheimer's disease biomarkers in distinguishing cognitively normal Aβ pathology-negative individuals from Alzheimer's disease Aβ pathology-positive individuals. Higher baseline CSF 14-3-3β levels were associated with accelerated cognitive decline, reduced hippocampus volumes and declining fluorodeoxyglucose-PET values over a 4-year follow-up period. Patients with mild cognitive impairment and high CSF 14-3-3β levels at baseline had a significantly increased risk [hazard ratio = 2.894 (1.599-5.238), P < 0.001] of progression to Alzheimer's disease dementia during follow-up. These findings indicate that CSF 14-3-3β may be a potential biomarker for Alzheimer's disease and could provide a more comprehensive understanding of the underlying pathological changes of Alzheimer's disease, as well as aid in the diagnosis and monitoring of disease progression.
Collapse
Affiliation(s)
- Qiang Qiang
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, 200040 Shanghai, China
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, 807-8555 Kitakyushu, Japan
| | - Loren Skudder-Hill
- Yuquan Hospital, Tsinghua University School of Clinical Medicine, 100084 Beijing, China
- School of Medicine, University of Auckland, 1023 Auckland, New Zealand
| | - Tomoko Toyota
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, 807-8555 Kitakyushu, Japan
| | - Zhe Huang
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, 807-8555 Kitakyushu, Japan
| | - Wenshi Wei
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, 200040 Shanghai, China
| | - Hiroaki Adachi
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, 807-8555 Kitakyushu, Japan
| | | |
Collapse
|
176
|
Corriveau-Lecavalier N, Botha H, Graff-Radford J, Switzer AR, Przybelski SA, Wiste HJ, Murray ME, Reichard RR, Dickson DW, Nguyen AT, Ramanan VK, McCarter SJ, Boeve BF, Machulda MM, Fields JA, Stricker NH, Nelson PT, Grothe MJ, Knopman DS, Lowe VJ, Petersen RC, Jack CR, Jones DT. A limbic-predominant amnestic neurodegenerative syndrome associated with TDP-43 pathology. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.19.23298314. [PMID: 38045300 PMCID: PMC10690340 DOI: 10.1101/2023.11.19.23298314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Limbic-predominant age-related TDP-43 encephalopathy (LATE) is a neuropathologically-defined disease that affects 40% of persons in advanced age, but its associated neurological syndrome is not defined. LATE neuropathological changes (LATE-NC) are frequently comorbid with Alzheimer's disease neuropathologic changes (ADNC). When seen in isolation, LATE-NC have been associated with a predominantly amnestic profile and slow clinical progression. We propose a set of clinical criteria for a limbic-predominant amnestic neurodegenerative syndrome (LANS) that is highly associated with LATE-NC but also other pathologic entities. The LANS criteria incorporate core, standard and advanced features that are measurable in vivo, including older age at evaluation, mild clinical syndrome, disproportionate hippocampal atrophy, impaired semantic memory, limbic hypometabolism, absence of neocortical degenerative patterns and low likelihood of neocortical tau, with degrees of certainty (highest, high, moderate, low). We operationalized this set of criteria using clinical, imaging and biomarker data to validate its associations with clinical and pathologic outcomes. We screened autopsied patients from Mayo Clinic (n = 922) and ADNI (n = 93) cohorts and applied the LANS criteria to those with an antemortem predominant amnestic syndrome (Mayo, n = 165; ADNI, n = 53). ADNC, ADNC/LATE-NC and LATE-NC accounted for 35%, 37% and 4% of cases in the Mayo cohort, respectively, and 30%, 22%, and 9% of cases in the ADNI cohort, respectively. The LANS criteria effectively categorized these cases, with ADNC having the lowest LANS likelihoods, LATE-NC patients having the highest likelihoods, and ADNC/LATE-NC patients having intermediate likelihoods. A logistic regression model using the LANS features as predictors of LATE-NC achieved a balanced accuracy of 74.6% in the Mayo cohort, and out-of-sample predictions in the ADNI cohort achieved a balanced accuracy of 73.3%. Patients with high LANS likelihoods had a milder and slower clinical course and more severe temporo-limbic degeneration compared to those with low likelihoods. Stratifying ADNC/LATE-NC patients from the Mayo cohort according to their LANS likelihood revealed that those with higher likelihoods had more temporo-limbic degeneration and a slower rate of cognitive decline, and those with lower likelihoods had more lateral temporo-parietal degeneration and a faster rate of cognitive decline. The implementation of LANS criteria has implications to disambiguate the different driving etiologies of progressive amnestic presentations in older age and guide prognosis, treatment, and clinical trials. The development of in vivo biomarkers specific to TDP-43 pathology are needed to refine molecular associations between LANS and LATE-NC and precise antemortem diagnoses of LATE.
Collapse
Affiliation(s)
- Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Heather J. Wiste
- Department of Quantitative Health Sciences, Mayo Clinic Rochester, MN, USA
| | | | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, MN, USA
| | | | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, MN, USA
| | | | | | | | - Mary M. Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Julie A. Fields
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Nikki H. Stricker
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Peter T. Nelson
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Michel J. Grothe
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center, Madrid, Spain
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Clifford R. Jack
- Department of Neuroscience, Mayo Clinic Jacksonville, FL, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - David T. Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
177
|
Kang JH, Korecka M, Lee EB, Cousins KAQ, Tropea TF, Chen-Plotkin AA, Irwin DJ, Wolk D, Brylska M, Wan Y, Shaw LM. Alzheimer Disease Biomarkers: Moving from CSF to Plasma for Reliable Detection of Amyloid and tau Pathology. Clin Chem 2023; 69:1247-1259. [PMID: 37725909 PMCID: PMC10895336 DOI: 10.1093/clinchem/hvad139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/07/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Development of validated biomarkers to detect early Alzheimer disease (AD) neuropathology is needed for therapeutic AD trials. Abnormal concentrations of "core" AD biomarkers, cerebrospinal fluid (CSF) amyloid beta1-42, total tau, and phosphorylated tau correlate well with neuroimaging biomarkers and autopsy findings. Nevertheless, given the limitations of established CSF and neuroimaging biomarkers, accelerated development of blood-based AD biomarkers is underway. CONTENT Here we describe the clinical significance of CSF and plasma AD biomarkers to detect disease pathology throughout the Alzheimer continuum and correlate with imaging biomarkers. Use of the AT(N) classification by CSF and imaging biomarkers provides a more objective biologically based diagnosis of AD than clinical diagnosis alone. Significant progress in measuring CSF AD biomarkers using extensively validated highly automated assay systems has facilitated their transition from research use only to approved in vitro diagnostics tests for clinical use. We summarize development of plasma AD biomarkers as screening tools for enrollment and monitoring participants in therapeutic trials and ultimately in clinical care. Finally, we discuss the challenges for AD biomarkers use in clinical trials and precision medicine, emphasizing the possible ethnocultural differences in the levels of AD biomarkers. SUMMARY CSF AD biomarker measurements using fully automated analytical platforms is possible. Building on this experience, validated blood-based biomarker tests are being implemented on highly automated immunoassay and mass spectrometry platforms. The progress made developing analytically and clinically validated plasma AD biomarkers within the AT(N) classification scheme can accelerate use of AD biomarkers in therapeutic trials and routine clinical practice.
Collapse
Affiliation(s)
- Ju Hee Kang
- Department of Pharmacology and Clinical Pharmacology, Research Center for Controlling Intercellular Communication, Inha University, Incheon, South Korea
| | - Magdalena Korecka
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Katheryn A Q Cousins
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Thomas F Tropea
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alice A Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Magdalena Brylska
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yang Wan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
178
|
Balajoo SM, Eickhoff SB, Masouleh SK, Plachti A, Waite L, Saberi A, Bahri MA, Bastin C, Salmon E, Hoffstaedter F, Palomero-Gallagher N, Genon S. Hippocampal metabolic subregions and networks: Behavioral, molecular, and pathological aging profiles. Alzheimers Dement 2023; 19:4787-4804. [PMID: 37014937 PMCID: PMC10698199 DOI: 10.1002/alz.13056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 04/06/2023]
Abstract
INTRODUCTION Hippocampal local and network dysfunction is the hallmark of Alzheimer's disease (AD). METHODS We characterized the spatial patterns of hippocampus differentiation based on brain co-metabolism in healthy elderly participants and demonstrated their relevance to study local metabolic changes and associated dysfunction in pathological aging. RESULTS The hippocampus can be differentiated into anterior/posterior and dorsal cornu ammonis (CA)/ventral (subiculum) subregions. While anterior/posterior CA show co-metabolism with different regions of the subcortical limbic networks, the anterior/posterior subiculum are parts of cortical networks supporting object-centered memory and higher cognitive demands, respectively. Both networks show relationships with the spatial patterns of gene expression pertaining to cell energy metabolism and AD's process. Finally, while local metabolism is generally lower in posterior regions, the anterior-posterior imbalance is maximal in late mild cognitive impairment with the anterior subiculum being relatively preserved. DISCUSSION Future studies should consider bidimensional hippocampal differentiation and in particular the posterior subicular region to better understand pathological aging.
Collapse
Affiliation(s)
- Somayeh Maleki Balajoo
- Institute of Systems Neuroscience, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
- Institute of Neuroscience and Medicine (INM-7), Research Centre Juelich, Juelich, Germany
| | - Simon B. Eickhoff
- Institute of Systems Neuroscience, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
- Institute of Neuroscience and Medicine (INM-7), Research Centre Juelich, Juelich, Germany
| | - Shahrzad Kharabian Masouleh
- Institute of Systems Neuroscience, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
- Institute of Neuroscience and Medicine (INM-7), Research Centre Juelich, Juelich, Germany
| | - Anna Plachti
- Institute of Systems Neuroscience, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
- Institute of Neuroscience and Medicine (INM-7), Research Centre Juelich, Juelich, Germany
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Laura Waite
- Institute of Neuroscience and Medicine (INM-7), Research Centre Juelich, Juelich, Germany
| | - Amin Saberi
- Institute of Systems Neuroscience, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
- Institute of Neuroscience and Medicine (INM-7), Research Centre Juelich, Juelich, Germany
- Otto Hahn Research Group for Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Mohamed Ali Bahri
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
| | - Christine Bastin
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
| | - Eric Salmon
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
- Department of Neurology, University Hospital of Liège, Liège, Belgium
| | - Felix Hoffstaedter
- Institute of Neuroscience and Medicine (INM-7), Research Centre Juelich, Juelich, Germany
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine (INM‑1), Research Centre Juelich, Juelich, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Cécile and Oskar Vogt Institute for Brain Research, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Sarah Genon
- Institute of Systems Neuroscience, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
- Institute of Neuroscience and Medicine (INM-7), Research Centre Juelich, Juelich, Germany
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
| |
Collapse
|
179
|
Blanco K, Salcidua S, Orellana P, Sauma-Pérez T, León T, Steinmetz LCL, Ibañez A, Duran-Aniotz C, de la Cruz R. Systematic review: fluid biomarkers and machine learning methods to improve the diagnosis from mild cognitive impairment to Alzheimer's disease. Alzheimers Res Ther 2023; 15:176. [PMID: 37838690 PMCID: PMC10576366 DOI: 10.1186/s13195-023-01304-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/15/2023] [Indexed: 10/16/2023]
Abstract
Mild cognitive impairment (MCI) is often considered an early stage of dementia, with estimated rates of progression to dementia up to 80-90% after approximately 6 years from the initial diagnosis. Diagnosis of cognitive impairment in dementia is typically based on clinical evaluation, neuropsychological assessments, cerebrospinal fluid (CSF) biomarkers, and neuroimaging. The main goal of diagnosing MCI is to determine its cause, particularly whether it is due to Alzheimer's disease (AD). However, only a limited percentage of the population has access to etiological confirmation, which has led to the emergence of peripheral fluid biomarkers as a diagnostic tool for dementias, including MCI due to AD. Recent advances in biofluid assays have enabled the use of sophisticated statistical models and multimodal machine learning (ML) algorithms for the diagnosis of MCI based on fluid biomarkers from CSF, peripheral blood, and saliva, among others. This approach has shown promise for identifying specific causes of MCI, including AD. After a PRISMA analysis, 29 articles revealed a trend towards using multimodal algorithms that incorporate additional biomarkers such as neuroimaging, neuropsychological tests, and genetic information. Particularly, neuroimaging is commonly used in conjunction with fluid biomarkers for both cross-sectional and longitudinal studies. Our systematic review suggests that cost-effective longitudinal multimodal monitoring data, representative of diverse cultural populations and utilizing white-box ML algorithms, could be a valuable contribution to the development of diagnostic models for AD due to MCI. Clinical assessment and biomarkers, together with ML techniques, could prove pivotal in improving diagnostic tools for MCI due to AD.
Collapse
Affiliation(s)
- Kevin Blanco
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Diagonal Las Torres 2640, Peñalolén, Santiago, Chile
| | - Stefanny Salcidua
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
- Faculty of Engineering and Sciences, Universidad Adolfo Ibáñez, Diagonal Las Torres 2700, Building D, Peñalolén, Santiago, Chile
| | - Paulina Orellana
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Diagonal Las Torres 2640, Peñalolén, Santiago, Chile
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Tania Sauma-Pérez
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Tomás León
- Global Brain Health Institute, Trinity College, Dublin, Ireland
- Memory and Neuropsychiatric Center (CMYN) Neurology Department, Hospital del Salvador and Faculty of Medicine, University of Chile, Santiago, Chile
| | - Lorena Cecilia López Steinmetz
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
- Technische Universität Berlin, Berlin, Deutschland
- Instituto de Investigaciones Psicológicas (IIPsi), Universidad Nacional de Córdoba (UNC) y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Agustín Ibañez
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
- Global Brain Health Institute, Trinity College, Dublin, Ireland
- Global Brain Health Institute, University of California San Francisco (UCSF), San Francisco, CA, USA
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, & National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Claudia Duran-Aniotz
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Diagonal Las Torres 2640, Peñalolén, Santiago, Chile.
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile.
| | - Rolando de la Cruz
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile.
- Faculty of Engineering and Sciences, Universidad Adolfo Ibáñez, Diagonal Las Torres 2700, Building D, Peñalolén, Santiago, Chile.
- Data Observatory Foundation, ANID Technology Center No. DO210001, Santiago, Chile.
| |
Collapse
|
180
|
Dubois B, von Arnim CAF, Burnie N, Bozeat S, Cummings J. Biomarkers in Alzheimer's disease: role in early and differential diagnosis and recognition of atypical variants. Alzheimers Res Ther 2023; 15:175. [PMID: 37833762 PMCID: PMC10571241 DOI: 10.1186/s13195-023-01314-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND Development of in vivo biomarkers has shifted the diagnosis of Alzheimer's disease (AD) from the later dementia stages of disease towards the earlier stages and has introduced the potential for pre-symptomatic diagnosis. The International Working Group recommends that AD diagnosis is restricted in the clinical setting to people with specific AD phenotypes and supportive biomarker findings. MAIN BODY In this review, we discuss the phenotypic presentation and use of biomarkers for the early diagnosis of typical and atypical AD and describe how this can support clinical decision making, benefit patient communication, and improve the patient journey. Early diagnosis is essential to optimize the benefits of available and emerging treatments. As atypical presentations of AD often mimic other dementias, differential diagnosis can be challenging and can be facilitated using AD biomarkers. However, AD biomarkers alone are not sufficient to confidently diagnose AD or predict disease progression and should be supplementary to clinical assessment to help inform the diagnosis of AD. CONCLUSIONS Use of AD biomarkers with incorporation of atypical AD phenotypes into diagnostic criteria will allow earlier diagnosis of patients with atypical clinical presentations that otherwise would have been misdiagnosed and treated inappropriately. Early diagnosis is essential to guide informed discussion, appropriate care and support, and individualized treatment. It is hoped that disease-modifying treatments will impact the underlying AD pathology; thus, determining the patient's AD phenotype will be a critical factor in guiding the therapeutic approach and the assessment of the effects of interventions.
Collapse
Affiliation(s)
- Bruno Dubois
- Assistance Publique-Hôpitaux de Paris (AP-HP), Memory and Alzheimer's Disease Institute, Sorbonne University, Paris, France
- Brain Institute, Sorbonne University, Paris, France
| | | | - Nerida Burnie
- General Practice, South West London CCG, London, UK
- London Dementia Clinical Network, London, UK
| | | | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| |
Collapse
|
181
|
Mieling M, Göttlich M, Yousuf M, Bunzeck N. Basal forebrain activity predicts functional degeneration in the entorhinal cortex in Alzheimer's disease. Brain Commun 2023; 5:fcad262. [PMID: 37901036 PMCID: PMC10608112 DOI: 10.1093/braincomms/fcad262] [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: 04/17/2023] [Revised: 08/23/2023] [Accepted: 10/07/2023] [Indexed: 10/31/2023] Open
Abstract
Recent models of Alzheimer's disease suggest the nucleus basalis of Meynert (NbM) as an early origin of structural degeneration followed by the entorhinal cortex (EC). However, the functional properties of NbM and EC regarding amyloid-β and hyperphosphorylated tau remain unclear. We analysed resting-state functional fMRI data with CSF assays from the Alzheimer's Disease Neuroimaging Initiative (n = 71) at baseline and 2 years later. At baseline, local activity, as quantified by fractional amplitude of low-frequency fluctuations, differentiated between normal and abnormal CSF groups in the NbM but not EC. Further, NbM activity linearly decreased as a function of CSF ratio, resembling the disease status. Finally, NbM activity predicted the annual percentage signal change in EC, but not the reverse, independent from CSF ratio. Our findings give novel insights into the pathogenesis of Alzheimer's disease by showing that local activity in NbM is affected by proteinopathology and predicts functional degeneration within the EC.
Collapse
Affiliation(s)
- Marthe Mieling
- Department of Psychology, University of Lübeck, Lübeck 23562, Germany
| | - Martin Göttlich
- Department of Neurology, University of Lübeck, Lübeck 23562, Germany
- Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck 23562, Germany
| | - Mushfa Yousuf
- Department of Psychology, University of Lübeck, Lübeck 23562, Germany
| | - Nico Bunzeck
- Department of Psychology, University of Lübeck, Lübeck 23562, Germany
- Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck 23562, Germany
| |
Collapse
|
182
|
Krell-Roesch J, Rakusa M, Syrjanen JA, van Harten AC, Lowe VJ, Jack CR, Kremers WK, Knopman DS, Stokin GB, Petersen RC, Vassilaki M, Geda YE. Association between CSF biomarkers of Alzheimer's disease and neuropsychiatric symptoms: Mayo Clinic Study of Aging. Alzheimers Dement 2023; 19:4498-4506. [PMID: 35142047 PMCID: PMC10433790 DOI: 10.1002/alz.12557] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 06/17/2021] [Accepted: 11/02/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION We examined the association between cerebrospinal fluid (CSF)-derived biomarkers of Alzheimer's disease and neuropsychiatric symptoms (NPS) in older non-demented adults. METHODS We included 784 persons (699 cognitively unimpaired, 85 with mild cognitive impairment) aged ≥ 50 years who underwent CSF amyloid beta (Aβ42), hyperphosphorylated tau 181 (p-tau), and total tau (t-tau) as well as NPS assessment using Beck Depression and Anxiety Inventories (BDI-II, BAI), and Neuropsychiatric Inventory Questionnaire (NPI-Q). RESULTS Lower CSF Aβ42, and higher t-tau/Aβ42 and p-tau/Aβ42 ratios were associated with BDI-II and BAI total scores, clinical depression (BDI-II ≥ 13), and clinical anxiety (BAI ≥ 10), as well as NPI-Q-assessed anxiety, apathy, and nighttime behavior. DISCUSSION CSF Aβ42, t-tau/Aβ42, and p-tau/Aβ42 ratios were associated with NPS in community-dwelling individuals free of dementia. If confirmed by a longitudinal cohort study, the findings have clinical relevance of taking into account the NPS status of individuals with abnormal CSF biomarkers.
Collapse
Affiliation(s)
- Janina Krell-Roesch
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
- Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Martin Rakusa
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, University Medical Centre Maribor, Maribor, Slovenia
| | - Jeremy A. Syrjanen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Argonde C. van Harten
- Alzheimer Center, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Walter K. Kremers
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Gorazd B. Stokin
- International Clinical Research Center, St. Anne’s Hospital, Brno, Czech Republic
| | - Ronald C. Petersen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Maria Vassilaki
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Yonas E. Geda
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
183
|
Bhujbal SS, Kad MM, Patole VC. Recent diagnostic techniques for the detection of Alzheimer's disease: a short review. Ir J Med Sci 2023; 192:2417-2426. [PMID: 36525239 DOI: 10.1007/s11845-022-03244-y] [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: 07/28/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is a neurological condition that affects millions of individuals around the world and for which there are few effective therapies. Dementia is characterized by the formation of senile plaques and neurofibrillary tangles, which is followed by neurotoxicity, which results in memory loss and mortality. Pathogenesis occurs several years before the onset of disease. As the disease-modifying drugs are most effective in the early stages of Alzheimer's disease, biomarkers for early detection of disease and their development are crucial. This review discusses the diagnostic utility, benefits, and limitations of traditional techniques such as neuroimaging, cognitive testing, positron emission tomography, and biomarkers, as well as the novel techniques such as artificial intelligence, machine learning, immunotherapy, and blood test approaches for early detection, understanding, and treatment of AD.
Collapse
Affiliation(s)
- Santosh S Bhujbal
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences & Research, Pimpri, Pune, India.
| | - Minal M Kad
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences & Research, Pimpri, Pune, India
| | - Vinita C Patole
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences & Research, Pimpri, Pune, India
| |
Collapse
|
184
|
Tao Q, Zhang C, Mercier G, Lunetta K, Ang TFA, Akhter‐Khan S, Zhang Z, Taylor A, Killiany RJ, Alosco M, Mez J, Au R, Zhang X, Farrer LA, Qiu WWQ. Identification of an APOE ε4-specific blood-based molecular pathway for Alzheimer's disease risk. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12490. [PMID: 37854772 PMCID: PMC10579631 DOI: 10.1002/dad2.12490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023]
Abstract
INTRODUCTION The precise apolipoprotein E (APOE) ε4-specific molecular pathway(s) for Alzheimer's disease (AD) risk are unclear. METHODS Plasma protein modules/cascades were analyzed using weighted gene co-expression network analysis (WGCNA) in the Alzheimer's Disease Neuroimaging Initiative study. Multivariable regression analyses were used to examine the associations among protein modules, AD diagnoses, cerebrospinal fluid (CSF) phosphorylated tau (p-tau), and brain glucose metabolism, stratified by APOE genotype. RESULTS The Green Module was associated with AD diagnosis in APOE ε4 homozygotes. Three proteins from this module, C-reactive protein (CRP), complement C3, and complement factor H (CFH), had dose-dependent associations with CSF p-tau and cognitive impairment only in APOE ε4 homozygotes. The link among these three proteins and glucose hypometabolism was observed in brain regions of the default mode network (DMN) in APOE ε4 homozygotes. A Framingham Heart Study validation study supported the findings for AD. DISCUSSION The study identifies the APOE ε4-specific CRP-C3-CFH inflammation pathway for AD, suggesting potential drug targets for the disease.Highlights: Identification of an APOE ε4 specific molecular pathway involving blood CRP, C3, and CFH for the risk of AD.CRP, C3, and CFH had dose-dependent associations with CSF p-Tau and brain glucose hypometabolism as well as with cognitive impairment only in APOE ε4 homozygotes.Targeting CRP, C3, and CFH may be protective and therapeutic for AD onset in APOE ε4 carriers.
Collapse
Affiliation(s)
- Qiushan Tao
- Department of Pharmacology, Physiology & BiophysicsBoston University School of MedicineBostonMassachusettsUSA
- Slone Epidemiology CenterSchool of Public HealthBoston University Medical Campus (BUMC)BostonMassachusettsUSA
| | - Chao Zhang
- Section of Computational BiomedicineDepartment of MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Gustavo Mercier
- Section of Molecular Imaging and Nuclear MedicineDepartment of RadiologyBoston University School of MedicineBostonMassachusettsUSA
| | - Kathryn Lunetta
- Slone Epidemiology CenterSchool of Public HealthBoston University Medical Campus (BUMC)BostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Ting Fang Alvin Ang
- Slone Epidemiology CenterSchool of Public HealthBoston University Medical Campus (BUMC)BostonMassachusettsUSA
- Department of Anatomy & NeurobiologyBoston University School of MedicineBostonMassachusettsUSA
| | - Samia Akhter‐Khan
- Department of Health Service & Population ResearchKing's College London, LondonDavid Goldberg CentreLondonUK
| | - Zhengrong Zhang
- Department of Pharmacology, Physiology & BiophysicsBoston University School of MedicineBostonMassachusettsUSA
| | - Andrew Taylor
- Department of OphthalmologyBoston University School of MedicineBostonMassachusettsUSA
| | - Ronald J. Killiany
- Department of Anatomy & NeurobiologyBoston University School of MedicineBostonMassachusettsUSA
| | - Michael Alosco
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
| | - Jesse Mez
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
- Alzheimer's Disease and CTE CentersBoston University School of MedicineBostonMassachusettsUSA
| | - Rhoda Au
- Slone Epidemiology CenterSchool of Public HealthBoston University Medical Campus (BUMC)BostonMassachusettsUSA
- Department of Anatomy & NeurobiologyBoston University School of MedicineBostonMassachusettsUSA
| | - Xiaoling Zhang
- Department of MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Lindsay A. Farrer
- Alzheimer's Disease and CTE CentersBoston University School of MedicineBostonMassachusettsUSA
- Department of MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Wendy Wei Qiao Qiu
- Department of Pharmacology, Physiology & BiophysicsBoston University School of MedicineBostonMassachusettsUSA
- Alzheimer's Disease and CTE CentersBoston University School of MedicineBostonMassachusettsUSA
- Department of PsychiatryBoston University School of MedicineBostonMassachusettsUSA
| | | |
Collapse
|
185
|
Bilgel M, An Y, Walker KA, Moghekar AR, Ashton NJ, Kac PR, Karikari TK, Blennow K, Zetterberg H, Jedynak BM, Thambisetty M, Ferrucci L, Resnick SM. Longitudinal changes in Alzheimer's-related plasma biomarkers and brain amyloid. Alzheimers Dement 2023; 19:4335-4345. [PMID: 37216632 PMCID: PMC10592628 DOI: 10.1002/alz.13157] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023]
Abstract
INTRODUCTION Understanding longitudinal plasma biomarker trajectories relative to brain amyloid changes can help devise Alzheimer's progression assessment strategies. METHODS We examined the temporal order of changes in plasma amyloid-β ratio (A β 42 / A β 40 ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}$ ), glial fibrillary acidic protein (GFAP), neurofilament light chain (NfL), and phosphorylated tau ratios (p-tau181 / A β 42 $\text{p-tau181}/\mathrm{A}{\beta}_{42}$ ,p-tau231 / A β 42 $\text{p-tau231}/\mathrm{A}{\beta}_{42}$ ) relative to 11 C-Pittsburgh compound B (PiB) positron emission tomography (PET) cortical amyloid burden (PiB-/+). Participants (n = 199) were cognitively normal at index visit with a median 6.1-year follow-up. RESULTS PiB groups exhibited different rates of longitudinal change inA β 42 / A β 40 ( β = 5.41 × 10 - 4 , SE = 1.95 × 10 - 4 , p = 0.0073 ) ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}\ ( {\beta \ = \ 5.41 \times {{10}}^{ - 4},{\rm{\ SE\ }} = \ 1.95 \times {{10}}^{ - 4},\ p\ = \ 0.0073} )$ . Change in brain amyloid correlated with change in GFAP (r = 0.5, 95% CI = [0.26, 0.68]). The greatest relative decline inA β 42 / A β 40 ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}$ (-1%/year) preceded brain amyloid positivity by 41 years (95% CI = [32, 53]). DISCUSSION PlasmaA β 42 / A β 40 ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}$ may begin declining decades prior to brain amyloid accumulation, whereas p-tau ratios, GFAP, and NfL increase closer in time. HIGHLIGHTS PlasmaA β 42 / A β 40 ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}$ declines over time among PiB- but does not change among PiB+. Phosphorylated-tau to Aβ42 ratios increase over time among PiB+ but do not change among PiB-. Rate of change in brain amyloid is correlated with change in GFAP and neurofilament light chain. The greatest decline inA β 42 / A β 40 ${{\rm A}\beta }_{42}/{{\rm A}\beta }_{40}$ may precede brain amyloid positivity by decades.
Collapse
Affiliation(s)
- Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Abhay R. Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RX, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research, Unit for Dementia at South London and Maudsley, NHS Foundation, London, SE5 8AF, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, 4019 Stavanger, Norway
| | - Przemysław R. Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
| | - Thomas K. Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Bruno M. Jedynak
- Department of Mathematics and Statistics, Portland State University, Portland, Oregon, 97201, USA
| | - Madhav Thambisetty
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, 21224, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, 21224, USA
| |
Collapse
|
186
|
Caprihan A, Hillmer L, Erhardt EB, Adair JC, Knoefel JE, Prestopnik J, Rosenberg GA. A trichotomy method for defining homogeneous subgroups in a dementia population. Ann Clin Transl Neurol 2023; 10:1802-1815. [PMID: 37602520 PMCID: PMC10578887 DOI: 10.1002/acn3.51869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/11/2023] [Accepted: 07/22/2023] [Indexed: 08/22/2023] Open
Abstract
INTRODUCTION Diagnosis of dementia in the aging brain is confounded by the presence of multiple pathologies. Mixed dementia (MX), a combination of Alzheimer's disease (AD) proteins with vascular disease (VD), is frequently found at autopsy, and has been difficult to diagnose during life. This report develops a method for separating the MX group and defining preclinical AD (presence of AD factors with normal cognition) and preclinical VD subgroups (presence of white matter damage with normal cognition). METHODS Clustering was based on three diagnostic axes: (1) AD factor (ADF) derived from cerebrospinal fluid proteins (Aβ42 and pTau), (2) VD factor (VDF) calculated from mean free water and peak width of skeletonized mean diffusivity in the white matter, and (3) Cognition (Cog) based on memory and executive function. The trichotomy method was applied to an Alzheimer's Disease Neuroimaging Initiative cohort (N = 538). RESULTS Eight biologically defined subgroups were identified which included the MX group with both high ADF and VDF (9.3%) and a preclinical VD group (3.9%), and a preclinical AD group (13.6%). Cog is significantly associated with both ADF and VDF, and the partial-correlation remains significant even when the effect of the other variable is removed (r(Cog, ADF/VDF removed) = 0.46, p < 10-28 and r(Cog, VDF/ADF removed) = 0.24, p < 10-7 ). DISCUSSION The trichotomy method creates eight biologically characterized patient groups, which includes MX, preclinical AD, and preclinical VD subgroups. Further longitudinal studies are needed to determine the utility of the 3-way clustering method with multimodal biological biomarkers.
Collapse
Affiliation(s)
| | - Laura Hillmer
- Center for Memory and AgingUniversity of New Mexico School of MedicineAlbuquerqueNew Mexico87106USA
| | - Erik Barry Erhardt
- Departments of Mathematics and StatisticsUniversity of New Mexico College of Arts and SciencesAlbuquerqueNew Mexico87106USA
| | - John C. Adair
- Center for Memory and AgingUniversity of New Mexico School of MedicineAlbuquerqueNew Mexico87106USA
- Department of NeurologyUniversity of New MexicoAlbuquerqueNew Mexico87106USA
| | - Janice E. Knoefel
- Center for Memory and AgingUniversity of New Mexico School of MedicineAlbuquerqueNew Mexico87106USA
- Department of NeurologyUniversity of New MexicoAlbuquerqueNew Mexico87106USA
| | - Jillian Prestopnik
- Center for Memory and AgingUniversity of New Mexico School of MedicineAlbuquerqueNew Mexico87106USA
| | - Gary A. Rosenberg
- Center for Memory and AgingUniversity of New Mexico School of MedicineAlbuquerqueNew Mexico87106USA
- Department of NeurologyUniversity of New MexicoAlbuquerqueNew Mexico87106USA
| | | |
Collapse
|
187
|
Waschkies KF, Soch J, Darna M, Richter A, Altenstein S, Beyle A, Brosseron F, Buchholz F, Butryn M, Dobisch L, Ewers M, Fliessbach K, Gabelin T, Glanz W, Goerss D, Gref D, Janowitz D, Kilimann I, Lohse A, Munk MH, Rauchmann BS, Rostamzadeh A, Roy N, Spruth EJ, Dechent P, Heneka MT, Hetzer S, Ramirez A, Scheffler K, Buerger K, Laske C, Perneczky R, Peters O, Priller J, Schneider A, Spottke A, Teipel S, Düzel E, Jessen F, Wiltfang J, Schott BH, Kizilirmak JM. Machine learning-based classification of Alzheimer's disease and its at-risk states using personality traits, anxiety, and depression. Int J Geriatr Psychiatry 2023; 38:e6007. [PMID: 37800601 DOI: 10.1002/gps.6007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 09/07/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is often preceded by stages of cognitive impairment, namely subjective cognitive decline (SCD) and mild cognitive impairment (MCI). While cerebrospinal fluid (CSF) biomarkers are established predictors of AD, other non-invasive candidate predictors include personality traits, anxiety, and depression, among others. These predictors offer non-invasive assessment and exhibit changes during AD development and preclinical stages. METHODS In a cross-sectional design, we comparatively evaluated the predictive value of personality traits (Big Five), geriatric anxiety and depression scores, resting-state functional magnetic resonance imaging activity of the default mode network, apoliprotein E (ApoE) genotype, and CSF biomarkers (tTau, pTau181, Aβ42/40 ratio) in a multi-class support vector machine classification. Participants included 189 healthy controls (HC), 338 individuals with SCD, 132 with amnestic MCI, and 74 with mild AD from the multicenter DZNE-Longitudinal Cognitive Impairment and Dementia Study (DELCODE). RESULTS Mean predictive accuracy across all participant groups was highest when utilizing a combination of personality, depression, and anxiety scores. HC were best predicted by a feature set comprised of depression and anxiety scores and participants with AD were best predicted by a feature set containing CSF biomarkers. Classification of participants with SCD or aMCI was near chance level for all assessed feature sets. CONCLUSION Our results demonstrate predictive value of personality trait and state scores for AD. Importantly, CSF biomarkers, personality, depression, anxiety, and ApoE genotype show complementary value for classification of AD and its at-risk stages.
Collapse
Affiliation(s)
- Konrad F Waschkies
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Joram Soch
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Margarita Darna
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Anni Richter
- Leibniz Institute for Neurobiology, Magdeburg, Germany
- German Center for Mental Health (DZPG), Munich, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Aline Beyle
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University of Bonn, Bonn, Germany
| | | | - Friederike Buchholz
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Michaela Butryn
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
| | - Tatjana Gabelin
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Doreen Goerss
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Daria Gref
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Andrea Lohse
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Matthias H Munk
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
- Department of Neuroradiology, University Hospital LMU, Munich, Germany
| | - Ayda Rostamzadeh
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Peter Dechent
- MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University Goettingen, Göttingen, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Stefan Hetzer
- Berlin Center for Advanced Neuroimaging, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, Texas, USA
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
- School of Medicine, Technical University of Munich, Department of Psychiatry and Psychotherapy, Munich, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Björn H Schott
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Jasmin M Kizilirmak
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Neurodidactics and NeuroLab, Institute for Psychology, University of Hildesheim, Hildesheim, Germany
| |
Collapse
|
188
|
Romano MF, Zhou X, Balachandra AR, Jadick MF, Qiu S, Nijhawan DA, Joshi PS, Mohammad S, Lee PH, Smith MJ, Paul AB, Mian AZ, Small JE, Chin SP, Au R, Kolachalama VB. Deep learning for risk-based stratification of cognitively impaired individuals. iScience 2023; 26:107522. [PMID: 37646016 PMCID: PMC10460987 DOI: 10.1016/j.isci.2023.107522] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/19/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023] Open
Abstract
Quantifying the risk of progression to Alzheimer's disease (AD) could help identify persons who could benefit from early interventions. We used data from the Alzheimer's Disease Neuroimaging Initiative (ADNI, n = 544, discovery cohort) and the National Alzheimer's Coordinating Center (NACC, n = 508, validation cohort), subdividing individuals with mild cognitive impairment (MCI) into risk groups based on cerebrospinal fluid amyloid-β levels and identifying differential gray matter patterns. We then created models that fused neural networks with survival analysis, trained using non-parcellated T1-weighted brain MRIs from ADNI data, to predict the trajectories of MCI to AD conversion within the NACC cohort (integrated Brier score: 0.192 [discovery], and 0.108 [validation]). Using modern interpretability techniques, we verified that regions important for model prediction are classically associated with AD. We confirmed AD diagnosis labels using postmortem data. We conclude that our framework provides a strategy for risk-based stratification of individuals with MCI and for identifying regions key for disease prognosis.
Collapse
Affiliation(s)
- Michael F. Romano
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Xiao Zhou
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Computer Science, Boston University, Boston, MA, USA
| | - Akshara R. Balachandra
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michalina F. Jadick
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Shangran Qiu
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Diya A. Nijhawan
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Prajakta S. Joshi
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of General Dentistry, Boston University School of Dental Medicine, Boston, MA, USA
- The Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Shariq Mohammad
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Peter H. Lee
- Department of Radiology, Lahey Hospital & Medical Center, Burlington, MA, USA
| | - Maximilian J. Smith
- Department of Radiology, Lahey Hospital & Medical Center, Burlington, MA, USA
| | - Aaron B. Paul
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Asim Z. Mian
- Department of Radiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Juan E. Small
- Department of Radiology, Lahey Hospital & Medical Center, Burlington, MA, USA
| | - Sang P. Chin
- Department of Computer Science, Boston University, Boston, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center of Mathematical Sciences & Applications, Harvard University, Cambridge, MA, USA
| | - Rhoda Au
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Boston University Alzheimer’s Disease Research Center, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Vijaya B. Kolachalama
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Computer Science, Boston University, Boston, MA, USA
- Boston University Alzheimer’s Disease Research Center, Boston, MA, USA
- Faculty of Computing & Data Sciences, Boston University, Boston, MA, USA
| |
Collapse
|
189
|
Perovnik M, Tang CC, Namías M, Eidelberg D. Longitudinal changes in metabolic network activity in early Alzheimer's disease. Alzheimers Dement 2023; 19:4061-4072. [PMID: 37204815 DOI: 10.1002/alz.13137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/20/2023]
Abstract
INTRODUCTION The progression of Alzheimer's disease (AD) has been linked to two metabolic networks, the AD-related pattern (ADRP) and the default mode network (DMN). METHODS Converting and clinically stable cognitively normal subjects (n = 47) and individuals with mild cognitive impairment (n = 96) underwent 2-[18 F]fluoro-2-deoxy-D-glucose (FDG) positron emission tomography (PET) three or more times over 6 years (nscans = 705). Expression levels for ADRP and DMN were measured in each subject and time point, and the resulting changes were correlated with cognitive performance. The role of network expression in predicting conversion to dementia was also evaluated. RESULTS Longitudinal increases in ADRP expression were observed in converters, while age-related DMN loss was seen in converters and nonconverters. Cognitive decline correlated with increases in ADRP and declines in DMN, but conversion to dementia was predicted only by baseline ADRP levels. DISCUSSION The results point to the potential utility of ADRP as an imaging biomarker of AD progression.
Collapse
Affiliation(s)
- Matej Perovnik
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Chris C Tang
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Mauro Namías
- Fundación Centro Diagnóstico Nuclear, Buenos Aires, Argentina
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Molecular Medicine and Neurology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
190
|
Sun HL, Yao XQ, Lei L, Jin WS, Bai YD, Zeng GH, Shi AY, Liang J, Zhu L, Liu YH, Wang YJ, Bu XL. Associations of Blood and Cerebrospinal Fluid Aβ and tau Levels with Renal Function. Mol Neurobiol 2023; 60:5343-5351. [PMID: 37310581 DOI: 10.1007/s12035-023-03420-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/25/2023] [Indexed: 06/14/2023]
Abstract
Amyloid β (Aβ) and tau play pivotal roles in the pathogenesis of Alzheimer's disease (AD). Previous studies have shown that brain-derived Aβ and tau can be cleared through transport into the periphery, and the kidneys may be vital organs involved in the clearance of Aβ and tau. However, the effects of deficiency in the clearance of Aβ and tau by the kidneys on brain AD-type pathologies in humans remain largely unknown. In this study, we first recruited 41 patients with chronic kidney disease (CKD) and 40 age- and sex-matched controls with normal renal function to analyze the associations of the estimated glomerular filtration rate (eGFR) with plasma Aβ and tau levels. To analyze the associations of eGFR with cerebrospinal fluid (CSF) AD biomarkers, we recruited 42 cognitively normal CKD patients and 150 cognitively normal controls with CSF samples. Compared with controls with normal renal function, CKD patients had higher plasma levels of Aβ40, Aβ42 and total tau (T-tau), lower CSF levels of Aβ40 and Aβ42 and higher levels of CSF T-tau/Aβ42 and phosphorylated tau (P-tau)/Aβ42. Plasma Aβ40, Aβ42, and T-tau levels were negatively correlated with eGFR. In addition, eGFR was negatively correlated with CSF levels of T-tau, T-tau/Aβ42, and P-tau/Aβ42 but positively correlated with Mini-Mental State Examination (MMSE) scores. Thus, this study showed that the decline in renal function was correlated with abnormal AD biomarkers and cognitive decline, which provides human evidence that renal function may be involved in the pathogenesis of AD.
Collapse
Affiliation(s)
- Hao-Lun Sun
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, China
| | - Xiu-Qing Yao
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Lei
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Wang-Sheng Jin
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Yu-Di Bai
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Gui-Hua Zeng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - An-Yu Shi
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Jun Liang
- Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, China
| | - Li Zhu
- Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, China
| | - Yu-Hui Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Xian-Le Bu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China.
| |
Collapse
|
191
|
Ta M, Blauwendraat C, Antar T, Leonard HL, Singleton AB, Nalls MA, Iwaki H. Genome-Wide Meta-Analysis of Cerebrospinal Fluid Biomarkers in Alzheimer's Disease and Parkinson's Disease Cohorts. Mov Disord 2023; 38:1697-1705. [PMID: 37539664 PMCID: PMC11459375 DOI: 10.1002/mds.29511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/14/2023] [Accepted: 05/30/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Amyloid-β, phosphorylated tau (p-tau), and total tau (t-tau) in cerebrospinal fluid are established biomarkers for Alzheimer's disease (AD). In other neurodegenerative diseases, such as Parkinson's disease (PD), these biomarkers have also been found to be altered, and the molecular mechanisms responsible for these alterations are still under investigation. Moreover, the interplay between these mechanisms and the diverse underlying disease states remains to be elucidated. OBJECTIVE To investigate genetic contributions to the AD biomarkers and assess the commonality and heterogeneity of the associations per underlying disease status. METHODS We conducted genome-wide association studies (GWASs) for the AD biomarkers on subjects from the Parkinson's Progression Markers Initiative, the Fox Investigation for New Discovery of Biomarkers, and the Alzheimer's Disease Neuroimaging Initiative, and meta-analyzed with the largest AD GWAS. We tested heterogeneity of associations of interest between different disease statuses (AD, PD, and control). RESULTS We observed three GWAS signals: the APOE locus for amyloid-β, the 3q28 locus between GEMC1 and OSTN for p-tau and t-tau, and the 7p22 locus (top hit: rs60871478, an intronic variant for DNAAF5, also known as HEATR2) for p-tau. The 7p22 locus is novel and colocalized with the brain DNAAF5 expression. Although no heterogeneity from underlying disease status was observed for the earlier GWAS signals, some disease risk loci suggested disease-specific associations with these biomarkers. CONCLUSIONS Our study identified a novel association at the intronic region of DNAAF5 associated with increased levels of p-tau across all diseases. We also observed some disease-specific genetic associations with these biomarkers. Published 2023. This article is a U.S. Government work and is in the public domain in the USA.
Collapse
Affiliation(s)
- Michael Ta
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Tarek Antar
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Hampton L Leonard
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
| | - Andrew B. Singleton
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD, USA 20892
| | - Mike A. Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
| | - Hirotaka Iwaki
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
| | | |
Collapse
|
192
|
Behfar Q, Richter N, Kural M, Clemens A, Behfar SK, Folkerts AK, Fassbender R, Kalbe E, Fink GR, Onur OA. Improved connectivity and cognition due to cognitive stimulation in Alzheimer's disease. Front Aging Neurosci 2023; 15:1140975. [PMID: 37662551 PMCID: PMC10470843 DOI: 10.3389/fnagi.2023.1140975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Background Due to the increasing prevalence of Alzheimer's disease (AD) and the limited efficacy of pharmacological treatment, the interest in non-pharmacological interventions, e.g., cognitive stimulation therapy (CST), to improve cognitive dysfunction and the quality of life of AD patients are on a steady rise. Objectives Here, we examined the efficacy of a CST program specifically conceptualized for AD dementia patients and the neural mechanisms underlying cognitive or behavioral benefits of CST. Methods Using neuropsychological tests and MRI-based measurements of functional connectivity, we examined the (neuro-) psychological status and network changes at two time points: pre vs. post-stimulation (8 to 12 weeks) in the intervention group (n = 15) who received the CST versus a no-intervention control group (n = 15). Results After CST, we observed significant improvement in the Mini-Mental State Examination (MMSE), the Alzheimer's Disease Assessment Scale, cognitive subsection (ADAS-cog), and the behavioral and psychological symptoms of dementia (BPSD) scores. These cognitive improvements were associated with an up-regulated functional connectivity between the left posterior hippocampus and the trunk of the left postcentral gyrus. Conclusion Our data indicate that CST seems to induce short-term global cognition and behavior improvements in mild to moderate AD dementia and enhances resting-state functional connectivity in learning- and memory-associated brain regions. These convergent results prove that even in mild to moderate dementia AD, neuroplasticity can be harnessed to alleviate cognitive impairment with CST.
Collapse
Affiliation(s)
- Qumars Behfar
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Juelich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nils Richter
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Juelich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Merve Kural
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Anne Clemens
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Stefan Kambiz Behfar
- Department of Information Systems, Geneva School of Business Administration (HES-SO Genéve), Carouge, Switzerland
| | - Ann-Kristin Folkerts
- Medical Psychology Neuropsychology and Gender Studies and Center for Neuropsychological Diagnostics and Intervention (CeNDI), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ronja Fassbender
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Juelich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Elke Kalbe
- Medical Psychology Neuropsychology and Gender Studies and Center for Neuropsychological Diagnostics and Intervention (CeNDI), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gereon R. Fink
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Juelich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Oezguer A. Onur
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Juelich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
193
|
Gu Y, Honig LS, Kang MS, Bahl A, Sanchez D, Reyes-Dumeyer D, Manly JJ, Lantigua RA, Dage JL, Brickman AM, Vardarajan BN, Mayeux R. Risk of Alzheimer's Disease is Associated with Longitudinal Changes in Plasma Biomarkers in the Multiethnic Washington Heights, Inwood Columbia Aging Project Cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.11.23293967. [PMID: 37645764 PMCID: PMC10462222 DOI: 10.1101/2023.08.11.23293967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) biomarkers can help differentiate cognitively unimpaired (CU) individuals from mild cognitive impairment (MCI) and dementia. The role of AD biomarkers in predicting cognitive impairment and AD needs examination. METHODS In 628 CU individuals from a multi-ethnic cohort, Aβ42, Aβ40, phosphorylated tau-181 (P-tau181), glial fibrillary acid protein (GFAP), and neurofilament light chain (NfL) were measured in plasma. RESULTS Higher baseline levels of P-tau181/Aβ42 ratio were associated with increased risk of incident dementia. A biomarker pattern (with elevated Aβ42/Aβ40 but low P-tau181/Aβ42) was associated with decreased dementia risk. Compared to CU, participants who developed MCI or dementia had a rapid decrease in the biomarker pattern reflecting AD-specific pathological change. DISCUSSION Elevated levels of AD biomarker P-tau181/Aβ42, by itself or combined with a low Aβ42/Aβ40 level, predicts clinically diagnosed AD. Individuals with a rapid change in these biomarkers may need close monitoring for the potential downward trajectory of cognition.
Collapse
Affiliation(s)
- Yian Gu
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New, York, New York, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York, USA
- Department of Epidemiology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York, USA
| | - Lawrence S. Honig
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New, York, New York, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York, USA
| | - Min Suk Kang
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New, York, New York, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York, USA
| | - Aanya Bahl
- Department of Epidemiology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York, USA
| | - Danurys Sanchez
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New, York, New York, USA
| | - Dolly Reyes-Dumeyer
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New, York, New York, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Jennifer J. Manly
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New, York, New York, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York, USA
| | - Rafael A. Lantigua
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York,New York, USA
| | - Jeffrey L. Dage
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New, York, New York, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York, USA
| | - Badri N Vardarajan
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New, York, New York, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York, USA
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New, York, New York, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York, USA
- Department of Epidemiology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
194
|
Arenare G, Manca R, Caffarra P, Venneri A. Associations between Neuropsychiatric Symptoms and Alzheimer's Disease Biomarkers in People with Mild Cognitive Impairment. Brain Sci 2023; 13:1195. [PMID: 37626552 PMCID: PMC10452057 DOI: 10.3390/brainsci13081195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Neuropsychiatric symptoms (NPS) are associated with faster decline in mild cognitive impairment (MCI). This study aimed to investigate the association between NPS severity and Alzheimer's disease (AD) biomarkers, i.e., amyloid-β (Aβ), phosphorylated tau protein (p-tau) and hippocampal volume ratio (HR), to characterise in more detail MCI patients with a poor prognosis. METHODS A total of 506 individuals with MCI and 99 cognitively unimpaired older adults were selected from the ADNI dataset. The patients were divided into three different groups based on their NPI-Q total scores: no NPS (n = 198), mild NPS (n = 160) and severe NPS (n = 148). Regression models were used to assess the association between the severity of NPS and each biomarker level and positivity status. RESULTS Cerebrospinal fluid Aβ levels were positively associated with older age and lower MMSE scores, while higher p-tau levels were associated with female sex and lower MMSE scores. Only patients with severe NPS had a lower HR (β = -0.18, p = 0.050), i.e., more pronounced medio-temporal atrophy, than those without NPS. DISCUSSION Only HR was associated with the presence of NPS, partially in line with previous evidence showing that severe NPS may be explained primarily by greater grey matter loss. Future longitudinal studies will be needed to ascertain the relevance of this finding.
Collapse
Affiliation(s)
- Giulia Arenare
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Riccardo Manca
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
- Department of Life Sciences, Brunel University London, Uxbridge UB8 3BH, UK
| | - Paolo Caffarra
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Annalena Venneri
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
- Department of Life Sciences, Brunel University London, Uxbridge UB8 3BH, UK
| | | |
Collapse
|
195
|
Yosypyshyn D, Kučikienė D, Ramakers I, Schulz JB, Reetz K, Costa AS. Clinical characteristics of patients with suspected Alzheimer's disease within a CSF Aß-ratio grey zone. Neurol Res Pract 2023; 5:40. [PMID: 37533121 PMCID: PMC10398972 DOI: 10.1186/s42466-023-00262-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND The AT(N) research framework for Alzheimer's disease (AD) remains unclear on how to best deal with borderline cases. Our aim was to characterise patients with suspected AD with a borderline Aß1-42/Aß1-40 ratio in cerebrospinal fluid. METHODS We analysed retrospective data from two cohorts (memory clinic cohort and ADNI) of patients (n = 63) with an Aß1-42/Aß1-40 ratio within a predefined borderline area-Q1 above the validated cut-off value(grey zone). We compared demographic, clinical, neuropsychological and neuroimaging features between grey zone patients and patients with low Aß1-42 (normal Aß ratio but pathological Aß1-42, n = 42) and patients with AD (pathological Aß, P-Tau, und T-Tau, n = 80). RESULTS Patients had mild cognitive impairment or mild dementia and a median age of 72 years. Demographic and general clinical characteristics did not differ between the groups. Patients in the grey zone group were the least impaired in cognition. However, they overlapped with the low Aß1-42 group in verbal episodic memory performance, especially in delayed recall and recognition. The grey zone group had less severe medial temporal atrophy, but mild posterior atrophy and mild white matter hyperintensities, similar to the low Aß1-42 group. CONCLUSIONS Patients in the Aß ratio grey zone were less impaired, but showed clinical overlap with patients on the AD continuum. These borderline patients may be at an earlier disease stage. Assuming an increased risk of AD and progressive cognitive decline, careful consideration of clinical follow-up is recommended when using dichotomous approaches to classify Aß status.
Collapse
Affiliation(s)
- Dariia Yosypyshyn
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Domantė Kučikienė
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Inez Ramakers
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Jörg B Schulz
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
- JARA Institute Molecular Neuroscience and Neuroimaging, RWTH Aachen & Forschungszentrum Jülich, Aachen, Germany
| | - Kathrin Reetz
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany.
- JARA Institute Molecular Neuroscience and Neuroimaging, RWTH Aachen & Forschungszentrum Jülich, Aachen, Germany.
| | - Ana Sofia Costa
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
- JARA Institute Molecular Neuroscience and Neuroimaging, RWTH Aachen & Forschungszentrum Jülich, Aachen, Germany
| |
Collapse
|
196
|
Wuestefeld A, Pichet Binette A, Berron D, Spotorno N, van Westen D, Stomrud E, Mattsson-Carlgren N, Strandberg O, Smith R, Palmqvist S, Glenn T, Moes S, Honer M, Arfanakis K, Barnes LL, Bennett DA, Schneider JA, Wisse LEM, Hansson O. Age-related and amyloid-beta-independent tau deposition and its downstream effects. Brain 2023; 146:3192-3205. [PMID: 37082959 PMCID: PMC10393402 DOI: 10.1093/brain/awad135] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/20/2023] [Accepted: 04/06/2023] [Indexed: 04/22/2023] Open
Abstract
Amyloid-β (Aβ) is hypothesized to facilitate the spread of tau pathology beyond the medial temporal lobe. However, there is evidence that, independently of Aβ, age-related tau pathology might be present outside of the medial temporal lobe. We therefore aimed to study age-related Aβ-independent tau deposition outside the medial temporal lobe in two large cohorts and to investigate potential downstream effects of this on cognition and structural measures. We included 545 cognitively unimpaired adults (40-92 years) from the BioFINDER-2 study (in vivo) and 639 (64-108 years) from the Rush Alzheimer's Disease Center cohorts (ex vivo). 18F-RO948- and 18F-flutemetamol-PET standardized uptake value ratios were calculated for regional tau and global/regional Aβ in vivo. Immunohistochemistry was used to estimate Aβ load and tangle density ex vivo. In vivo medial temporal lobe volumes (subiculum, cornu ammonis 1) and cortical thickness (entorhinal cortex, Brodmann area 35) were obtained using Automated Segmentation for Hippocampal Subfields packages. Thickness of early and late neocortical Alzheimer's disease regions was determined using FreeSurfer. Global cognition and episodic memory were estimated to quantify cognitive functioning. In vivo age-related tau deposition was observed in the medial temporal lobe and in frontal and parietal cortical regions, which was statistically significant when adjusting for Aβ. This was also observed in individuals with low Aβ load. Tau deposition was negatively associated with cortical volumes and thickness in temporal and parietal regions independently of Aβ. The associations between age and cortical volume or thickness were partially mediated via tau in regions with early Alzheimer's disease pathology, i.e. early tau and/or Aβ pathology (subiculum/Brodmann area 35/precuneus/posterior cingulate). Finally, the associations between age and cognition were partially mediated via tau in Brodmann area 35, even when including Aβ-PET as covariate. Results were validated in the ex vivo cohort showing age-related and Aβ-independent increases in tau aggregates in and outside the medial temporal lobe. Ex vivo age-cognition associations were mediated by medial and inferior temporal tau tangle density, while correcting for Aβ density. Taken together, our study provides support for primary age-related tauopathy even outside the medial temporal lobe in vivo and ex vivo, with downstream effects on structure and cognition. These results have implications for our understanding of the spreading of tau outside the medial temporal lobe, also in the context of Alzheimer's disease. Moreover, this study suggests the potential utility of tau-targeting treatments in primary age-related tauopathy, likely already in preclinical stages in individuals with low Aβ pathology.
Collapse
Affiliation(s)
- Anika Wuestefeld
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - David Berron
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - Danielle van Westen
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, SE-222 42 Lund, Sweden
- Image and Function, Skåne University Hospital, SE-205 02 Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, SE-205 02 Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Department of Neurology, Skåne University Hospital, SE-205 02 Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Department of Neurology, Skåne University Hospital, SE-205 02 Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, SE-205 02 Malmö, Sweden
| | - Trevor Glenn
- Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Svenja Moes
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Michael Honer
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Konstantinos Arfanakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Lisa L Barnes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Julie A Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Laura E M Wisse
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, SE-222 42 Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, SE-205 02 Malmö, Sweden
| |
Collapse
|
197
|
Nisenbaum L, Martone R, Chen T, Rajagovindan R, Dent G, Beaver J, Rubel C, Racine A, He P, Harrison K, Dean R, Vandijck M, Haeberlein SB. CSF biomarker concordance with amyloid PET in Phase 3 studies of aducanumab. Alzheimers Dement 2023; 19:3379-3388. [PMID: 36795603 DOI: 10.1002/alz.12919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 02/17/2023]
Abstract
INTRODUCTION We assessed the use of cerebrospinal fluid (CSF) biomarkers as an alternative to positron emission tomography (PET) for brain amyloid beta (Aβ) pathology confirmation in the EMERGE and ENGAGE clinical trials. METHODS EMERGE and ENGAGE were randomized, placebo-controlled, Phase 3 trials of aducanumab in participants with early Alzheimer's disease. Concordance between CSF biomarkers (Aβ42, Aβ40, phosphorylated tau 181, and total tau) and amyloid PET status (visual read) at screening was examined. RESULTS Robust concordance between CSF biomarkers and amyloid PET visual status was observed (for Aβ42/Aβ40, AUC: 0.90; 95% CI: 0.83-0.97; p < 0.0001), confirming CSF biomarkers as a reliable alternative to amyloid PET in these studies. Compared with single CSF biomarkers, CSF biomarker ratios showed better agreement with amyloid PET visual reads, demonstrating high diagnostic accuracy. DISCUSSION These analyses add to the growing body of evidence supporting CSF biomarkers as reliable alternatives to amyloid PET imaging for brain Aβ pathology confirmation. HIGHLIGHTS CSF biomarkers and amyloid PET concordance were assessed in Ph3 aducanumab trials. Robust concordance between CSF biomarkers and amyloid PET was observed. CSF biomarker ratios increased diagnostic accuracy over single CSF biomarkers. CSF Aβ42/Aβ40 demonstrated high concordance with amyloid PET. Results support CSF biomarker testing as a reliable alternative to amyloid PET.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ping He
- Biogen, Cambridge, Massachusetts, USA
| | | | - Robert Dean
- Robert A. Dean Consulting, LLC, Indianapolis, Indiana, USA
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
198
|
Wang ZB, Tan L, Wang HF, Chen SD, Fu Y, Gao PY, Ma YH, Guo Y, Hou JH, Zhang DD, Yu JT. Differences between ante mortem Alzheimer's disease biomarkers in predicting neuropathology at autopsy. Alzheimers Dement 2023; 19:3613-3624. [PMID: 36840620 DOI: 10.1002/alz.12997] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 02/26/2023]
Abstract
INTRODUCTION This study aimed to assess whether biomarkers related to amyloid, tau, and neurodegeneration can accurately predict Alzheimer's disease (AD) neuropathology at autopsy in early and late clinical stages. METHODS We included 100 participants who had ante mortem biomarker measurements and underwent post mortem neuropathological examination. Based on ante mortem clinical diagnosis, participants were divided into non-dementia and dementia, as early or late clinical stages. RESULTS Amyloid positron emission tomography (PET) and cerebrospinal fluid (CSF) amyloid beta (Aβ)42/phosphorylated tau (p-tau)181 showed excellent performance in differentiating autopsy-confirmed AD and predicting the risk of neuropathological changes in early and late clinical stages. However, CSF Aβ42 performed better in the early clinical stage, while CSF p-tau181, CSF t-tau, and plasma p-tau181 performed better in the late clinical stage. DISCUSSION Our findings provide important clinical information that, if using PET, CSF, and plasma biomarkers to detect AD pathology, researchers must consider their differential performances at different clinical stages of AD. HIGHLIGHTS Amyloid PET and CSF Aβ42/p-tau181 were the most promising candidate biomarkers for predicting AD pathology. CSF Aβ42 can serve as a candidate predictive biomarker in the early clinical stage of AD. CSF p-tau181, CSF t-tau, and plasma p-tau181 can serve as candidate predictive biomarkers in the late clinical stage of AD. Combining APOE ε4 genotypes can significantly improve the predictive accuracy of AD-related biomarkers for AD pathology.
Collapse
Affiliation(s)
- Zhi-Bo Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hui-Fu Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Pei-Yang Gao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia-Hui Hou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Dan-Dan Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
199
|
Kerrebijn I, Wainberg M, Zhukovsky P, Chen Y, Davie M, Felsky D, Tripathy SJ. Case-control virtual histology elucidates cell types associated with cortical thickness differences in Alzheimer's disease. Neuroimage 2023; 276:120177. [PMID: 37211192 DOI: 10.1016/j.neuroimage.2023.120177] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 05/08/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023] Open
Abstract
Many neuropsychiatric disorders are characterised by altered cortical thickness, but the cell types underlying these changes remain largely unknown. Virtual histology (VH) approaches map regional patterns of gene expression with regional patterns of MRI-derived phenotypes, such as cortical thickness, to identify cell types associated with case-control differences in those MRI measures. However, this method does not incorporate valuable information of case-control differences in cell type abundance. We developed a novel method, termed case-control virtual histology (CCVH), and applied it to Alzheimer's disease (AD) and dementia cohorts. Leveraging a multi-region gene expression dataset of AD cases (n = 40) and controls (n = 20), we quantified AD case-control differential expression of cell type-specific markers across 13 brain regions. We then correlated these expression effects with MRI-derived AD case-control cortical thickness differences across the same regions. Cell types with spatially concordant AD-related effects were identified through resampling marker correlation coefficients. Among regions thinner in AD, gene expression patterns identified by CCVH suggested fewer excitatory and inhibitory neurons, and greater proportions of astrocytes, microglia, oligodendrocytes, oligodendrocyte precursor cells, and endothelial cells in AD cases vs. controls. In contrast, original VH identified expression patterns suggesting that excitatory but not inhibitory neuron abundance was associated with thinner cortex in AD, despite the fact that both types of neurons are known to be lost in the disorder. Compared to original VH, cell types identified through CCVH are more likely to directly underlie cortical thickness differences in AD. Sensitivity analyses suggest our results are largely robust to specific analysis choices, like numbers of cell type-specific marker genes used and background gene sets used to construct null models. As more multi-region brain expression datasets become available, CCVH will be useful for identifying the cellular correlates of cortical thickness across neuropsychiatric illnesses.
Collapse
Affiliation(s)
- Isabel Kerrebijn
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Michael Wainberg
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Peter Zhukovsky
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Yuxiao Chen
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Melanie Davie
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Daniel Felsky
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto ON, Canada
| | - Shreejoy J Tripathy
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
200
|
Deming Y, Vasiljevic E, Morrow A, Miao J, Van Hulle C, Jonaitis E, Ma Y, Whitenack V, Kollmorgen G, Wild N, Suridjan I, Shaw LM, Asthana S, Carlsson CM, Johnson SC, Zetterberg H, Blennow K, Bendlin BB, Lu Q, Engelman CD. Neuropathology-based APOE genetic risk score better quantifies Alzheimer's risk. Alzheimers Dement 2023; 19:3406-3416. [PMID: 36795776 PMCID: PMC10427737 DOI: 10.1002/alz.12990] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 02/17/2023]
Abstract
INTRODUCTION Apolipoprotein E (APOE) ε4-carrier status or ε4 allele count are included in analyses to account for the APOE genetic effect on Alzheimer's disease (AD); however, this does not account for protective effects of APOE ε2 or heterogeneous effect of ε2, ε3, and ε4 haplotypes. METHODS We leveraged results from an autopsy-confirmed AD study to generate a weighted risk score for APOE (APOE-npscore). We regressed cerebrospinal fluid (CSF) amyloid and tau biomarkers on APOE variables from the Wisconsin Registry for Alzheimer's Prevention (WRAP), Wisconsin Alzheimer's Disease Research Center (WADRC), and Alzheimer's Disease Neuroimaging Initiative (ADNI). RESULTS The APOE-npscore explained more variance and provided a better model fit for all three CSF measures than APOE ε4-carrier status and ε4 allele count. These findings were replicated in ADNI and observed in subsets of cognitively unimpaired (CU) participants. DISCUSSION The APOE-npscore reflects the genetic effect on neuropathology and provides an improved method to account for APOE in AD-related analyses.
Collapse
Affiliation(s)
- Yuetiva Deming
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Eva Vasiljevic
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Autumn Morrow
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jiacheng Miao
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Carol Van Hulle
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Erin Jonaitis
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yue Ma
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Vanessa Whitenack
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sanjay Asthana
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Geriatric Research Education and Clinical Center, Madison, Wisconsin, USA
| | - Cynthia M Carlsson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Geriatric Research Education and Clinical Center, Madison, Wisconsin, USA
| | - Sterling C Johnson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Geriatric Research Education and Clinical Center, Madison, Wisconsin, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Barbara B Bendlin
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Qiongshi Lu
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Corinne D Engelman
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|