1
|
Bamford AR, Adams JN, Kim S, McMillan LC, Malhas R, Mapstone M, Hitt BD, Yassa MA, Thomas EA. The amyloid beta 42/38 ratio as a plasma biomarker of early memory deficits in cognitively unimpaired older adults. Neurobiol Aging 2024; 144:12-18. [PMID: 39241563 DOI: 10.1016/j.neurobiolaging.2024.08.009] [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: 03/09/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
The amyloid beta (Aβ) 42/40 ratio has been widely studied as a biomarker in Alzheimer's disease (AD); however, other Aβ peptides could also represent relevant biomarkers. We measured levels of Aβ38/40/42 in plasma samples from cognitively-unimpaired older adults and determined the relationships between Aβ levels and amyloid positron-emission-tomography (PET) and performance on a learning and memory task. We found that all Aβ peptides individually and the Aβ42/40 ratio, but not the Aβ42/38 ratio, were significantly correlated with brain amyloid (Aβ-PET). Multiple linear modeling, adjusting for age, sex, education, APOE4 and Aβ-PET showed significant associations between the Aβ42/38 ratio and memory. Further, associations between the Aβ42/38 ratio and learning scores were stronger in males and in Aβ-PET-negative individuals. In contrast, no significant associations were detected between the Aβ42/40 ratio and any learning measure. These studies implicate the Aβ42/38 ratio as a biomarker to assess early memory deficits and underscore the utility of the Aβ38 fragment as an important biomarker in the AD field.
Collapse
Affiliation(s)
- Alison R Bamford
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA; Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA
| | - Jenna N Adams
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, USA
| | - Soyun Kim
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, USA
| | - Liv C McMillan
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Rond Malhas
- Department of Neurology, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Mark Mapstone
- Department of Neurology, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Brian D Hitt
- Department of Neurology, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Michael A Yassa
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, USA
| | - Elizabeth A Thomas
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA; Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
2
|
Prabha S, Sajad M, Hasan GM, Islam A, Imtaiyaz Hassan M, Thakur SC. Recent advancement in understanding of Alzheimer's disease: Risk factors, subtypes, and drug targets and potential therapeutics. Ageing Res Rev 2024; 101:102476. [PMID: 39222668 DOI: 10.1016/j.arr.2024.102476] [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: 07/13/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Alzheimer's disease (AD) is a significant neocortical degenerative disorder characterized by the progressive loss of neurons and secondary alterations in white matter tracts. Understanding the risk factors and mechanisms underlying AD is crucial for developing effective treatments. The risk factors associated with AD encompass a wide range of variables, including gender differences, family history, and genetic predispositions. Additionally, environmental factors such as air pollution and lifestyle-related conditions like cardiovascular disease, gut pathogens, and liver pathology contribute substantially to the development and progression of AD and its subtypes. This review provides current update and deeper insights into the role of diverse risk factors, categorizing AD into its distinct subtypes and elucidating their specific pathophysiological mechanisms. Unlike previous studies that often focus on isolated aspects of AD, our review integrates these factors to offer a comprehensive understanding of the disease. Furthermore, the review explores a variety of drug targets linked to the neuropathology of different AD subtypes, highlighting the potential for targeted therapeutic interventions. We further discussed the novel therapeutic options and categorized them according to their targets. The roles of different drug targets were comprehensively studied, and the mechanism of action of their inhibitors was discussed in detail. By comprehensively covering the interplay of risk factors, subtype differentiation, and drug targets, this review provides a deeper understanding of AD and suggests directions for future research and therapeutic strategies.
Collapse
Affiliation(s)
- Sneh Prabha
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohd Sajad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| | - Sonu Chand Thakur
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
3
|
Čihák M, Horáková H, Vyhnálek M, Veverová K, Matušková V, Laczó J, Hort J, Nikolai T. Evaluation of Differential Diagnostics Potential of Uniform Data Set 2 Neuropsychology Battery Using Alzheimer's Disease Biomarkers. Arch Clin Neuropsychol 2024; 39:839-848. [PMID: 38582748 PMCID: PMC11504696 DOI: 10.1093/arclin/acae028] [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: 10/18/2023] [Revised: 02/20/2024] [Accepted: 03/12/2024] [Indexed: 04/08/2024] Open
Abstract
OBJECTIVE This study aims to evaluate the efficacy of the Uniform Data Set (UDS) 2 battery in distinguishing between individuals with mild cognitive impairment (MCI) attributable to Alzheimer's disease (MCI-AD) and those with MCI due to other causes (MCI-nonAD), based on contemporary AT(N) biomarker criteria. Despite the implementation of the novel UDS 3 battery, the UDS 2 battery is still used in several non-English-speaking countries. METHODS We employed a cross-sectional design. A total of 113 Czech participants with MCI underwent a comprehensive diagnostic assessment, including cerebrospinal fluid biomarker evaluation, resulting in two groups: 45 individuals with prodromal AD (A+T+) and 68 participants with non-Alzheimer's pathological changes or normal AD biomarkers (A-). Multivariable logistic regression analyses were employed with neuropsychological test scores and demographic variables as predictors and AD status as an outcome. Model 1 included UDS 2 scores that differed between AD and non-AD groups (Logical Memory delayed recall), Model 2 employed also Letter Fluency and Rey's Auditory Verbal Learning Test (RAVLT). The two models were compared using area under the receiver operating characteristic curves. We also created separate logistic regression models for each of the UDS 2 scores. RESULTS Worse performance in delayed recall of Logical Memory significantly predicted the presence of positive AD biomarkers. In addition, the inclusion of Letter Fluency RAVLT into the model significantly enhanced its discriminative capacity. CONCLUSION Our findings demonstrate that using Letter Fluency and RAVLT alongside the UDS 2 battery can enhance its potential for differential diagnostics.
Collapse
Affiliation(s)
- Martin Čihák
- Department of Neurology, First Faculty of Medicine, Charles University, 121 08 Prague, Czech Republic
| | - Hana Horáková
- Department of Neurology, Second Faculty of Medicine and Motol University Hospital, Charles University, 150 06 Prague, Czech Republic
- Department of Clinical Psychology, Motol University Hospital, 150 06 Prague, Czech Republic
| | - Martin Vyhnálek
- Department of Neurology, First Faculty of Medicine, Charles University, 121 08 Prague, Czech Republic
| | - Kateřina Veverová
- Department of Neurology, Second Faculty of Medicine and Motol University Hospital, Charles University, 150 06 Prague, Czech Republic
- Department of Psychology, Faculty of Arts, Charles University, 116 38 Prague, Czech Republic
| | - Veronika Matušková
- Department of Neurology, Second Faculty of Medicine and Motol University Hospital, Charles University, 150 06 Prague, Czech Republic
| | - Jan Laczó
- Department of Neurology, Second Faculty of Medicine and Motol University Hospital, Charles University, 150 06 Prague, Czech Republic
| | - Jakub Hort
- Department of Neurology, Second Faculty of Medicine and Motol University Hospital, Charles University, 150 06 Prague, Czech Republic
| | - Tomáš Nikolai
- Department of Neurology, Second Faculty of Medicine and Motol University Hospital, Charles University, 150 06 Prague, Czech Republic
- Department of Clinical Psychology, Motol University Hospital, 150 06 Prague, Czech Republic
- Department of Psychology, Faculty of Arts, Charles University, 116 38 Prague, Czech Republic
| |
Collapse
|
4
|
Andrade K, Pacella V. The unique role of anosognosia in the clinical progression of Alzheimer's disease: a disorder-network perspective. Commun Biol 2024; 7:1384. [PMID: 39448784 PMCID: PMC11502706 DOI: 10.1038/s42003-024-07076-7] [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: 06/20/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Alzheimer's disease (AD) encompasses a long continuum from a preclinical phase, characterized by neuropathological alterations albeit normal cognition, to a symptomatic phase, marked by its clinical manifestations. Yet, the neural mechanisms responsible for cognitive decline in AD patients remain poorly understood. Here, we posit that anosognosia, emerging from an error-monitoring failure due to early amyloid-β deposits in the posterior cingulate cortex, plays a causal role in the clinical progression of AD by preventing patients from being aware of their deficits and implementing strategies to cope with their difficulties, thus fostering a vicious circle of cognitive decline.
Collapse
Affiliation(s)
- Katia Andrade
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Assistance Publique-Hôpitaux de Paris (AP-HP), Sorbonne University, Pitié-Salpêtrière Hospital, 75013, Paris, France.
- FrontLab, Paris Brain Institute (Institut du Cerveau, ICM), AP-HP, Pitié-Salpêtrière Hospital, 75013, Paris, France.
| | - Valentina Pacella
- IUSS Cognitive Neuroscience (ICON) Center, Scuola Universitaria Superiore IUSS, Pavia, 27100, Italy
- Brain Connectivity and Behaviour Laboratory, Paris, France
| |
Collapse
|
5
|
Meng L, Jin H, Yulug B, Altay O, Li X, Hanoglu L, Cankaya S, Coskun E, Idil E, Nogaylar R, Ozsimsek A, Shoaie S, Turkez H, Nielsen J, Zhang C, Borén J, Uhlén M, Mardinoglu A. Multi-omics analysis reveals the key factors involved in the severity of the Alzheimer's disease. Alzheimers Res Ther 2024; 16:213. [PMID: 39358810 PMCID: PMC11448018 DOI: 10.1186/s13195-024-01578-6] [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: 02/26/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder with a global impact, yet its pathogenesis remains poorly understood. While age, metabolic abnormalities, and accumulation of neurotoxic substances are potential risk factors for AD, their effects are confounded by other factors. To address this challenge, we first utilized multi-omics data from 87 well phenotyped AD patients and generated plasma proteomics and metabolomics data, as well as gut and saliva metagenomics data to investigate the molecular-level alterations accounting the host-microbiome interactions. Second, we analyzed individual omics data and identified the key parameters involved in the severity of the dementia in AD patients. Next, we employed Artificial Intelligence (AI) based models to predict AD severity based on the significantly altered features identified in each omics analysis. Based on our integrative analysis, we found the clinical relevance of plasma proteins, including SKAP1 and NEFL, plasma metabolites including homovanillate and glutamate, and Paraprevotella clara in gut microbiome in predicting the AD severity. Finally, we validated the predictive power of our AI based models by generating additional multi-omics data from the same group of AD patients by following up for 3 months. Hence, we observed that these results may have important implications for the development of potential diagnostic and therapeutic approaches for AD patients.
Collapse
Affiliation(s)
- Lingqi Meng
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Han Jin
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Burak Yulug
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Ozlem Altay
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Xiangyu Li
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Lutfu Hanoglu
- Department of Neurology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Seyda Cankaya
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Ebru Coskun
- Department of Neurology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ezgi Idil
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Rahim Nogaylar
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Ahmet Ozsimsek
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Saeed Shoaie
- Centre for Host-Microbiome Interaction's, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden.
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden.
- Centre for Host-Microbiome Interaction's, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK.
| |
Collapse
|
6
|
Puerta R, de Rojas I, García-González P, Olivé C, Sotolongo-Grau O, García-Sánchez A, García-Gutiérrez F, Montrreal L, Pablo Tartari J, Sanabria Á, Pytel V, Lage C, Quintela I, Aguilera N, Rodriguez-Rodriguez E, Alarcón-Martín E, Orellana A, Pastor P, Pérez-Tur J, Piñol-Ripoll G, de Munian AL, García-Alberca JM, Royo JL, Bullido MJ, Álvarez V, Real LM, Anchuelo AC, Gómez-Garre D, Larrad MTM, Franco-Macías E, Mir P, Medina M, Sánchez-Valle R, Dols-Icardo O, Sáez ME, Carracedo Á, Tárraga L, Alegret M, Valero S, Marquié M, Boada M, Juan PS, Cavazos JE, Cabrera A, Cano A. Connecting genomic and proteomic signatures of amyloid burden in the brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.06.24313124. [PMID: 39281766 PMCID: PMC11398581 DOI: 10.1101/2024.09.06.24313124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background Alzheimer's disease (AD) has a high heritable component characteristic of complex diseases, yet many of the genetic risk factors remain unknown. We combined genome-wide association studies (GWAS) on amyloid endophenotypes measured in cerebrospinal fluid (CSF) and positron emission tomography (PET) as surrogates of amyloid pathology, which may be helpful to understand the underlying biology of the disease. Methods We performed a meta-analysis of GWAS of CSF Aβ42 and PET measures combining six independent cohorts (n=2,076). Due to the opposite effect direction of Aβ phenotypes in CSF and PET measures, only genetic signals in the opposite direction were considered for analysis (n=376,599). Polygenic risk scores (PRS) were calculated and evaluated for AD status and amyloid endophenotypes. We then searched the CSF proteome signature of brain amyloidosis using SOMAscan proteomic data (Ace cohort, n=1,008) and connected it with GWAS results of loci modulating amyloidosis. Finally, we compared our results with a large meta-analysis using publicly available datasets in CSF (n=13,409) and PET (n=13,116). This combined approach enabled the identification of overlapping genes and proteins associated with amyloid burden and the assessment of their biological significance using enrichment analyses. Results After filtering the meta-GWAS, we observed genome-wide significance in the rs429358-APOE locus and nine suggestive hits were annotated. We replicated the APOE loci using the large CSF-PET meta-GWAS and identified multiple AD-associated genes as well as the novel GADL1 locus. Additionally, we found a significant association between the AD PRS and amyloid levels, whereas no significant association was found between any Aβ PRS with AD risk. CSF SOMAscan analysis identified 1,387 FDR-significant proteins associated with CSF Aβ42 levels. The overlap among GWAS loci and proteins associated with amyloid burden was very poor (n=35). The enrichment analysis of overlapping hits strongly suggested several signalling pathways connecting amyloidosis with the anchored component of the plasma membrane, synapse physiology and mental disorders that were replicated in the large CSF-PET meta-analysis. Conclusions The strategy of combining CSF and PET amyloid endophenotypes GWAS with CSF proteome analyses might be effective for identifying signals associated with the AD pathological process and elucidate causative molecular mechanisms behind the amyloid mobilization in AD.
Collapse
Affiliation(s)
- Raquel Puerta
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- Universitat de Barcelona (UB)
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Clàudia Olivé
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | | | | | | | - Laura Montrreal
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Ángela Sanabria
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Vanesa Pytel
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Carmen Lage
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Inés Quintela
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Nuria Aguilera
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Eloy Rodriguez-Rodriguez
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | | | - Adelina Orellana
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pau Pastor
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain
- The Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Jordi Pérez-Tur
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unitat de Genètica Molecular, Institut de Biomedicina de València-CSIC, Valencia, Spain
- Unidad Mixta de Neurologia Genètica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Hospital Universitari Santa Maria de Lleida, Lleida, Spain
- Institut de Recerca Biomedica de Lleida (IRBLLeida), Lleida, Spain
| | - Adolfo López de Munian
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Department of Neurology. Hospital Universitario Donostia. San Sebastian, Spain
- Department of Neurosciences. Faculty of Medicine and Nursery. University of the Basque Country, San Sebastián, Spain
- Neurosciences Area. Instituto Biodonostia. San Sebastian, Spain
| | - Jose María García-Alberca
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Alzheimer Research Center & Memory Clinic, Andalusian Institute for Neuroscience, Málaga, Spain
| | - Jose Luís Royo
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
| | - María Jesús Bullido
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC)
- Instituto de Investigacion Sanitaria ‘Hospital la Paz’ (IdIPaz), Madrid, Spain
- Universidad Autónoma de Madrid
| | - Victoria Álvarez
- Laboratorio de Genética. Hospital Universitario Central de Asturias, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Luis Miguel Real
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
- Unidad Clínica de Enfermedades Infecciosas y Microbiología.Hospital Universitario de Valme, Sevilla, Spain
| | - Arturo Corbatón Anchuelo
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
| | - Dulcenombre Gómez-Garre
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos; Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid (UCM)
- Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - María Teresa Martínez Larrad
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)
| | - Emilio Franco-Macías
- Dementia Unit, Department of Neurology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
| | - Pablo Mir
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología. Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Miguel Medina
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center
| | - Raquel Sánchez-Valle
- Alzheimer’s disease and other cognitive disorders unit. Service of Neurology. Hospital Clínic of Barcelona. Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Oriol Dols-Icardo
- Department of Neurology, Sant Pau Memory Unit, Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Ángel Carracedo
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica – CIBERER-IDIS, Santiago de Compostela, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Montse Alegret
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pascual Sánchez Juan
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Jose Enrique Cavazos
- South Texas Medical Science Training Program, University of Texas Health San Antonio, San Antonio
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| | - Alfredo Cabrera
- Neuroscience Therapeutic Area, Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Amanda Cano
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Alzheimer’s Disease Neuroimaging Initiative.
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| |
Collapse
|
7
|
Karikari T, Chen Y, Zeng X, Olvera-Rojas M, Sehrawat A, Lafferty T, Pascoal T, Villemagne V, Solis-Urra P, Triviño-Ibañez E, Gómez-Rí M, Cohen A, Ikonomovic M, Esteban-Cornejo I, Erickson K, Lopez O, Yates N. A streamlined, resource-efficient immunoprecipitation-mass spectrometry method for quantifying plasma amyloid-β biomarkers in Alzheimer's disease. RESEARCH SQUARE 2024:rs.3.rs-4947448. [PMID: 39281858 PMCID: PMC11398558 DOI: 10.21203/rs.3.rs-4947448/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
High-performance, resource-efficient methods for plasma amyloid-β (Aβ) quantification in Alzheimer's disease are lacking; existing mass spectrometry-based assays are resource- and time-intensive. We developed a streamlined mass spectrometry method with a single immunoprecipitation step, an optimized buffer system, and ≤75% less antibody requirement. Analytical and clinical performances were compared with an in-house reproduced version of a well-known two-step assay. The streamlined assay showed high dilution linearity (r2>0.99) and precision (< 10% coefficient of variation), low quantification limits (Aβ1-40: 12.5 pg/ml; Aβ1-42: 3.125 pg/ml), and high signal correlation (r2~0.7) with the two-step immunoprecipitation assay. The novel single-step assay showed more efficient recovery of Aβ peptides via fewer immunoprecipitation steps, with significantly higher signal-to-noise ratios, even at plasma sample volumes down to 50 pl. Both assays had equivalent performances in distinguishing non-elevated vs. elevated brain Aβ-PET individuals. The new method enables simplified yet robust evaluation of plasma Aβ biomarkers in Alzheimer's disease.
Collapse
|
8
|
Shao K, Hu X, Kleineidam L, Stark M, Altenstein S, Amthauer H, Boecker H, Buchert R, Buerger K, Butryn M, Cai Y, Cai Y, Cosma NC, Chen G, Chen Z, Daamen M, Drzezga A, Düzel E, Essler M, Ewers M, Fliessbach K, Gaertner FC, Glanz W, Guo T, Hansen N, He B, Janowitz D, Kilimann I, Krause BJ, Lan G, Lange C, Laske C, Li Y, Li R, Liu L, Lu J, Meng F, Munk MH, Peters O, Perneczky R, Priller J, Ramirez A, Rauchmann B, Reimold M, Rominger A, Rostamzadeh A, Roy‐Kluth N, Schneider A, Spottke A, Spruth EJ, Sun P, Teipel S, Wang X, Wei M, Wei Y, Wiltfang J, Yan S, Yang J, Yu X, Zhang M, Zhang L, Wagner M, Jessen F, Han Y, Kuhn E. Amyloid and SCD jointly predict cognitive decline across Chinese and German cohorts. Alzheimers Dement 2024; 20:5926-5939. [PMID: 39072956 PMCID: PMC11497667 DOI: 10.1002/alz.14119] [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: 11/17/2023] [Revised: 04/10/2024] [Accepted: 05/11/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Subjective cognitive decline (SCD) in amyloid-positive (Aβ+) individuals was proposed as a clinical indicator of Stage 2 in the Alzheimer's disease (AD) continuum, but this requires further validation across cultures, measures, and recruitment strategies. METHODS Eight hundred twenty-one participants from SILCODE and DELCODE cohorts, including normal controls (NC) and individuals with SCD recruited from the community or from memory clinics, underwent neuropsychological assessments over up to 6 years. Amyloid positivity was derived from positron emission tomography or plasma biomarkers. Global cognitive change was analyzed using linear mixed-effects models. RESULTS In the combined and stratified cohorts, Aβ+ participants with SCD showed steeper cognitive decline or diminished practice effects compared with NC or Aβ- participants with SCD. These findings were confirmed using different operationalizations of SCD and amyloid positivity, and across different SCD recruitment settings. DISCUSSION Aβ+ individuals with SCD in German and Chinese populations showed greater global cognitive decline and could be targeted for interventional trials. HIGHLIGHTS SCD in amyloid-positive (Aβ+) participants predicts a steeper cognitive decline. This finding does not rely on specific SCD or amyloid operationalization. This finding is not specific to SCD patients recruited from memory clinics. This finding is valid in both German and Chinese populations. Aβ+ older adults with SCD could be a target population for interventional trials.
Collapse
Affiliation(s)
- Kai Shao
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of PsychiatryMedical FacultyUniversity of CologneCologneGermany
| | - Xiaochen Hu
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of PsychiatryMedical FacultyUniversity of CologneCologneGermany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| | - Melina Stark
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE)BerlinGermany
- Department of Psychiatry and PsychotherapyCharitéBerlinGermany
| | - Holger Amthauer
- Department of Nuclear MedicineCharité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, and Humboldt‐Universität zu BerlinBerlinGermany
| | - Henning Boecker
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Clinical Functional Imaging Group, Department of Diagnostic and Interventional RadiologyUniversity Hospital BonnBonnGermany
| | - Ralph Buchert
- Department of Nuclear MedicineCharité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, and Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
- Department of Diagnostic and Interventional Radiology and Nuclear MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
- Institute for Stroke and Dementia Research (ISD)University Hospital, LMU MunichMunichGermany
| | - Michaela Butryn
- German Center for Neurodegenerative Diseases (DZNE)MagdeburgGermany
- Institute of Cognitive Neurology and Dementia Research (IKND)Otto‐von‐Guericke UniversityMagdeburgGermany
| | - Yanning Cai
- Department of clinical biobankXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Yue Cai
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Nicoleta Carmen Cosma
- Department of Psychiatry and PsychotherapyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Guanqun Chen
- Department of NeurologyBeijing ChaoYang Hospital of Capital Medical UniversityBeijingChina
| | - Zhigeng Chen
- Department of Radiology and Nuclear MedicineXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Marcel Daamen
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Alexander Drzezga
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Institute of Neuroscience and Medicine (INM‐2)Molecular Organization of the Brain, Forschungszentrum JülichJülichGermany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE)MagdeburgGermany
- Institute of Cognitive Neurology and Dementia Research (IKND)Otto‐von‐Guericke UniversityMagdeburgGermany
| | - Markus Essler
- Department of Nuclear MedicineUniversity Hospital BonnBonnGermany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
- Institute for Stroke and Dementia Research (ISD)University Hospital, LMU MunichMunichGermany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| | | | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE)MagdeburgGermany
- Institute of Cognitive Neurology and Dementia Research (IKND)Otto‐von‐Guericke UniversityMagdeburgGermany
| | - Tengfei Guo
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center GoettingenUniversity of GoettingenGoettingenGermany
| | - Beiqi He
- School of Information and Communication EngineeringHainan UniversityHaikouChina
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD)University Hospital, LMU MunichMunichGermany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE)RostockGermany
- Department of Psychosomatic MedicineRostock University Medical CenterRostockGermany
| | - Bernd J. Krause
- Department of Nuclear MedicineRostock University Medical CentreRostockGermany
| | - Guoyu Lan
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhenChina
| | - Catharina Lange
- Department of Nuclear MedicineCharité ‐ Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, and Humboldt‐Universität zu BerlinBerlinGermany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and PsychotherapyUniversity of TübingenTübingenGermany
| | - Yuxia Li
- Department of NeurologyTangshan Central HospitalTanshanChina
| | - Ruixian Li
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Lin Liu
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhenChina
| | - Jie Lu
- Department of Radiology and Nuclear MedicineXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Fansheng Meng
- Medical Imaging Department of Hainan Cancer HospitalHaikouChina
| | - Matthias H. Munk
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Department of Psychiatry and PsychotherapyUniversity of TübingenTübingenGermany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE)BerlinGermany
- Department of Psychiatry and PsychotherapyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
- Department of Psychiatry and PsychotherapyUniversity Hospital, LMU MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy) MunichMunichGermany
- Ageing Epidemiology Research Unit (AGE), School of Public HealthImperial College LondonLondonUK
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE)BerlinGermany
- Department of Psychiatry and PsychotherapyCharitéBerlinGermany
- University of Edinburgh and UK DRIEdinburghUK
- School of Medicine, Department of Psychiatry and PsychotherapyTechnical University of MunichMunichGermany
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
- Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneKölnGermany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital CologneUniversity of CologneKölnGermany
- Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesSan AntonioTexasUSA
| | - Boris‐Stephan Rauchmann
- Department of Psychiatry and PsychotherapyUniversity Hospital, LMU MunichMunichGermany
- Sheffield Institute for Translational Neuroscience (SITraN)University of SheffieldSheffieldUK
- Department of NeuroradiologyUniversity Hospital LMUMunichGermany
| | - Matthias Reimold
- Department of Nuclear Medicine and Clinical Molecular ImagingEberhard‐Karls‐UniversityTuebingenGermany
| | - Axel Rominger
- Department of Nuclear MedicineLudwig‐Maximilian‐University MunichMunichGermany
- Department of Nuclear Medicine, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
| | - Ayda Rostamzadeh
- Department of PsychiatryMedical FacultyUniversity of CologneCologneGermany
| | - Nina Roy‐Kluth
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of NeurologyUniversity of BonnBonnGermany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE)BerlinGermany
- Department of Psychiatry and PsychotherapyCharitéBerlinGermany
| | - Pan Sun
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhenChina
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE)RostockGermany
- Department of Psychosomatic MedicineRostock University Medical CenterRostockGermany
| | - Xiao Wang
- Department of Psychiatry and PsychotherapyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Min Wei
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Yongzhe Wei
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center GoettingenUniversity of GoettingenGoettingenGermany
- German Center for Neurodegenerative Diseases (DZNE)GoettingenGermany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical SciencesUniversity of AveiroAveiroPortugal
| | - Shaozhen Yan
- Department of Radiology and Nuclear MedicineXuanWu Hospital of Capital Medical UniversityBeijingChina
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
| | - Jie Yang
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Xianfeng Yu
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Mingkai Zhang
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
| | - Liang Zhang
- School of Information and Communication EngineeringHainan UniversityHaikouChina
| | | | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of PsychiatryMedical FacultyUniversity of CologneCologneGermany
- Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneKölnGermany
| | - Ying Han
- Department of NeurologyXuanWu Hospital of Capital Medical UniversityBeijingChina
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- School of Biomedical EngineeringHainan UniversityHaikouChina
- Center of Alzheimer's DiseaseBeijing Institute for Brain DisordersBeijingChina
- National Clinical Research Center for Geriatric DisordersBeijingChina
- The Central Hospital of KaramayXinjiangChina
| | - Elizabeth Kuhn
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Old Age Psychiatry and Cognitive DisordersUniversity of Bonn Medical CenterBonnGermany
| |
Collapse
|
9
|
Fang S, Zhang K, Liu D, Yang Y, Xi H, Xie W, Diao K, Rao Z, Wang D, Yang W. Polyphenol-based polymer nanoparticles for inhibiting amyloid protein aggregation: recent advances and perspectives. Front Nutr 2024; 11:1408620. [PMID: 39135555 PMCID: PMC11317421 DOI: 10.3389/fnut.2024.1408620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
Polyphenols are a group of naturally occurring compounds that possess a range of biological properties capable of potentially mitigating or preventing the progression of age-related cognitive decline and Alzheimer's disease (AD). AD is a chronic neurodegenerative disease known as one of the fast-growing diseases, especially in the elderly population. Moreover, as the primary etiology of dementia, it poses challenges for both familial and societal structures, while also imposing a significant economic strain. There is currently no pharmacological intervention that has demonstrated efficacy in treating AD. While polyphenols have exhibited potential in inhibiting the pathological hallmarks of AD, their limited bioavailability poses a significant challenge in their therapeutic application. Furthermore, in order to address the therapeutic constraints, several polymer nanoparticles are being explored as improved therapeutic delivery systems to optimize the pharmacokinetic characteristics of polyphenols. Polymer nanoparticles have demonstrated advantageous characteristics in facilitating the delivery of polyphenols across the blood-brain barrier, resulting in their efficient distribution within the brain. This review focuses on amyloid-related diseases and the role of polyphenols in them, in addition to discussing the anti-amyloid effects and applications of polyphenol-based polymer nanoparticles.
Collapse
Affiliation(s)
- Shuzhen Fang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Kangyi Zhang
- State Key Laboratory of Tea Plant Biology and Utilization, Key Laboratory of Food Nutrition and Safety, School of Tea, Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Danqing Liu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Yulong Yang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Hu Xi
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Wenting Xie
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Ke Diao
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhihong Rao
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Wenming Yang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| |
Collapse
|
10
|
Orioli R, Belluti F, Gobbi S, Rampa A, Bisi A. Naturally Inspired Coumarin Derivatives in Alzheimer's Disease Drug Discovery: Latest Advances and Current Challenges. Molecules 2024; 29:3514. [PMID: 39124919 PMCID: PMC11313984 DOI: 10.3390/molecules29153514] [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: 06/28/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
The main feature of neurodegenerative diseases, including Alzheimer's disease, is the network of complex and not fully recognized neuronal pathways and targets involved in their onset and progression. The therapeutic treatment, at present mainly symptomatic, could benefit from a polypharmacological approach based on the development of a single molecular entity designed to simultaneously modulate different validated biological targets. This strategy is principally based on molecular hybridization, obtained by linking or merging different chemical moieties acting with synergistic and/or complementary mechanisms. The coumarin core, widely found in nature, endowed with a recognized broad spectrum of pharmacological activities, large synthetic accessibility and favourable pharmacokinetic properties, appears as a valuable, privileged scaffold to be properly modified in order to obtain compounds able to engage different selected targets. The scientific literature has long been interested in the multifaceted profiles of coumarin derivatives, and in this review, a survey of the most important results of the last four years, on both natural and synthetic coumarin-based compounds, regarding the development of anti-Alzheimer's compounds is reported.
Collapse
Affiliation(s)
| | | | | | - Angela Rampa
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy; (R.O.); (F.B.); (S.G.)
| | - Alessandra Bisi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy; (R.O.); (F.B.); (S.G.)
| |
Collapse
|
11
|
Kasri A, Camporesi E, Gkanatsiou E, Boluda S, Brinkmalm G, Stimmer L, Ge J, Hanrieder J, Villain N, Duyckaerts C, Vermeiren Y, Pape SE, Nicolas G, Laquerrière A, De Deyn PP, Wallon D, Blennow K, Strydom A, Zetterberg H, Potier MC. Amyloid-β peptide signature associated with cerebral amyloid angiopathy in familial Alzheimer's disease with APPdup and Down syndrome. Acta Neuropathol 2024; 148:8. [PMID: 39026031 PMCID: PMC11258176 DOI: 10.1007/s00401-024-02756-4] [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: 02/02/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024]
Abstract
Alzheimer's disease (AD) is characterized by extracellular amyloid plaques containing amyloid-β (Aβ) peptides, intraneuronal neurofibrillary tangles, extracellular neuropil threads, and dystrophic neurites surrounding plaques composed of hyperphosphorylated tau protein (pTau). Aβ can also deposit in blood vessel walls leading to cerebral amyloid angiopathy (CAA). While amyloid plaques in AD brains are constant, CAA varies among cases. The study focuses on differences observed between rare and poorly studied patient groups with APP duplications (APPdup) and Down syndrome (DS) reported to have higher frequencies of elevated CAA levels in comparison to sporadic AD (sAD), most of APP mutations, and controls. We compared Aβ and tau pathologies in postmortem brain tissues across cases and Aβ peptides using mass spectrometry (MS). We further characterized the spatial distribution of Aβ peptides with MS-brain imaging. While intraparenchymal Aβ deposits were numerous in sAD, DS with AD (DS-AD) and AD with APP mutations, these were less abundant in APPdup. On the contrary, Aβ deposits in the blood vessels were abundant in APPdup and DS-AD while only APPdup cases displayed high Aβ deposits in capillaries. Investigation of Aβ peptide profiles showed a specific increase in Aβx-37, Aβx-38 and Aβx-40 but not Aβx-42 in APPdup cases and to a lower extent in DS-AD cases. Interestingly, N-truncated Aβ2-x peptides were particularly increased in APPdup compared to all other groups. This result was confirmed by MS-imaging of leptomeningeal and parenchymal vessels from an APPdup case, suggesting that CAA is associated with accumulation of shorter Aβ peptides truncated both at N- and C-termini in blood vessels. Altogether, this study identified striking differences in the localization and composition of Aβ deposits between AD cases, particularly APPdup and DS-AD, both carrying three genomic copies of the APP gene. Detection of specific Aβ peptides in CSF or plasma of these patients could improve the diagnosis of CAA and their inclusion in anti-amyloid immunotherapy treatments.
Collapse
Affiliation(s)
- Amal Kasri
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Elena Camporesi
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Eleni Gkanatsiou
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Susana Boluda
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Department of Neuropathology Raymond Escourolle, AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Gunnar Brinkmalm
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Lev Stimmer
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Junyue Ge
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jörg Hanrieder
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Nicolas Villain
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Charles Duyckaerts
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Department of Neuropathology Raymond Escourolle, AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Yannick Vermeiren
- Department of Biomedical Sciences, Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Division of Human Nutrition and Health, Chair Group Nutritional Biology, Wageningen University and Research (WUR), Wageningen, The Netherlands
| | - Sarah E Pape
- Institute of Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London, UK
| | - Gaël Nicolas
- Department of Genetics, CNRMAJ, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, F-76000, Rouen, France
| | - Annie Laquerrière
- Department of Pathology, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, F-76000, Rouen, France
| | - Peter Paul De Deyn
- Department of Biomedical Sciences, Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - David Wallon
- Department of Neurology, CNRMAJ, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, 76000, Rouen, France
| | - Kaj Blennow
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, Department of Neurology, Institute On Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People's Republic of China
| | - Andre Strydom
- Institute of Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London, UK
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
- Department of Neurology and Alzheimer Center, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.
- UK Dementia Research Institute at UCL, London, UK.
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China.
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA.
| | - Marie-Claude Potier
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France.
| |
Collapse
|
12
|
Bonanni R, Cariati I, Cifelli P, Frank C, Annino G, Tancredi V, D'Arcangelo G. Exercise to Counteract Alzheimer's Disease: What Do Fluid Biomarkers Say? Int J Mol Sci 2024; 25:6951. [PMID: 39000060 PMCID: PMC11241657 DOI: 10.3390/ijms25136951] [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: 05/14/2024] [Revised: 06/14/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Neurodegenerative diseases (NDs) represent an unsolved problem to date with an ever-increasing population incidence. Particularly, Alzheimer's disease (AD) is the most widespread ND characterized by an accumulation of amyloid aggregates of beta-amyloid (Aβ) and Tau proteins that lead to neuronal death and subsequent cognitive decline. Although neuroimaging techniques are needed to diagnose AD, the investigation of biomarkers within body fluids could provide important information on neurodegeneration. Indeed, as there is no definitive solution for AD, the monitoring of these biomarkers is of strategic importance as they are useful for both diagnosing AD and assessing the progression of the neurodegenerative state. In this context, exercise is known to be an effective non-pharmacological management strategy for AD that can counteract cognitive decline and neurodegeneration. However, investigation of the concentration of fluid biomarkers in AD patients undergoing exercise protocols has led to unclear and often conflicting results, suggesting the need to clarify the role of exercise in modulating fluid biomarkers in AD. Therefore, this critical literature review aims to gather evidence on the main fluid biomarkers of AD and the modulatory effects of exercise to clarify the efficacy and usefulness of this non-pharmacological strategy in counteracting neurodegeneration in AD.
Collapse
Affiliation(s)
- Roberto Bonanni
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Ida Cariati
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Pierangelo Cifelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Claudio Frank
- UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Giuseppe Annino
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Sports Engineering Laboratory, Department of Industrial Engineering, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Virginia Tancredi
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Giovanna D'Arcangelo
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| |
Collapse
|
13
|
Lopez E, Etxebarria-Elezgarai J, García-Sebastián M, Altuna M, Ecay-Torres M, Estanga A, Tainta M, López C, Martínez-Lage P, Amigo JM, Seifert A. Unlocking Preclinical Alzheimer's: A Multi-Year Label-Free In Vitro Raman Spectroscopy Study Empowered by Chemometrics. Int J Mol Sci 2024; 25:4737. [PMID: 38731955 PMCID: PMC11084676 DOI: 10.3390/ijms25094737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder, the early detection of which is crucial for timely intervention and enrollment in clinical trials. However, the preclinical diagnosis of Alzheimer's encounters difficulties with gold-standard methods. The current definitive diagnosis of Alzheimer's still relies on expensive instrumentation and post-mortem histological examinations. Here, we explore label-free Raman spectroscopy with machine learning as an alternative to preclinical Alzheimer's diagnosis. A special feature of this study is the inclusion of patient samples from different cohorts, sampled and measured in different years. To develop reliable classification models, partial least squares discriminant analysis in combination with variable selection methods identified discriminative molecules, including nucleic acids, amino acids, proteins, and carbohydrates such as taurine/hypotaurine and guanine, when applied to Raman spectra taken from dried samples of cerebrospinal fluid. The robustness of the model is remarkable, as the discriminative molecules could be identified in different cohorts and years. A unified model notably classifies preclinical Alzheimer's, which is particularly surprising because of Raman spectroscopy's high sensitivity regarding different measurement conditions. The presented results demonstrate the capability of Raman spectroscopy to detect preclinical Alzheimer's disease for the first time and offer invaluable opportunities for future clinical applications and diagnostic methods.
Collapse
Affiliation(s)
- Eneko Lopez
- CIC nanoGUNE BRTA, 20018 San Sebasián, Spain; (E.L.); (J.E.-E.)
- Department of Physics, University of the Basque Country (UPV/EHU), 20018 San Sebastián, Spain
| | | | - Maite García-Sebastián
- Center for Research and Advanced Therapies, CITA-Alzhéimer Foundation, 20009 San Sebastián, Spain; (M.G.-S.); (M.A.); (M.E.-T.); (A.E.); (M.T.); (C.L.); (P.M.-L.)
| | - Miren Altuna
- Center for Research and Advanced Therapies, CITA-Alzhéimer Foundation, 20009 San Sebastián, Spain; (M.G.-S.); (M.A.); (M.E.-T.); (A.E.); (M.T.); (C.L.); (P.M.-L.)
| | - Mirian Ecay-Torres
- Center for Research and Advanced Therapies, CITA-Alzhéimer Foundation, 20009 San Sebastián, Spain; (M.G.-S.); (M.A.); (M.E.-T.); (A.E.); (M.T.); (C.L.); (P.M.-L.)
| | - Ainara Estanga
- Center for Research and Advanced Therapies, CITA-Alzhéimer Foundation, 20009 San Sebastián, Spain; (M.G.-S.); (M.A.); (M.E.-T.); (A.E.); (M.T.); (C.L.); (P.M.-L.)
| | - Mikel Tainta
- Center for Research and Advanced Therapies, CITA-Alzhéimer Foundation, 20009 San Sebastián, Spain; (M.G.-S.); (M.A.); (M.E.-T.); (A.E.); (M.T.); (C.L.); (P.M.-L.)
| | - Carolina López
- Center for Research and Advanced Therapies, CITA-Alzhéimer Foundation, 20009 San Sebastián, Spain; (M.G.-S.); (M.A.); (M.E.-T.); (A.E.); (M.T.); (C.L.); (P.M.-L.)
| | - Pablo Martínez-Lage
- Center for Research and Advanced Therapies, CITA-Alzhéimer Foundation, 20009 San Sebastián, Spain; (M.G.-S.); (M.A.); (M.E.-T.); (A.E.); (M.T.); (C.L.); (P.M.-L.)
| | - Jose Manuel Amigo
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
- Department of Analytical Chemistry, University of the Basque Country, 48940 Leioa, Spain
| | - Andreas Seifert
- CIC nanoGUNE BRTA, 20018 San Sebasián, Spain; (E.L.); (J.E.-E.)
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
14
|
Lista S, Mapstone M, Caraci F, Emanuele E, López-Ortiz S, Martín-Hernández J, Triaca V, Imbimbo C, Gabelle A, Mielke MM, Nisticò R, Santos-Lozano A, Imbimbo BP. A critical appraisal of blood-based biomarkers for Alzheimer's disease. Ageing Res Rev 2024; 96:102290. [PMID: 38580173 DOI: 10.1016/j.arr.2024.102290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/18/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024]
Abstract
Biomarkers that predict the clinical onset of Alzheimer's disease (AD) enable the identification of individuals in the early, preclinical stages of the disease. Detecting AD at this point may allow for more effective therapeutic interventions and optimized enrollment for clinical trials of novel drugs. The current biological diagnosis of AD is based on the AT(N) classification system with the measurement of brain deposition of amyloid-β (Aβ) ("A"), tau pathology ("T"), and neurodegeneration ("N"). Diagnostic cut-offs for Aβ1-42, the Aβ1-42/Aβ1-40 ratio, tau and hyperphosphorylated-tau concentrations in cerebrospinal fluid have been defined and may support AD clinical diagnosis. Blood-based biomarkers of the AT(N) categories have been described in the AD continuum. Cross-sectional and longitudinal studies have shown that the combination of blood biomarkers tracking neuroaxonal injury (neurofilament light chain) and neuroinflammatory pathways (glial fibrillary acidic protein) enhance sensitivity and specificity of AD clinical diagnosis and improve the prediction of AD onset. However, no international accepted cut-offs have been identified for these blood biomarkers. A kit for blood Aβ1-42/Aβ1-40 is commercially available in the U.S.; however, it does not provide a diagnosis, but simply estimates the risk of developing AD. Although blood-based AD biomarkers have a great potential in the diagnostic work-up of AD, they are not ready for the routine clinical use.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Mark Mapstone
- Department of Neurology, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA.
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania 95125, Italy; Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, Troina 94018, Italy.
| | | | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Juan Martín-Hernández
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Rome 00015, Italy.
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy.
| | - Audrey Gabelle
- Memory Resources and Research Center, Montpellier University of Excellence i-site, Montpellier 34295, France.
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA.
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", Rome 00133, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome 00143, Italy.
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain; Physical Activity and Health Research Group (PaHerg), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid 28041, Spain.
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma 43122, Italy.
| |
Collapse
|
15
|
Geng J, Zhang Y, Chen H, Shi H, Wu Z, Chen J, Luo F. Associations between Alzheimer's disease biomarkers and postoperative delirium or cognitive dysfunction: A meta-analysis and trial sequential analysis of prospective clinical trials. Eur J Anaesthesiol 2024; 41:234-244. [PMID: 38038408 PMCID: PMC10842675 DOI: 10.1097/eja.0000000000001933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
BACKGROUND The relationship between Alzheimer's disease biomarkers and postoperative complications, such as postoperative delirium (POD) and postoperative cognitive dysfunction (POCD), remains a subject of ongoing debate. OBJECTIVE This meta-analysis aimed to determine whether there is an association between perioperative Alzheimer's disease biomarkers and postoperative complications. DESIGN We conducted a meta-analysis of observational clinical studies that explored the correlation between Alzheimer's disease biomarkers and POD or POCD in patients who have undergone surgery, following PRISMA guidelines. The protocol was previously published (INPLASY: INPLASY202350001). DATA SOURCES A comprehensive search was conducted across PubMed, Embase, Web of Science, and Cochrane databases until March 2023. ELIGIBILITY CRITERIA Surgical patients aged at least 18 years, studies focusing on POD or POCD, research involving Alzheimer's disease biomarkers, including Aβ or tau in blood or cerebrospinal fluid (CSF), and availability of the full text. RESULTS Our meta-analysis included 15 studies: six focusing on POD and nine on POCD. The findings revealed a negative correlation between preoperative CSF β-amyloid 42 (Aβ42) levels and the onset of POD [mean difference -86.1, 95% confidence interval (CI), -114.15 to -58.05, I2 : 47%]; this association was strongly supported by trial sequential analysis (TSA). A similar negative correlation was discerned between preoperative CSF Aβ42 levels and the incidence of POCD (-165.01, 95% CI, -261.48 to -68.53, I2 : 95%). The TSA also provided robust evidence for this finding; however, the evidence remains insufficient to confirm a relationship between other Alzheimer's disease biomarkers [β-amyloid 40 (Aβ40), total tau (T-tau), phosphorylated tau (P-tau), and Aβ42/T-tau ratio] and POD or POCD. CONCLUSION The study results indicate a negative correlation between preoperative CSF Aβ42 levels and the occurrence of both POD and POCD. Future investigations are warranted to identify the predictive cutoff value of preoperative CSF Aβ42 for POD and POCD.
Collapse
Affiliation(s)
- Jun Geng
- From the Department of Anaesthesiology, Jiangyin Hospital Affiliated to Nantong University, Wuxi City, Jiangsu Province, China (JG, YZ, HC, HH, ZW, JC) and Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Gongshu District, Hangzhou City, Zhejiang Province, China (FL)
| | | | | | | | | | | | | |
Collapse
|
16
|
Moradi E, Prakash M, Hall A, Solomon A, Strange B, Tohka J. Machine learning prediction of future amyloid beta positivity in amyloid-negative individuals. Alzheimers Res Ther 2024; 16:46. [PMID: 38414035 PMCID: PMC10900722 DOI: 10.1186/s13195-024-01415-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 02/11/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND The pathophysiology of Alzheimer's disease (AD) involves β -amyloid (A β ) accumulation. Early identification of individuals with abnormal β -amyloid levels is crucial, but A β quantification with positron emission tomography (PET) and cerebrospinal fluid (CSF) is invasive and expensive. METHODS We propose a machine learning framework using standard non-invasive (MRI, demographics, APOE, neuropsychology) measures to predict future A β -positivity in A β -negative individuals. We separately study A β -positivity defined by PET and CSF. RESULTS Cross-validated AUC for 4-year A β conversion prediction was 0.78 for the CSF-based and 0.68 for the PET-based A β definitions. Although not trained for the clinical status-change prediction, the CSF-based model excelled in predicting future mild cognitive impairment (MCI)/dementia conversion in cognitively normal/MCI individuals (AUCs, respectively, 0.76 and 0.89 with a separate dataset). CONCLUSION Standard measures have potential in detecting future A β -positivity and assessing conversion risk, even in cognitively normal individuals. The CSF-based definition led to better predictions than the PET-based definition.
Collapse
Affiliation(s)
- Elaheh Moradi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, 70150, Finland.
| | - Mithilesh Prakash
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, 70150, Finland
| | - Anette Hall
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
- Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institute, Stockholm, Sweden
| | - Alina Solomon
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
- Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institute, Stockholm, Sweden
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | - Bryan Strange
- Laboratory for Clinical Neuroscience, Center for Biomedical Technology, Universidad Politécnica de Madrid, IdISSC, Madrid, Spain
- Reina Sofia Centre for Alzheimer's Research, Madrid, Spain
| | - Jussi Tohka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, 70150, Finland
| |
Collapse
|
17
|
Twait EL, Wu JH, Kamarioti M, Basten M, van der Flier WM, Gerritsen L, Geerlings MI. Association of amyloid-beta with depression or depressive symptoms in older adults without dementia: a systematic review and meta-analysis. Transl Psychiatry 2024; 14:25. [PMID: 38225253 PMCID: PMC10789765 DOI: 10.1038/s41398-024-02739-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/05/2023] [Accepted: 01/05/2024] [Indexed: 01/17/2024] Open
Abstract
Several lines of evidence have indicated that depression might be a prodromal symptom of Alzheimer's disease (AD). This systematic review and meta-analysis investigated the cross-sectional association between amyloid-beta, one of the key pathologies defining AD, and depression or depressive symptoms in older adults without dementia. A systematic search in PubMed yielded 689 peer-reviewed articles. After full-text screening, nine CSF studies, 11 PET studies, and five plasma studies were included. No association between amyloid-beta and depression or depressive symptoms were found using cerebrospinal fluid (CSF) (0.15; 95% CI: -0.08; 0.37), positron emission topography (PET) (Cohen's d: 0.09; 95% CI: -0.05; 0.24), or plasma (-0.01; 95% CI: -0.23; 0.22). However, subgroup analyses revealed an association in plasma studies of individuals with cognitive impairment. A trend of an association was found in the studies using CSF and PET. This systematic review and meta-analysis suggested that depressive symptoms may be part of the prodromal stage of dementia.
Collapse
Affiliation(s)
- Emma L Twait
- Amsterdam UMC, location Vrije Universiteit, Department of General Practice, Van der Boechorststraat 7, Amsterdam, The Netherlands
- Amsterdam Public Health; Aging & Later life, and Personalized Medicine, Amsterdam, The Netherlands
- Amsterdam Neuroscience; Neurodegeneration, and Mood, Anxiety, Psychosis, Stress, and Sleep, Amsterdam, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Jen-Hao Wu
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Maria Kamarioti
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Maartje Basten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Lotte Gerritsen
- Department of Psychology, Utrecht University, Utrecht, The Netherlands
| | - Mirjam I Geerlings
- Amsterdam UMC, location Vrije Universiteit, Department of General Practice, Van der Boechorststraat 7, Amsterdam, The Netherlands.
- Amsterdam Neuroscience; Neurodegeneration, and Mood, Anxiety, Psychosis, Stress, and Sleep, Amsterdam, The Netherlands.
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands.
- Amsterdam UMC, location University of Amsterdam, Department of General Practice, Meibergdreef 9, Amsterdam, The Netherlands.
| |
Collapse
|
18
|
Lojo-Ramírez JA, Guerra-Gómez M, Marín-Cabañas AM, Fernández-Rodríguez P, Bernal Sánchez-Arjona M, Franco-Macías E, García-Solís D. Correlation Between Amyloid PET Imaging and Discordant Cerebrospinal Fluid Biomarkers Results in Patients with Suspected Alzheimer's Disease. J Alzheimers Dis 2024; 97:447-458. [PMID: 38143353 DOI: 10.3233/jad-230744] [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: 12/26/2023]
Abstract
BACKGROUND Although the concordance between cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers and amyloid-PET findings is well known, there are no data regarding the concordance of amyloid-PET with inconclusive CSF values of amyloid-β (Aβ)1 - 42 and p-tau for the diagnosis of AD. OBJECTIVE To investigate the relationship between the amyloid-PET results with discordant AD biomarkers values in CSF (Aβ1 - 42+/p-tau-or Aβ1 - 42-/p-tau+). METHODS An observational retrospective study, including 62 patients with mild cognitive impairment (32/62) or dementia (30/62), suspicious of AD who had undergone a lumbar puncture to determine CSF AD biomarkers, and presented discordant values in CSF between Aβ1 - 42 and p-tau (Aβ1 - 42+/p-tau-or Aβ1 - 42-/p-tau+). All of them, underwent an amyloid-PET with 18F-Florbetaben. An extensive neuropsychological testing as part of their diagnostic process (MMSE and TMA-93), was performed, and it was also obtained the Global Deterioration Scale. RESULTS Comparing the discordant CSF results of each patient with the cerebral amyloid-PET results, we found that in the group with Aβ1 - 42+ and p-tau-CSF values, the amyloid-PET was positive in 51.2% and negative in 48.8% of patients, while in the group with Aβ1 - 42-and p-Tau+ CSF values, the amyloid-PET was positive in 52.6% of patients and negative in 47.4% of them. No significant association was found (p = 0.951) between the results of amyloid-PET and the two divergent groups in CSF. CONCLUSIONS No significant relationship was observed between the results of discordant AD biomarkers in CSF and the result of amyloid-PET. No trend in amyloid-PET results was observed in relation to CSF biomarker values.
Collapse
Affiliation(s)
| | - Miriam Guerra-Gómez
- Department of Nuclear Medicine, Virgen del Rocío University Hospital, Seville, Spain
| | | | | | | | - Emilio Franco-Macías
- Memory Unit, Department of Neurology, Virgen del Rocío University Hospital, Seville, Spain
| | - David García-Solís
- Department of Nuclear Medicine, Virgen del Rocío University Hospital, Seville, Spain
| |
Collapse
|
19
|
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
|
20
|
Liu X, Chen J, Meng C, Zhou L, Liu Y. Serum neurofilament light chain and cognition decline in US elderly: A cross-sectional study. Ann Clin Transl Neurol 2024; 11:17-29. [PMID: 37902309 PMCID: PMC10791034 DOI: 10.1002/acn3.51929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023] Open
Abstract
OBJECTIVE Early identification of cognitive impairment in neurodegenerative diseases like Alzheimer's disease (AD) is crucial. Neurofilament, a potential biomarker for neurological disorders, has gained attention. Our study aims to investigate the relationship between serum neurofilament light (sNfL) levels and cognitive function in elderly individuals in the United States. METHODS This cross-sectional study analyzed data from participants aged 60 and above in the National Health and Nutrition Examination Survey (2013-2014). We collected sNfL levels, cognitive function tests, sociodemographic characteristics, comorbidities, and other variables. Weighted multiple linear regression models examined the relationship between ln(sNfL) and cognitive scores. Restricted cubic spline (RCS) visualization explored nonlinear relationships. The stratified analysis examined subgroups' ln(sNfL) and cognitive function association. RESULTS The study included 446 participants (47.73% male). Participants with ln(sNfL) levels between 2.58 and 2.81 pg/mL (second quintile) performed relatively well in cognitive tests. After adjusting for multiple factors, ln(sNfL) levels were negatively correlated with cognitive function, with adjusted β (95% CI) as follows: immediate recall test (IRT): -0.763 (-1.301 to -0.224), delayed recall test (DRT): -0.308 (-0.576 to -0.04), animal fluency test (AFT): -1.616 (-2.639 to -0.594), and digit symbol substitution test (DSST): -2.790 (-4.369 to -1.21). RCS curves showed nonlinear relationships between ln(sNfL) and DRT, AFT, with inflection points around 2.7 pg/mL. The stratified analysis revealed a negative correlation between ln(sNfL) and cognition in specific subgroups with distinct features, with an interaction between diabetes and ln(sNfL). INTERPRETATION Higher sNfL levels are associated with poorer cognitive function in the elderly population of the United States. sNfL shows promise as a potential biomarker for early identification of cognitive decline.
Collapse
Affiliation(s)
- Xiaodong Liu
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| | - Jun Chen
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| | - Chen Meng
- Department of Anesthesiology, Taihe HospitalHubei University of MedicineShiyanHubeiChina
| | - Lan Zhou
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| | - Yong Liu
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| |
Collapse
|
21
|
Singh S, Mahajan M, Kumar D, Singh K, Chowdhary M, Amit. An inclusive study of recent advancements in Alzheimer's disease: A comprehensive review. Neuropeptides 2023; 102:102369. [PMID: 37611472 DOI: 10.1016/j.npep.2023.102369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023]
Abstract
Alzheimer's disease (AD) has remained elusive in revealing its pathophysiology and mechanism of development. In this review paper, we attempt to highlight several theories that abound about the exact pathway of AD development. The number of cases worldwide has prompted a constant flow of research to detect high-risk patients, slow the progression of the disease and discover improved methods of treatment that may prove effective. We shall focus on the two main classes of drugs that are currently in use; and emerging ones with novel mechanisms that are under development. As of late there has also been increased attention towards factors that were previously thought to be unrelated to AD, such as the gut microbiome, lifestyle habits, and diet. Studies have now shown that all these factors make an impact on AD progression, thus bringing to our attention more areas that could hold the key to combating this disease. This paper covers all the aforementioned factors concisely. We also briefly explore the relationship between mental health and AD, both before and after the diagnosis of the disease.
Collapse
Affiliation(s)
- Sukanya Singh
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Mitali Mahajan
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Dhawal Kumar
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Kunika Singh
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Mehvish Chowdhary
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Amit
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India.
| |
Collapse
|
22
|
Guillén N, Contador J, Buongiorno M, Álvarez I, Culell N, Alcolea D, Lleó A, Fortea J, Piñol-Ripoll G, Carnes-Vendrell A, Lourdes Ispierto M, Vilas D, Puig-Pijoan A, Fernández-Lebrero A, Balasa M, Sánchez-Valle R, Lladó A. Agreement of cerebrospinal fluid biomarkers and amyloid-PET in a multicenter study. Eur Arch Psychiatry Clin Neurosci 2023:10.1007/s00406-023-01701-y. [PMID: 37898567 DOI: 10.1007/s00406-023-01701-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 10/02/2023] [Indexed: 10/30/2023]
Abstract
Core Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarkers have shown incomplete agreement with amyloid-positron emission tomography (PET). Our goal was to analyze the agreement between AD CSF biomarkers and amyloid-PET in a multicenter study. Retrospective multicenter study (5 centers). Participants who underwent both CSF biomarkers and amyloid-PET scan within 18 months were included. Clinical diagnoses were made according to latest diagnostic criteria by the attending clinicians. CSF Amyloid Beta1-42 (Aβ1-42, A), phosphorliated tau 181 (pTau181, T) and total tau (tTau, N) biomarkers were considered normal (-) or abnormal ( +) according to cutoffs of each center. Amyloid-PET was visually classified as positive/negative. Agreement between CSF biomarkers and amyloid-PET was analyzed by overall percent agreement (OPA). 236 participants were included (mean age 67.9 years (SD 9.1), MMSE score 24.5 (SD 4.1)). Diagnoses were mild cognitive impairment or dementia due to AD (49%), Lewy body dementia (22%), frontotemporal dementia (10%) and others (19%). Mean time between tests was 5.1 months (SD 4.1). OPA between single CSF biomarkers and amyloid-PET was 74% for Aβ1-42, 75% for pTau181, 73% for tTau. The use of biomarker ratios improved OPA: 87% for Aβ1-42/Aβ1-40 (n = 155), 88% for pTau181/Aβ1-42 (n = 94) and 82% for tTau/Aβ1-42 (n = 160). A + T + N + cases showed the highest agreement between CSF biomarkers and amyloid-PET (96%), followed by A-T-N- cases (89%). Aβ1-42/Aβ1-40 was a better marker of cerebral amyloid deposition, as identified by amyloid tracers, than Aβ1-42 alone. Combined biomarkers in CSF predicted amyloid-PET result better than single biomarkers.
Collapse
Affiliation(s)
- Núria Guillén
- Alzheimer's Disease and Other Cognitive Disorders Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Carrer Villarroel, 170, 08036, Barcelona, Spain
| | - José Contador
- Alzheimer's Disease and Other Cognitive Disorders Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Carrer Villarroel, 170, 08036, Barcelona, Spain
| | - Mariateresa Buongiorno
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació Docència i Recerca Mútua Terrassa, Terrassa, Spain
| | - Ignacio Álvarez
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació Docència i Recerca Mútua Terrassa, Terrassa, Spain
| | - Natalia Culell
- Memory Disorders Unit, Department of Neurology, Hospital Universitari Mutua de Terrassa, Terrassa, Spain
- Fundació Docència i Recerca Mútua Terrassa, Terrassa, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau-Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau-Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau-Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Madrid, Spain
| | - Gerard Piñol-Ripoll
- Clinical Neuroscience Research, Unitat Trastorns Cognitius, IRBLleida, Santa Maria University Hospital, Lleida, Spain
| | - Anna Carnes-Vendrell
- Clinical Neuroscience Research, Unitat Trastorns Cognitius, IRBLleida, Santa Maria University Hospital, Lleida, Spain
| | - María Lourdes Ispierto
- Neurodegenerative Diseases Unit, Neurology Service and Neurosciences Department, University Hospital Germans Trias i Pujol (HUGTP), Badalona, Spain
| | - Dolores Vilas
- Neurodegenerative Diseases Unit, Neurology Service and Neurosciences Department, University Hospital Germans Trias i Pujol (HUGTP), Badalona, Spain
| | - Albert Puig-Pijoan
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Aida Fernández-Lebrero
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Carrer Villarroel, 170, 08036, Barcelona, Spain
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Carrer Villarroel, 170, 08036, Barcelona, Spain
- Institute of Neurosciences, Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Carrer Villarroel, 170, 08036, Barcelona, Spain.
- Institute of Neurosciences, Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
23
|
Yuan Y, Chen L, Kong L, Qiu L, Fu Z, Sun M, Liu Y, Cheng M, Ma S, Wang X, Zhao C, Jiang J, Zhang X, Wang L, Gao L. Histidine modulates amyloid-like assembly of peptide nanomaterials and confers enzyme-like activity. Nat Commun 2023; 14:5808. [PMID: 37726302 PMCID: PMC10509148 DOI: 10.1038/s41467-023-41591-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
Amyloid-like assembly is not only associated with pathological events, but also leads to the development of novel nanomaterials with unique properties. Herein, using Fmoc diphenylalanine peptide (Fmoc-F-F) as a minimalistic model, we found that histidine can modulate the assembly behavior of Fmoc-F-F and induce enzyme-like catalysis. Specifically, the presence of histidine rearranges the β structure of Fmoc-F-F to assemble nanofilaments, resulting in the formation of active site to mimic peroxidase-like activity that catalyzes ROS generation. A similar catalytic property is also observed in Aβ assembled filaments, which is correlated with the spatial proximity between intermolecular histidine and F-F. Notably, the assembled Aβ filaments are able to induce cellular ROS elevation and damage neuron cells, providing an insight into the pathological relationship between Aβ aggregation and Alzheimer's disease. These findings highlight the potential of histidine as a modulator in amyloid-like assembly of peptide nanomaterials exerting enzyme-like catalysis.
Collapse
Affiliation(s)
- Ye Yuan
- Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, 130012, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Lei Chen
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lingfei Kong
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lingling Qiu
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, China
| | - Zhendong Fu
- Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Minmin Sun
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuan Liu
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Miaomiao Cheng
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Saiyu Ma
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaonan Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Changhui Zhao
- Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Jing Jiang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xinzheng Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liping Wang
- Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, 130012, China.
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
24
|
Surpi A, Murgia M, López-Amoedo S, González-Gómez MA, Piñeiro Y, Rivas J, Perugini V, Santin M, Sobrino T, Greco P, Campos F, Dediu VA. Magnetic separation and concentration of Aβ 1-42 molecules dispersed at the threshold concentration for Alzheimer's disease diagnosis in clinically-relevant volumes of sample. J Nanobiotechnology 2023; 21:329. [PMID: 37710290 PMCID: PMC10503095 DOI: 10.1186/s12951-023-02095-8] [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: 04/18/2023] [Accepted: 09/02/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of dementia and loss of autonomy in the elderly, implying a progressive cognitive decline and limitation of social activities. The progressive aging of the population is expected to exacerbate this problem in the next decades. Therefore, there is an urgent need to develop quantitative diagnostic methodologies to assess the onset the disease and its progression especially in the initial phases. RESULTS Here we describe a novel technology to extract one of the most important molecular biomarkers of AD (Aβ1-42) from a clinically-relevant volume - 100 µl - therein dispersed in a range of concentrations critical for AD early diagnosis. We demonstrate that it is possible to immunocapture Aβ1-42 on 20 nm wide magnetic nanoparticles functionalized with hyperbranced KVLFF aptamers. Then, it is possible to transport them through microfluidic environments to a detection system where virtually all (~ 90%) the Aβ1-42 molecules are concentrated in a dense plug of ca.50 nl. The technology is based on magnetic actuation by permanent magnets, specifically designed to generate high gradient magnetic fields. These fields, applied through submillimeter-wide channels, can concentrate, and confine magnetic nanoparticles (MNPs) into a droplet with an optimized shape that maximizes the probability of capturing highly diluted molecular biomarkers. These advancements are expected to provide efficient protocols for the concentration and manipulation of molecular biomarkers from clinical samples, enhancing the accuracy and the sensitivity of diagnostic technologies. CONCLUSIONS This easy to automate technology allows an efficient separation of AD molecular biomarkers from volumes of biological solutions complying with the current clinical protocols and, ultimately, leads to accurate measurements of biomarkers. The technology paves a new way for a quantitative AD diagnosis at the earliest stage and it is also adaptable for the biomarker analysis of other pathologies.
Collapse
Affiliation(s)
- Alessandro Surpi
- Istituto per lo Studio dei Materiali Nanostrutturati (CNR-ISMN), Bologna, 40129, Italy.
- Istituto per la Microelettronica e i Microsistemi, IMM-CNR, 40129, Bologna, Italy.
| | - Mauro Murgia
- Istituto per lo Studio dei Materiali Nanostrutturati (CNR-ISMN), Bologna, 40129, Italy
- Center for Translational Neurophysiology (IIT), Italian Institute of Technology, Ferrara, 44121, Italy
| | - Sonia López-Amoedo
- Translational Stroke Laboratory (TREAT), Clinical Neurosciences Research Laboratory (LINC) , Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15782, Spain
| | - Manuel A González-Gómez
- NANOMAG Laboratory, Applied Physics Department, iMATUS Materials Institute, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Yolanda Piñeiro
- NANOMAG Laboratory, Applied Physics Department, iMATUS Materials Institute, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - José Rivas
- NANOMAG Laboratory, Applied Physics Department, iMATUS Materials Institute, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Valeria Perugini
- Centre for Regenerative Medicine and Devices, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Matteo Santin
- Centre for Regenerative Medicine and Devices, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Tomás Sobrino
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, 28029, Spain
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, Spain
| | - Pierpaolo Greco
- Center for Translational Neurophysiology (IIT), Italian Institute of Technology, Ferrara, 44121, Italy
- Dipartimento di Neuroscienze e Riabilitazione, Università di Ferrara, Ferrara, 44121, Italy
| | - Francisco Campos
- Translational Stroke Laboratory (TREAT), Clinical Neurosciences Research Laboratory (LINC) , Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15782, Spain.
| | - Valentin Alek Dediu
- Istituto per lo Studio dei Materiali Nanostrutturati (CNR-ISMN), Bologna, 40129, Italy.
| |
Collapse
|
25
|
Haque R, Watson CM, Liu J, Carter EK, Duong DM, Lah JJ, Wingo AP, Roberts BR, Johnson EC, Saykin AJ, Shaw LM, Seyfried NT, Wingo TS, Levey AI. A protein panel in cerebrospinal fluid for diagnostic and predictive assessment of Alzheimer's disease. Sci Transl Med 2023; 15:eadg4122. [PMID: 37672565 PMCID: PMC10880442 DOI: 10.1126/scitranslmed.adg4122] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 08/17/2023] [Indexed: 09/08/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with heterogenous pathophysiological changes that develop years before the onset of clinical symptoms. These preclinical changes have generated considerable interest in identifying markers for the pathophysiological mechanisms linked to AD and AD-related disorders (ADRD). On the basis of our prior work integrating cerebrospinal fluid (CSF) and brain proteome networks, we developed a reliable and high-throughput mass spectrometry-selected reaction monitoring assay that targets 48 key proteins altered in CSF. To test the diagnostic utility of these proteins and compare them with existing AD biomarkers, CSF collected at baseline visits was assayed from 706 participants recruited from the Alzheimer's Disease Neuroimaging Initiative. We found that the targeted CSF panel of 48 proteins (CSF 48 panel) performed at least as well as existing AD CSF biomarkers (Aβ42, tTau, and pTau181) for predicting clinical diagnosis, FDG PET, hippocampal volume, and measures of cognitive and dementia severity. In addition, for each of those outcomes, the CSF 48 panel plus the existing AD CSF biomarkers significantly improved diagnostic performance. Furthermore, the CSF 48 panel plus existing AD CSF biomarkers significantly improved predictions for changes in FDG PET, hippocampal volume, and measures of cognitive decline and dementia severity compared with either measure alone. A potential reason for these improvements is that the CSF 48 panel reflects a range of altered biology observed in AD/ADRD. In conclusion, we show that the CSF 48 panel complements existing AD CSF biomarkers to improve diagnosis and predict future cognitive decline and dementia severity.
Collapse
Affiliation(s)
- Rafi Haque
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA, 30329
| | - Caroline M. Watson
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA, 30329
| | - Jiaqi Liu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA, 30329
| | - E. Kathleen Carter
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA, 30322
| | - Duc M. Duong
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA, 30322
| | - James J. Lah
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA, 30329
| | - Aliza P. Wingo
- Division of Mental Health, Atlanta VA Medical Center, Decatur, GA, USA, 30033
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA, 30329
| | - Blaine R. Roberts
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA, 30322
| | - Erik C.B. Johnson
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA, 30322
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA, 46204
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA, 19104
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA, 19104
| | - Nicholas T. Seyfried
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA, 30322
| | - Thomas S. Wingo
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA, 30322
| | - Allan I. Levey
- Goizueta Alzheimer’s Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA, 30329
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA, 30329
| |
Collapse
|
26
|
Cozachenco D, Zimmer ER, Lourenco MV. Emerging concepts towards a translational framework in Alzheimer's disease. Neurosci Biobehav Rev 2023; 152:105246. [PMID: 37236385 DOI: 10.1016/j.neubiorev.2023.105246] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
Over the past decades, significant efforts have been made to understand the precise mechanisms underlying the pathogenesis of Alzheimer's disease (AD), the most common cause of dementia. However, clinical trials targeting AD pathological hallmarks have consistently failed. Refinement of AD conceptualization, modeling, and assessment is key to developing successful therapies. Here, we review critical findings and discuss emerging ideas to integrate molecular mechanisms and clinical approaches in AD. We further propose a refined workflow for animal studies incorporating multimodal biomarkers used in clinical studies - delineating critical paths for drug discovery and translation. Addressing unresolved questions with the proposed conceptual and experimental framework may accelerate the development of effective disease-modifying strategies for AD.
Collapse
Affiliation(s)
- Danielle Cozachenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Eduardo R Zimmer
- Department of Pharmacology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Graduate Program in Biological Sciences: Biochemistry (PPGBioq), UFRGS, Porto Alegre, RS, Brazil; Pharmacology and Therapeutics (PPGFT), UFRGS, Porto Alegre, RS, Brazil; McGill Centre for Studies in Aging, McGill University, Montreal, Canada; Brain Institute of Rio Grande do Sul, PUCRS, Porto Alegre, Brazil.
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
27
|
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: 1.0] [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
|
28
|
Kapoor A, Gaubert A, Yew B, Jang JY, Dutt S, Li Y, Alitin JPM, Nguyen A, Ho JK, Blanken AE, Sible IJ, Marshall A, Shenasa F, Rodgers KE, Martini AC, Head E, Nation DA. Enlarged perivascular spaces and plasma Aβ42/Aβ40 ratio in older adults without dementia. Neurobiol Aging 2023; 128:43-48. [PMID: 37156179 PMCID: PMC10852216 DOI: 10.1016/j.neurobiolaging.2023.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 05/10/2023]
Abstract
Dilation of perivascular spaces (PVS) in the brain may indicate poor fluid drainage due to the accumulation of perivascular cell debris, waste, and proteins, including amyloid-beta (Aβ). No prior study has assessed whether plasma Aβ levels are related to PVS in older adults without dementia. Independently living older adults (N = 56, mean age = 68.2 years; Standard deviation (SD) = 6.5; 30.4% male) free of dementia or clinical stroke were recruited from the community and underwent brain MRI and venipuncture. PVS were qualitatively scored and dichotomized to low PVS burden (scores 0-1,) or high PVS burden (score>1). Plasma was assayed using a Quanterix Simoa Kit to quantify Aβ42 and Aβ40 levels. A significant difference was observed in plasma Aβ42/Aβ40 ratio between low and high PVS burden, controlling for age (F[1, 53] = 5.59, p = 0.022, η2 = 0.10), with lower Aβ42/Aβ40 ratio in the high PVS burden group. Dilation of PVS is associated with a lower plasma Aβ42/Aβ40 ratio, which may indicate higher cortical amyloid deposition. Future longitudinal studies examining PVS changes, and the pathogenesis of AD are warranted.
Collapse
Affiliation(s)
- Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Aimée Gaubert
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Belinda Yew
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Jung Yun Jang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Shubir Dutt
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Yanrong Li
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - John Paul M Alitin
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Amy Nguyen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Jean K Ho
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Anna E Blanken
- San Francisco Veterans Affairs Health Care System & Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Isabel J Sible
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Anisa Marshall
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Fatemah Shenasa
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Kathleen E Rodgers
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Alessandra C Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Daniel A Nation
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
29
|
Liu JJ, Long YF, Xu P, Guo HD, Cui GH. Pathogenesis of miR-155 on nonmodifiable and modifiable risk factors in Alzheimer's disease. Alzheimers Res Ther 2023; 15:122. [PMID: 37452431 PMCID: PMC10347850 DOI: 10.1186/s13195-023-01264-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/28/2023] [Indexed: 07/18/2023]
Abstract
Alzheimer's disease (AD) is a common age-related neurodegenerative disease in the central nervous system and is the primary cause of dementia. It is clinically characterized by the memory impairment, aphasia, apraxia, agnosia, visuospatial and executive dysfunction, behavioral changes, and so on. Incidence of this disease was bound up with age, genetic factors, cardiovascular and cerebrovascular dysfunction, and other basic diseases, but the exact etiology has not been clarified. MicroRNAs (miRNAs) are small endogenous non-coding RNAs that were involved in the regulation of post-transcriptional gene expression. miRNAs have been extensively studied as noninvasive potential biomarkers for disease due to their relative stability in bodily fluids. In addition, they play a significant role in the physiological and pathological processes of various neurological disorders, including stroke, AD, and Parkinson's disease. MiR-155, as an important pro-inflammatory mediator of neuroinflammation, was reported to participate in the progression of β-amyloid peptide and tau via regulating immunity and inflammation. In this review, we put emphasis on the effects of miR-155 on AD and explore the underlying biological mechanisms which could provide a novel approach for diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Jia-Jia Liu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yun-Fan Long
- Department of Neurology, Shanghai No. 9 People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Peng Xu
- Affiliated Hospital of Jining Medical University, Jining, 272000, Shandong, China.
| | - Hai-Dong Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Guo-Hong Cui
- Department of Neurology, Shanghai No. 9 People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
30
|
Twait EL, Min B, Beran M, Vonk JMJ, Geerlings MI. The cross-sectional association between amyloid burden and white matter hyperintensities in older adults without cognitive impairment: A systematic review and meta-analysis. Ageing Res Rev 2023; 88:101952. [PMID: 37178806 DOI: 10.1016/j.arr.2023.101952] [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: 01/04/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, characterized by the aggregation of amyloid-beta (Aβ) proteins into plaques. Individuals with AD frequently show mixed pathologies, often caused by cerebral small vessel disease (CSVD), resulting in lesions such as white matter hyperintensities (WMH). The current systematic review and meta-analysis investigated the cross-sectional relationship between amyloid burden and WMH in older adults without objective cognitive impairment. A systematic search performed in PubMed, Embase, and PsycINFO yielded 13 eligible studies. Aβ was assessed using PET, CSF, or plasma measurements. Two meta-analyses were performed: one on Cohen's d metrics and one on correlation coefficients. The meta-analyses revealed an overall weighted small-to-medium Cohen's d of 0.55 (95% CI: 0.31-0.78) in CSF, an overall correlation of 0.31 (0.09-0.50) in CSF, and a large Cohen's d of 0.96 (95% CI: 0.66-1.27) in PET. Only two studies assessed this relationship in plasma, with an effect size of - 0.20 (95% CI: -0.75 to 0.34). These findings indicate a relationship between both amyloid and vascular pathologies in cognitively normal adults in PET and CSF. Future studies should assess the possible relationship of blood amyloid-beta and WMH for broader identification of at risk individuals showing mixed pathology in preclinical stages.
Collapse
Affiliation(s)
- Emma L Twait
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Amsterdam UMC, Location Vrije Universiteit, Department of General Practice, Amsterdam, The Netherlands; Research Institute Amsterdam Public Health, Research Programme Aging & Later life, and Research Programme Personalized Medicine, Amsterdam, The Netherlands
| | - Britt Min
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Bachelor Program Biomedical Sciences, Faculty of Medicine, Utrecht University, Utrecht, The Netherlands
| | - Magdalena Beran
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; School for Cardiovascular Disease (CARIM), Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands
| | - Jet M J Vonk
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Neurology, Memory and Aging Center, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Mirjam I Geerlings
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Research Institute Amsterdam Public Health, Research Programme Aging & Later life, and Research Programme Personalized Medicine, Amsterdam, The Netherlands; Research Institute Amsterdam Neuroscience, Research Programme Neurodegeneration, and Research Programme Mood, Anxiety, Psychosis, Stress, and Sleep, Amsterdam, The Netherlands; Amsterdam UMC, location University of Amsterdam, Department of General Practice, Amsterdam, The Netherlands.
| |
Collapse
|
31
|
Zhang Y, Chen H, Li R, Sterling K, Song W. Amyloid β-based therapy for Alzheimer's disease: challenges, successes and future. Signal Transduct Target Ther 2023; 8:248. [PMID: 37386015 PMCID: PMC10310781 DOI: 10.1038/s41392-023-01484-7] [Citation(s) in RCA: 150] [Impact Index Per Article: 150.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 07/01/2023] Open
Abstract
Amyloid β protein (Aβ) is the main component of neuritic plaques in Alzheimer's disease (AD), and its accumulation has been considered as the molecular driver of Alzheimer's pathogenesis and progression. Aβ has been the prime target for the development of AD therapy. However, the repeated failures of Aβ-targeted clinical trials have cast considerable doubt on the amyloid cascade hypothesis and whether the development of Alzheimer's drug has followed the correct course. However, the recent successes of Aβ targeted trials have assuaged those doubts. In this review, we discussed the evolution of the amyloid cascade hypothesis over the last 30 years and summarized its application in Alzheimer's diagnosis and modification. In particular, we extensively discussed the pitfalls, promises and important unanswered questions regarding the current anti-Aβ therapy, as well as strategies for further study and development of more feasible Aβ-targeted approaches in the optimization of AD prevention and treatment.
Collapse
Affiliation(s)
- Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Huaqiu Chen
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ran Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China.
| |
Collapse
|
32
|
Dang C, Wang Y, Li Q, Lu Y. Neuroimaging modalities in the detection of Alzheimer's disease-associated biomarkers. PSYCHORADIOLOGY 2023; 3:kkad009. [PMID: 38666112 PMCID: PMC11003434 DOI: 10.1093/psyrad/kkad009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/04/2023] [Accepted: 06/20/2023] [Indexed: 04/28/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Neuropathological changes in AD patients occur up to 10-20 years before the emergence of clinical symptoms. Specific diagnosis and appropriate intervention strategies are crucial during the phase of mild cognitive impairment (MCI) and AD. The detection of biomarkers has emerged as a promising tool for tracking the efficacy of potential therapies, making an early disease diagnosis, and prejudging treatment prognosis. Specifically, multiple neuroimaging modalities, including magnetic resonance imaging (MRI), positron emission tomography, optical imaging, and single photon emission-computed tomography, have provided a few potential biomarkers for clinical application. The MRI modalities described in this review include structural MRI, functional MRI, diffusion tensor imaging, magnetic resonance spectroscopy, and arterial spin labelling. These techniques allow the detection of presymptomatic diagnostic biomarkers in the brains of cognitively normal elderly people and might also be used to monitor AD disease progression after the onset of clinical symptoms. This review highlights potential biomarkers, merits, and demerits of different neuroimaging modalities and their clinical value in MCI and AD patients. Further studies are necessary to explore more biomarkers and overcome the limitations of multiple neuroimaging modalities for inclusion in diagnostic criteria for AD.
Collapse
Affiliation(s)
- Chun Dang
- Department of Periodical Press, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yanchao Wang
- Department of Neurology, Chifeng University of Affiliated Hospital, Chifeng 024000, China
| | - Qian Li
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Yaoheng Lu
- Department of General Surgery, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Chengdu 610000, China
| |
Collapse
|
33
|
Mendes GO, Pita SSDR, Carvalho PBD, Silva MPD, Taranto AG, Leite FHA. Molecular Multi-Target Approach for Human Acetylcholinesterase, Butyrylcholinesterase and β-Secretase 1: Next Generation for Alzheimer's Disease Treatment. Pharmaceuticals (Basel) 2023; 16:880. [PMID: 37375827 DOI: 10.3390/ph16060880] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative condition characterized by progressive memory loss and other affected cognitive functions. Pharmacological therapy of AD relies on inhibitors of the enzymes acetylcholinesterase (AChE) and butyrylcholinesterase (BChE), offering only a palliative effect and being incapable of stopping or reversing the neurodegenerative process. However, recent studies have shown that inhibiting the enzyme β-secretase 1 (BACE-1) may be able to stop neurodegeneration, making it a promising target. Considering these three enzymatic targets, it becomes feasible to apply computational techniques to guide the identification and planning of molecules capable of binding to all of them. After virtually screening 2119 molecules from a library, 13 hybrids were built and further screened by triple pharmacophoric model, molecular docking, and molecular dynamics (t = 200 ns). The selected hybrid G meets all stereo-electronic requirements to bind to AChE, BChE, and BACE-1 and offers a promising structure for future synthesis, enzymatic testing, and validation.
Collapse
Affiliation(s)
- Géssica Oliveira Mendes
- Laboratory of Molecular Modeling, Department of Health, State University of Feira de Santana, Salvador 44036-900, BA, Brazil
- Postgraduate Program in Pharmaceutical Sciences, State University of Feira de Santana, Salvador 44036-900, BA, Brazil
| | - Samuel Silva da Rocha Pita
- Postgraduate Program in Pharmaceutical Sciences, State University of Feira de Santana, Salvador 44036-900, BA, Brazil
- Laboratory of Bioinformatics and Molecular Modeling (LaBiMM), Pharmacy College, Federal University of Bahia (UFBA), Salvador 40170-110, BA, Brazil
| | | | - Michel Pires da Silva
- Laboratory of Bioinformatics and Drug Design, Department of Bioengineering, Federal University of Sao Joao del-Rei, São João del-Rei 36301-1601, MG, Brazil
- Federal Center for Technological Education of Minas Gerais, Department of Informatics, Management and Design, R. Álvares de Azevedo, 400, Bela Vista, Divinópolis 35503-822, MG, Brazil
| | - Alex Gutterres Taranto
- Laboratory of Bioinformatics and Drug Design, Department of Bioengineering, Federal University of Sao Joao del-Rei, São João del-Rei 36301-1601, MG, Brazil
| | - Franco Henrique Andrade Leite
- Laboratory of Molecular Modeling, Department of Health, State University of Feira de Santana, Salvador 44036-900, BA, Brazil
- Postgraduate Program in Pharmaceutical Sciences, State University of Feira de Santana, Salvador 44036-900, BA, Brazil
| |
Collapse
|
34
|
Pan F, Huang Y, Cai X, Wang Y, Guan Y, Deng J, Yang D, Zhu J, Zhao Y, Xie F, Fang Z, Guo Q. Integrated algorithm combining plasma biomarkers and cognitive assessments accurately predicts brain β-amyloid pathology. COMMUNICATIONS MEDICINE 2023; 3:65. [PMID: 37165172 PMCID: PMC10172320 DOI: 10.1038/s43856-023-00295-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 04/27/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Accurate prediction of cerebral amyloidosis with easily available indicators is urgently needed for diagnosis and treatment of Alzheimer's disease (AD). METHODS We examined plasma Aβ42, Aβ40, T-tau, P-tau181, and NfL, with APOE genotypes, cognitive test scores and key demographics in a large Chinese cohort (N = 609, aged 40 to 84 years) covering full AD spectrum. Data-driven integrated computational models were developed to predict brain β-amyloid (Aβ) pathology. RESULTS Our computational models accurately predict brain Aβ positivity (area under the ROC curves (AUC) = 0.94). The results are validated in Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. Particularly, the models have the highest prediction power (AUC = 0.97) in mild cognitive impairment (MCI) participants. Three levels of models are designed with different accuracies and complexities. The model which only consists of plasma biomarkers can predict Aβ positivity in amnestic MCI (aMCI) patients with AUC = 0.89. Generally the models perform better in participants without comorbidities or family histories. CONCLUSIONS The innovative integrated models provide opportunity to assess Aβ pathology in a non-invasive and cost-effective way, which might facilitate AD-drug development, early screening, clinical diagnosis and prognosis evaluation.
Collapse
Affiliation(s)
- Fengfeng Pan
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yanlu Huang
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiao Cai
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China
| | - Ying Wang
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiale Deng
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China
| | - Dake Yang
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China
| | - Jinhang Zhu
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China
| | - Yike Zhao
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Zhuo Fang
- Department of Data & Analytics, WuXi Diagnostics Innovation Research Institute, Shanghai, China.
| | - Qihao Guo
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
35
|
Régy M, Dugravot A, Sabia S, Bouaziz-Amar E, Paquet C, Hanseeuw B, Singh-Manoux A, Dumurgier J. Association between ATN profiles and mortality in a clinical cohort of patients with cognitive disorders. Alzheimers Res Ther 2023; 15:77. [PMID: 37038213 PMCID: PMC10088112 DOI: 10.1186/s13195-023-01220-x] [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: 11/07/2022] [Accepted: 03/28/2023] [Indexed: 04/12/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the 5th leading cause of death in people 65 years and older. The ATN classification reflects a biological definition of AD pathology with markers of Aβ deposition (A), pathologic tau (T), and neurodegeneration (N). Little is known about the relationship between ATN status and the risk of mortality, leading us to examine this association in a relatively large population of patients seen at a memory clinic for cognitive disorders. METHODS Data were drawn from the BioCogBank Study, including patients seen for cognitive disorders in Lariboisiere Hospital (Paris, France), followed up to 15 years. All participants underwent a lumbar puncture for an assessment of the levels of CSF tau (tau), phosphorylated tau (p-tau181), and β-amyloid 42 peptide (Aβ42). Vital status on July 1, 2020, was recorded for each participant using the national mortality register. Individuals were categorized according to their ATN profiles based on CSF Aβ42 or Aβ42/40 ratio, p-tau181, and tau. Kaplan-Meier and multivariate Cox analyses were performed with A-T-N - participants as the reference using a short (5 years) and long follow-up (15 years). RESULTS Of the 1353 patients in the study (mean age: 68 years old, 53% of women, mean MMSE score: 22.6), 262 died during the follow-up. At 5 years of follow-up, A-T-N + individuals had the highest risk of mortality in Kaplan-Meier and adjusted Cox analyses [HR (95% CI) = 2.93 (1.31-6.56)]. At 15 years of follow-up, patients in the AD spectrum had a higher mortality risk with a gradient effect for biomarker positivity: A-T + [HR = 1.63 (1.04-2.55)], A + T - [HR = 2.17 (1.44-3.26)], and A + T + individuals [HR = 2.38 (1.66-3.39)], compared to A-T-N - patients. Adjustments on potential confounders had little impact on these associations. CONCLUSION This study shows ATN profiles to be associated with mortality in a relatively large patient cohort based on a memory clinic. Patients with isolated evidence of neurodegeneration had a higher mortality rate in the short follow-up, and patients with the AD profile had the highest mortality rate in the long follow-up.
Collapse
Affiliation(s)
- Mélina Régy
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
- Université Catholique de Louvain, Brussels, Belgium
| | - Aline Dugravot
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
| | - Séverine Sabia
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
- Division of Psychiatry, Faculty of Brain Sciences, University College London, London, UK
| | - Elodie Bouaziz-Amar
- Université Paris-Cité, Department of Biochemistry, GHU APHP Nord Lariboisière Fernand-Widal Hospital, Paris, France
| | - Claire Paquet
- Université Paris-Cité, Inserm U1144, Cognitive Neurology Center, GHU APHP Nord Lariboisière Fernand-Widal Hospital, Paris, France
| | - Bernard Hanseeuw
- Université Catholique de Louvain, Brussels, Belgium
- Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Archana Singh-Manoux
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
- Division of Psychiatry, Faculty of Brain Sciences, University College London, London, UK
| | - Julien Dumurgier
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France.
| |
Collapse
|
36
|
Li Z, Lin H, Zhang Q, Shi R, Xu H, Yang F, Jiang X, Wang L, Han Y, Jiang J. Individual Proportion Loss of Functional Connectivity Strength: A Novel Individual Functional Connectivity Biomarker for Subjective Cognitive Decline Populations. BIOLOGY 2023; 12:564. [PMID: 37106764 PMCID: PMC10135935 DOI: 10.3390/biology12040564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/04/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023]
Abstract
High individual variation in the subjective cognitive decline (SCD) population makes functional connectivity (FC) biomarkers unstable. This study proposed a novel individual FC index, named individual proportion loss of functional connectivity strength (IPLFCS), and explored potential biomarkers for SCD using this new index. We proposed an IPLFCS analysis framework and compared it with traditional FC in Chinese and Western cohorts. Post hoc tests were used to determine biomarkers. Pearson's correlation analysis was used to investigate the correlation between neuropsychological scores or cortical amyloid deposits and IPLFCS biomarkers. Receiver operating characteristic curves were utilized to evaluate the ability of potential biomarkers to distinguish between groups. IPLFCS of the left middle temporal gyrus (LMTG) was identified as a potential biomarker. The IPLFC was correlated with the traditional FC (r = 0.956, p < 0.001; r = 0.946, p < 0.001) and cortical amyloid deposition (r = -0.245, p = 0.029; r = -0.185, p = 0.048) in both cohorts. Furthermore, the IPLFCS decreased across the Alzheimer's disease (AD) continuum. Its diagnostic efficiency was superior to that of existing fMRI biomarkers. These findings suggest that IPLFCS of the LMTG could be a potential biomarker of SCD.
Collapse
Affiliation(s)
- Zhuoyuan Li
- School of Communication and Information Engineering, Shanghai University, Shanghai 200444, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, China
| | - Hua Lin
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Qi Zhang
- School of Communication and Information Engineering, Shanghai University, Shanghai 200444, China
| | - Rong Shi
- School of Communication and Information Engineering, Shanghai University, Shanghai 200444, China
| | - Huanyu Xu
- School of Communication and Information Engineering, Shanghai University, Shanghai 200444, China
| | - Fan Yang
- School of Communication and Information Engineering, Shanghai University, Shanghai 200444, China
| | - Xueyan Jiang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Luyao Wang
- Institute of Biomedical Engineering, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
- School of Biomedical Engineering, Hainan University, Haikou 570228, China
- Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Clinical Research Center for Geriatric Disorders, Beijing 100053, China
| | - Jiehui Jiang
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, China
- Institute of Biomedical Engineering, School of Life Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
37
|
Chun MY, Jang H, Kim HJ, Kim JP, Gallacher J, Allué JA, Sarasa L, Castillo S, Pascual-Lucas M, Na DL, Seo SW. Contribution of clinical information to the predictive performance of plasma β-amyloid levels for amyloid positron emission tomography positivity. Front Aging Neurosci 2023; 15:1126799. [PMID: 36998318 PMCID: PMC10044013 DOI: 10.3389/fnagi.2023.1126799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/24/2023] [Indexed: 03/15/2023] Open
Abstract
BackgroundEarly detection of β-amyloid (Aβ) accumulation, a major biomarker for Alzheimer’s disease (AD), has become important. As fluid biomarkers, the accuracy of cerebrospinal fluid (CSF) Aβ for predicting Aβ deposition on positron emission tomography (PET) has been extensively studied, and the development of plasma Aβ is beginning to receive increased attention recently. In the present study, we aimed to determine whether APOE genotypes, age, and cognitive status increase the predictive performance of plasma Aβ and CSF Aβ levels for Aβ PET positivity.MethodsWe recruited 488 participants who underwent both plasma Aβ and Aβ PET studies (Cohort 1) and 217 participants who underwent both cerebrospinal fluid (CSF) Aβ and Aβ PET studies (Cohort 2). Plasma and CSF samples were analyzed using ABtest-MS, an antibody-free liquid chromatography-differential mobility spectrometry-triple quadrupole mass spectrometry method and INNOTEST enzyme-linked immunosorbent assay kits, respectively. To evaluate the predictive performance of plasma Aβ and CSF Aβ, respectively, logistic regression and receiver operating characteristic analyses were performed.ResultsWhen predicting Aβ PET status, both plasma Aβ42/40 ratio and CSF Aβ42 showed high accuracy (plasma Aβ area under the curve (AUC) 0.814; CSF Aβ AUC 0.848). In the plasma Aβ models, the AUC values were higher than plasma Aβ alone model, when the models were combined with either cognitive stage (p < 0.001) or APOE genotype (p = 0.011). On the other hand, there was no difference between the CSF Aβ models, when these variables were added.ConclusionPlasma Aβ might be a useful predictor of Aβ deposition on PET status as much as CSF Aβ, particularly when considered with clinical information such as APOE genotype and cognitive stage.
Collapse
Affiliation(s)
- Min Young Chun
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Republic of Korea
| | - Hyemin Jang
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- *Correspondence: Hyemin Jang, ; Sang Won Seo,
| | - Hee Jin Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Jun Pyo Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - John Gallacher
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | - Duk L. Na
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Sang Won Seo
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- *Correspondence: Hyemin Jang, ; Sang Won Seo,
| | | |
Collapse
|
38
|
Souza ID, Anderson JL, Tumas V, Queiroz MEC. Direct coupling of fiber-in-tube solid-phase microextraction with tandem mass spectrometry to determine amyloid beta peptides as biomarkers for Alzheimer's disease in cerebrospinal fluid samples. Talanta 2023; 254:124186. [PMID: 36521326 DOI: 10.1016/j.talanta.2022.124186] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/29/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Current research efforts at neurological diseases have focused on identifying novel biomarkers to aid in diagnosis, to provide accurate prognostic information, and to monitor disease progression. This study presents the direct coupling of fiber-in-tube solid-phase microextraction to tandem mass spectrometry as a reliable method to determine amyloid beta peptides (Aβ38, Aβ40, and Aβ42) as biomarkers for Alzheimer's disease in cerebrospinal fluid (CSF) samples. To obtain the biocompatible fiber-in-tube SPME capillary, a PEEK tube segment was longitudinally packed with fine fibers [nitinol wires coated with a zwitterionic polymeric ionic liquid], to act as selective extraction medium. The fiber-in-tube SPME-MS/MS method integrated analyte extraction/enrichment and sample cleanup (exclusion of interferents) into one step. The method provided lower limits of quantification (LLOQ: 0.2 ng mL-1 for Aβ38 and 0.1 ng mL-1 for Aβ40 and Aβ42), high precision (CV lower than 11.6%), and high accuracy (relative standard deviation lower than 15.1%). This method was successfully applied to determine Aβ peptides in CSF samples obtained from AD patients (n = 8) and controls (healthy volunteers, n = 10). Results showed that Aβ42 levels in the CSF samples obtained from AD patients were significantly lower compared to healthy controls (p < 0.05). On the basis of the ROC analysis results, the Aβ42/Aβ40 ratio (AUC = 0.950, p < 0.01; 95%) performed significantly better than Aβ42 alone (AUC = 0.913, p < 0.01; 95%) in discriminating between AD patients and healthy controls and presented better diagnostic ability for AD. The novelties of this study are not only related to evaluating Aβ peptides as AD biomarkers, but also to demonstrating direct online coupling of fiber-in-tube SPME with MS/MS as a quantitative high-throughput method for bioanalysis.
Collapse
Affiliation(s)
- Israel D Souza
- Departamento de Química, Faculdade de Filosofia Ciências e Letras de Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Jared L Anderson
- Department of Chemistry, Iowa State University, Ames, IA, United States
| | - Vitor Tumas
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School of University of São Paulo, Ribeirao Preto, SP, Brazil
| | - Maria Eugênia C Queiroz
- Departamento de Química, Faculdade de Filosofia Ciências e Letras de Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
39
|
Wang S, Ke S, Liu S, Wang E, Pan T. APOE ε4 status and plasma p-tau181 interact to influence cognitive performance among non-demented older adults. Neurosci Lett 2023; 796:137052. [PMID: 36608927 DOI: 10.1016/j.neulet.2023.137052] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/25/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
OBJECTIVE In this study, we aimed to investigate the relationships among plasma p-tau181, APOE ε4, and cognitive performance in non-demented elderly individuals. METHODS We used individuals (n = 630) with cognitive normal (CN, n = 182) and mild cognitive impairment (MCI, n = 448). Multiple linear regression models were performed to test the effects of APOE ε4 × plasma p-tau181 interaction on MMSE, CDR-SOB, ADAS-cog13, and RAVLT immediate recall. All models adjusted for age, sex, and education. RESULTS In total, our study comprised 630 samples including 364 APOE ε4 non-carriers and 266 APOE ε4 carriers. In APOE ε4 carriers, plasma p-tau181 was significantly associated with MMSE (B = -0.04, p = 0.003), ADAS-Cog13 (B [unstandardized coefficient] = 0.21, p < 0.001), CDR-SB (B = 0.02, p = 0.003) and RAVLT immediate recall ((B = -0.17, p = 0.035). After correcting for Aβ status and diagnosis, the interaction between APOE ε4 and plasma p-tau181 was significant or marginally significant associations for RAVLT immediate recall (p = 0.076), MMSE (p = 0.011), CDR (p = 0.008), and ADAS-Cog13 (p < 0.001). CONCLUSIONS Our findings suggested that plasma p-tau181 levels predicted cognitive performance among non-demented older adults, but only in the APOE ε4 carriers.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of Neurology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, China
| | - Shaofa Ke
- Department of Neurology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, China
| | - Suzhi Liu
- Department of Neurology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, China
| | - En Wang
- Department of Neurology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, China
| | - Tengwei Pan
- Department of Neurology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, China.
| |
Collapse
|
40
|
Baik K, Jeon S, Park M, Lee YG, Lee PH, Sohn YH, Ye BS. Comparison Between 18F-Florapronol and 18F-Florbetaben Imaging in Patients With Cognitive Impairment. J Clin Neurol 2023; 19:260-269. [PMID: 36775276 PMCID: PMC10169926 DOI: 10.3988/jcn.2022.0207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 02/14/2023] Open
Abstract
BACKGROUND AND PURPOSE To determine the imaging characteristics and cutoff value of 18F-florapronol (FC119S) quantitative analysis for detecting β-amyloid positivity and Alzheimer's disease (AD), we compared the findings of FC119S and 18F-florbetaben (FBB) positron- emission tomography (PET) in patients with cognitive impairment. METHODS We prospectively enrolled 35 patients with cognitive impairment who underwent FBB-PET, FC119S-PET, and brain magnetic resonance imaging. We measured global and vertex-wise standardized uptake value ratios (SUVRs) using a surface-based method with the cerebellar gray matter as reference. Optimal global FC119S SUVR cutoffs were determined using receiver operating characteristic curves for β-amyloid positivity based on the global FBB SUVR of 1.478 and presence of AD, respectively. We evaluated the global and vertex-wise SUVR correlations between the two tracers. In addition, we performed correlation analysis for global or vertex-wise SUVR of each tracer with the vertex-wise cortical thicknesses. RESULTS The optimal global FC119S SUVR cutoff value was 1.385 both for detecting β-amyloid positivity and for detecting AD. Based on the global SUVR cutoff value of each tracer, 32 (91.4%) patients had concordant β-amyloid positivity. The SUVRs of FC119S and FBB had strong global (r=0.72) and vertex-wise (r>0.7) correlations in the overall cortices, except for the parietal and temporal cortices (0.4<r<0.7). The FC119S SUVR had significant negative vertex-wise correlations with cortical thicknesses in the posterior cingulate, anterior cingulate, parietal, posterior temporal, and occipital cortices. CONCLUSIONS Quantitative FC119S-PET analysis provided reliable information for detecting β-amyloid deposition and the presence of AD.
Collapse
Affiliation(s)
- Kyoungwon Baik
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Seun Jeon
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea.,Brain Research Institute, Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Mincheol Park
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea.,Department of Neurology, Chung-Ang University College of Medicine and Graduate School of Medicine, Gwangmyeong Hospital, Gwangmyeong, Korea
| | - Young-Gun Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Young H Sohn
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
41
|
Spooner A, Mohammadi G, Sachdev PS, Brodaty H, Sowmya A. Ensemble feature selection with data-driven thresholding for Alzheimer's disease biomarker discovery. BMC Bioinformatics 2023; 24:9. [PMID: 36624372 PMCID: PMC9830744 DOI: 10.1186/s12859-022-05132-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Feature selection is often used to identify the important features in a dataset but can produce unstable results when applied to high-dimensional data. The stability of feature selection can be improved with the use of feature selection ensembles, which aggregate the results of multiple base feature selectors. However, a threshold must be applied to the final aggregated feature set to separate the relevant features from the redundant ones. A fixed threshold, which is typically used, offers no guarantee that the final set of selected features contains only relevant features. This work examines a selection of data-driven thresholds to automatically identify the relevant features in an ensemble feature selector and evaluates their predictive accuracy and stability. Ensemble feature selection with data-driven thresholding is applied to two real-world studies of Alzheimer's disease. Alzheimer's disease is a progressive neurodegenerative disease with no known cure, that begins at least 2-3 decades before overt symptoms appear, presenting an opportunity for researchers to identify early biomarkers that might identify patients at risk of developing Alzheimer's disease. RESULTS The ensemble feature selectors, combined with data-driven thresholds, produced more stable results, on the whole, than the equivalent individual feature selectors, showing an improvement in stability of up to 34%. The most successful data-driven thresholds were the robust rank aggregation threshold and the threshold algorithm threshold from the field of information retrieval. The features identified by applying these methods to datasets from Alzheimer's disease studies reflect current findings in the AD literature. CONCLUSIONS Data-driven thresholds applied to ensemble feature selectors provide more stable, and therefore more reproducible, selections of features than individual feature selectors, without loss of performance. The use of a data-driven threshold eliminates the need to choose a fixed threshold a-priori and can select a more meaningful set of features. A reliable and compact set of features can produce more interpretable models by identifying the factors that are important in understanding a disease.
Collapse
Affiliation(s)
- Annette Spooner
- grid.1005.40000 0004 4902 0432School of Computer Science and Engineering, University of New South Wales, Sydney, Australia
| | - Gelareh Mohammadi
- grid.1005.40000 0004 4902 0432School of Computer Science and Engineering, University of New South Wales, Sydney, Australia
| | - Perminder S. Sachdev
- grid.1005.40000 0004 4902 0432Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Henry Brodaty
- grid.1005.40000 0004 4902 0432Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry & Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Arcot Sowmya
- grid.1005.40000 0004 4902 0432School of Computer Science and Engineering, University of New South Wales, Sydney, Australia
| | | |
Collapse
|
42
|
Wang J, Jin C, Zhou J, Zhou R, Tian M, Lee HJ, Zhang H. PET molecular imaging for pathophysiological visualization in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2023; 50:765-783. [PMID: 36372804 PMCID: PMC9852140 DOI: 10.1007/s00259-022-05999-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/09/2022] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) is the most common dementia worldwide. The exact etiology of AD is unclear as yet, and no effective treatments are currently available, making AD a tremendous burden posed on the whole society. As AD is a multifaceted and heterogeneous disease, and most biomarkers are dynamic in the course of AD, a range of biomarkers should be established to evaluate the severity and prognosis. Positron emission tomography (PET) offers a great opportunity to visualize AD from diverse perspectives by using radiolabeled agents involved in various pathophysiological processes; PET imaging technique helps to explore the pathomechanisms of AD comprehensively and find out the most appropriate biomarker in each AD phase, leading to a better evaluation of the disease. In this review, we discuss the application of PET in the course of AD and summarized radiolabeled compounds with favorable imaging characteristics.
Collapse
Affiliation(s)
- Jing Wang
- grid.412465.0Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XInstitute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, 310009 Zhejiang China ,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009 Zhejiang China
| | - Chentao Jin
- grid.412465.0Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009 Zhejiang China
| | - Jinyun Zhou
- grid.412465.0Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009 Zhejiang China
| | - Rui Zhou
- grid.412465.0Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009 Zhejiang China
| | - Mei Tian
- grid.412465.0Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XInstitute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, 310009 Zhejiang China ,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009 Zhejiang China
| | - Hyeon Jeong Lee
- grid.13402.340000 0004 1759 700XCollege of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310014 Zhejiang China
| | - Hong Zhang
- grid.412465.0Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XInstitute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, 310009 Zhejiang China ,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCollege of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310014 Zhejiang China ,grid.13402.340000 0004 1759 700XKey Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, 310014 Zhejiang China
| |
Collapse
|
43
|
Vicente MC, Paneghini JL, Stabile AM, Amorim M, Anibal Silva CE, Patrone LGA, Cunha TM, Bícego KC, Almeida MC, Carrettiero DC, Gargaglioni LH. Inhibition of Pro-Inflammatory Microglia with Minocycline Improves Cognitive and Sleep-Wake Dysfunction Under Respiratory Stress in a Sporadic Model for Alzheimer's Disease. J Alzheimers Dis 2023; 95:317-337. [PMID: 37522205 DOI: 10.3233/jad-230151] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
BACKGROUND Neuroinflammation in Alzheimer's disease (AD) can occur due to excessive activation of microglia in response to the accumulation of amyloid-β peptide (Aβ). Previously, we demonstrated an increased expression of this peptide in the locus coeruleus (LC) in a sporadic model for AD (streptozotocin, STZ; 2 mg/kg, ICV). We hypothesized that the STZ-AD model exhibits neuroinflammation, and treatment with an inhibitor of microglia (minocycline) can reverse the cognitive, respiratory, sleep, and molecular disorders of this model. OBJECTIVE To evaluate the effect of minocycline treatment in STZ model disorders. METHODS We treated control and STZ-treated rats for five days with minocycline (30 mg/kg, IP) and evaluated cognitive performance, chemoreflex response to hypercapnia and hypoxia, and total sleep time. Additionally, quantification of Aβ, microglia analyses, and relative expression of cytokines in the LC were performed. RESULTS Minocycline treatment improved learning and memory, which was concomitant with a decrease in microglial cell density and re-establishment of morphological changes induced by STZ in the LC region. Minocycline did not reverse the STZ-induced increase in CO2 sensitivity during wakefulness. However, it restored the daytime sleep-wake cycle in STZ-treated animals to the same levels as those observed in control animals. In the LC, levels of A and expression of Il10, Il1b, and Mcp1 mRNA remained unaffected by minocycline, but we found a strong trend of minocycline effect on Tnf- α. CONCLUSION Our findings suggest that minocycline effectively reduces microglial recruitment and the inflammatory morphological profile in the LC, while it recovers cognitive performance and restores the sleep-wake pattern impaired by STZ.
Collapse
Affiliation(s)
- Mariane C Vicente
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV, Jaboticabal, SP, Brazil
- Mary S. Easton Center for Alzheimer's Research and Care, Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Julia L Paneghini
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV, Jaboticabal, SP, Brazil
| | - Angelita M Stabile
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Mateus Amorim
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Conceição E Anibal Silva
- Department of Pharmachology, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV, Jaboticabal, SP, Brazil
| | - Thiago M Cunha
- Department of Pharmachology, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV, Jaboticabal, SP, Brazil
| | - Maria C Almeida
- Center for Natural and Human Sciences, Federal University of ABC, São Bernardo do Campo, Brazil
| | - Daniel C Carrettiero
- Center for Natural and Human Sciences, Federal University of ABC, São Bernardo do Campo, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV, Jaboticabal, SP, Brazil
| |
Collapse
|
44
|
Gao F, Zhang PF, Gao J, Song J, Chi S. The CCL2 rs4586 SNP Is Associated with Slower Amyloid-β Deposition and Faster Tau Accumulation of Alzheimer's Disease. J Alzheimers Dis 2022; 90:1647-1657. [PMID: 36314210 DOI: 10.3233/jad-220716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND CC-chemokine ligand 2 (CCL2), the key immunomodulatory chemokine for microglial activation, has been implicated in the pathogenesis of Alzheimer's disease (AD). Whether the association of CCL2 single nucleotide polymorphisms (SNPs) and the risk of AD is still controversial. OBJECTIVE We aimed to investigate whether CCL2 rs4586 SNP is associated with the pathological changes and cognitive decline of AD. METHODS A total of 486 participants with longitudinal cerebrospinal fluid (CSF) amyloid-β (Aβ) and phospho-tau (P-tau) biomarkers, 18F-Florbetapir and 18F-flortaucipir-positron emission tomography (PET), and cognitive assessments from the Alzheimer's disease Neuroimaging Initiative were included in the study. The effects of CCL2 rs4586 SNP on the pathological changes and cognitive decline of AD were assessed with linear mixed-effects models and evaluated according to the Aβ-status so as to identify whether the effects were independent of Aβ status. RESULTS CCL2 rs4586-CC carriers exhibited a slower global Aβ-PET accumulation, particularly within stage I and stage II. However, they exhibited a faster accumulation of CSF P-tau and global tau-PET standard uptake value ratios, especially in Braak I and Braak III/IV and the inferior temporal gyrus. The congruent effects of CCL2 rs4586 on tau accumulation existed only in the Aβ-group, as is shown in global tau-PET and Braak I. However, CCL2 rs4586 was not associated with the cognitive decline. CONCLUSION Our findings showed that the CCL2 rs4586-CC (versus TT/TC) genotype was associated with slower Aβ deposition and faster tau accumulation, and the latter of which was independent of Aβ status.
Collapse
Affiliation(s)
- Fan Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Peng-Fei Zhang
- Department of Medicine, Hangzhou Juno Genomics Inc, Hangzhou, China
| | - Jing Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jinghui Song
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Song Chi
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | | |
Collapse
|
45
|
Zhao YL, Ou YN, Ma YH, Huang YY, Bi YL, Tan L, Yu JT. Association between Life’s Simple 7 and cerebrospinal fluid biomarkers of Alzheimer’s disease pathology in cognitively intact adults: the CABLE study. Alzheimers Res Ther 2022; 14:74. [PMID: 35619174 PMCID: PMC9134665 DOI: 10.1186/s13195-022-01019-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/16/2022] [Indexed: 11/10/2022]
Abstract
Abstract
Introduction
This study sought to explore the association between Life’s Simple 7 (LS7) and cerebrospinal fluid (CSF) Alzheimer’s disease (AD) pathological biomarkers in the cognitively normal northern Chinese population.
Methods
From the Chinese Alzheimer’s Biomarker and LifestylE (CABLE) study, 1106 cognitively normal participants were enrolled. The mean age was 62.34 years, and 39.6% were female. LS7 scores were summed with each metric assigned 0, 1, or 2 scores. The multiple linear regression models were used to investigate the association between LS7 scores and CSF AD biomarkers.
Results
We found that LS7 scores were significantly associated with CSF AD pathologies, including Aβ42/40 (β = 0.034, P = .041), p-tau181 (β = − 0.043, P = .006), and t-tau (β = − 0.044, P = .003). In subscales, the biological metrics (blood pressure, cholesterol, glucose) were significantly related to CSF tau-related biomarkers. These associations were observed in the APOE ε4 allele non-carriers, yet not in carriers. The relationship of behavior metrics was found in the middle age and males.
Conclusion
Improving LS7 scores might do a favor to alleviate the pathology of AD in the preclinical stage, especially among the APOE ε4 allele non-carriers.
Collapse
|
46
|
Bélanger E, Couch E, Carroll MS, DePasquale N, Gadbois EA, Shepherd-Banigan M, Jutkowitz E, Van Houtven CH, Plassman BL, Wetle TT. Advance directives among cognitively impaired persons who had an amyloid PET scan and their care partners: a mixed-methods study. BMC Palliat Care 2022; 21:194. [PMID: 36336690 PMCID: PMC9638311 DOI: 10.1186/s12904-022-01082-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 10/17/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Little research exists on the role of β-amyloid PET scans as part of Alzheimer's diagnostic tests and documentation of end-of-life preferences for persons with cognitive impairment. The study objectives were to examine the association of amyloid PET scan results (elevated vs. not elevated amyloid levels) and diagnostic category (mild cognitive impairment vs. dementia) with the likelihood of having an advance directive (reported a median of 4.5 months post-scan); to explore perceptions of PET scan results and their influence on planning for the future among persons with cognitive impairment and their care partners. METHODS Sequential, explanatory mixed-methods design using data from dyads in the CARE-IDEAS study: advance directives as a factor of diagnostic category and scan result using multivariable logistic regression models; thematic analysis of semi-structured interviews with persons with cognitive impairment and care partners to explore how scan results influenced documentation of future healthcare preferences. Participants included 1784 persons with cognitive impairment and care partners from the CARE-IDEAS study, and a subsample of 100 semi-structured telephone interviews. RESULTS 81.6% of dyads reported an advance directive. Non-Hispanic, White participants had higher rates of advance directives. There was no significant association between having an advance directive and scan results. Qualitative analysis provided insight into perceived urgency to have advance directives, evolving healthcare preferences, and the context of completing advance directives. CONCLUSIONS Although amyloid PET scans prompted persons with cognitive impairment and care partners to consider progressive cognitive impairment as part of evolving healthcare preferences, we found substantial variability in the perceived urgency of documentation.
Collapse
Affiliation(s)
- Emmanuelle Bélanger
- Center for Gerontology and Healthcare Research, Brown University School of Public Health, 121 South Main Street, 6th Fl., Providence, RI, 02903, USA.
- Department of Health Services, Policy & Practice, Brown University School of Public Health, Providence, RI, USA.
| | - Elyse Couch
- Center for Gerontology and Healthcare Research, Brown University School of Public Health, 121 South Main Street, 6th Fl., Providence, RI, 02903, USA
| | - Michaela S Carroll
- Center for Gerontology and Healthcare Research, Brown University School of Public Health, 121 South Main Street, 6th Fl., Providence, RI, 02903, USA
| | - Nicole DePasquale
- Division of General Internal Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Emily A Gadbois
- Center for Gerontology and Healthcare Research, Brown University School of Public Health, 121 South Main Street, 6th Fl., Providence, RI, 02903, USA
- Department of Health Services, Policy & Practice, Brown University School of Public Health, Providence, RI, USA
| | - Megan Shepherd-Banigan
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
- Duke-Margolis Center for Health Policy, Durham, NC, USA
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham VA Health Care System, Durham, NC, USA
| | - Eric Jutkowitz
- Center for Gerontology and Healthcare Research, Brown University School of Public Health, 121 South Main Street, 6th Fl., Providence, RI, 02903, USA
- Department of Health Services, Policy & Practice, Brown University School of Public Health, Providence, RI, USA
| | - Courtney H Van Houtven
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
- Duke-Margolis Center for Health Policy, Durham, NC, USA
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham VA Health Care System, Durham, NC, USA
| | - Brenda L Plassman
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Terrie T Wetle
- Center for Gerontology and Healthcare Research, Brown University School of Public Health, 121 South Main Street, 6th Fl., Providence, RI, 02903, USA
- Department of Health Services, Policy & Practice, Brown University School of Public Health, Providence, RI, USA
| |
Collapse
|
47
|
Blackman J, Stankeviciute L, Arenaza-Urquijo EM, Suárez-Calvet M, Sánchez-Benavides G, Vilor-Tejedor N, Iranzo A, Molinuevo JL, Gispert JD, Coulthard E, Grau-Rivera O. Cross-sectional and longitudinal association of sleep and Alzheimer biomarkers in cognitively unimpaired adults. Brain Commun 2022; 4:fcac257. [PMID: 36337343 PMCID: PMC9630979 DOI: 10.1093/braincomms/fcac257] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 05/25/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Sleep abnormalities are prevalent in Alzheimer’s disease, with sleep quality already impaired at its preclinical stage. Epidemiological and experimental data point to sleep abnormalities contributing to the risk of Alzheimer’s disease. However, previous studies are limited by either a lack of Alzheimer’s disease biomarkers, reduced sample size or cross-sectional design. Understanding if, when, and how poor sleep contributes to Alzheimer’s disease progression is important so that therapies can be targeted to the right phase of the disease. Using the largest cohort to date, the European Prevention of Alzheimer’s Dementia Longitudinal Cohort Study, we test the hypotheses that poor sleep is associated with core Alzheimer’s disease CSF biomarkers cross-sectionally and predicts future increments of Alzheimer’s disease pathology in people without identifiable symptoms of Alzheimer’s disease at baseline. This study included 1168 adults aged over 50 years with CSF core Alzheimer’s disease biomarkers (total tau, phosphorylated tau and amyloid-beta), cognitive performance, and sleep quality (Pittsburgh sleep quality index questionnaire) data. We used multivariate linear regressions to analyse associations between core Alzheimer’s disease biomarkers and the following Pittsburgh sleep quality index measures: total score of sleep quality, binarized score (poor sleep categorized as Pittsburgh sleep quality index > 5), sleep latency, duration, efficiency and disturbance. On a subsample of 332 participants with CSF taken at baseline and after an average period of 1.5 years, we assessed the effect of baseline sleep quality on change in Alzheimer’s disease biomarkers over time. Cross-sectional analyses revealed that poor sleep quality (Pittsburgh sleep quality index total > 5) was significantly associated with higher CSF t-tau; shorter sleep duration (<7 h) was associated with higher CSF p-tau and t-tau; and a higher degree of sleep disturbance (1–9 versus 0 and >9 versus 0) was associated with lower CSF amyloid-beta. Longitudinal analyses showed that greater sleep disturbances (1–9 versus 0 and >9 versus 0) were associated with a decrease in CSF Aβ42 over time. This study demonstrates that self-reported poor sleep quality is associated with greater Alzheimer’s disease-related pathology in cognitively unimpaired individuals, with longitudinal results further strengthening the hypothesis that disrupted sleep may represent a risk factor for Alzheimer’s disease. This highlights the need for future work to test the efficacy of preventive practices, designed to improve sleep at pre-symptomatic stages of disease, on reducing Alzheimer’s disease pathology.
Collapse
Affiliation(s)
- Jonathan Blackman
- North Bristol NHS Trust , Bristol BS10 5NB , UK
- Bristol Medical School, University of Bristol , Bristol BS8 1UD , UK
| | - Laura Stankeviciute
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- Universitat Pompeu Fabra , Barcelona 08005 , Spain
| | - Eider M Arenaza-Urquijo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- IMIM (Hospital del Mar Medical Research Institute) , Barcelona 08003 , Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) , Madrid 28029 , Spain
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- IMIM (Hospital del Mar Medical Research Institute) , Barcelona 08003 , Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) , Madrid 28029 , Spain
- Servei de Neurologia, Hospital del Mar , Barcelona 08003 , Spain
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- IMIM (Hospital del Mar Medical Research Institute) , Barcelona 08003 , Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) , Madrid 28029 , Spain
| | - Natalia Vilor-Tejedor
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- Universitat Pompeu Fabra , Barcelona 08005 , Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology , Barcelona 08003 , Spain
- Department of Clinical Genetics, Erasmus University Medical Center , Rotterdam 3015 GD , The Netherlands
| | - Alejandro Iranzo
- Neurology Service, Hospital Clínic de Barcelona and Institut D'Investigacions Biomèdiques , Barcelona 08036 , Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic de Barcelona , Barcelona 28029 , Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- IMIM (Hospital del Mar Medical Research Institute) , Barcelona 08003 , Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) , Madrid 28029 , Spain
| | - Elizabeth Coulthard
- North Bristol NHS Trust , Bristol BS10 5NB , UK
- Bristol Medical School, University of Bristol , Bristol BS8 1UD , UK
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation , Barcelona 08005 , Spain
- IMIM (Hospital del Mar Medical Research Institute) , Barcelona 08003 , Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) , Madrid 28029 , Spain
- Servei de Neurologia, Hospital del Mar , Barcelona 08003 , Spain
| | | |
Collapse
|
48
|
Nojima H, Ito S, Kushida A, Abe A, Motsuchi W, Verbel D, Vandijck M, Jannes G, Vandenbroucke I, Aoyagi K. Clinical utility of cerebrospinal fluid biomarkers measured by LUMIPULSE ® system. Ann Clin Transl Neurol 2022; 9:1898-1909. [PMID: 36321325 PMCID: PMC9735374 DOI: 10.1002/acn3.51681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVES Cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease (AD) are well-established in research settings, but their use in routine clinical practice remains a largely unexploited potential. Here, we examined the relationship between CSF biomarkers, measured by a fully automated immunoassay platform, and brain β-amyloid (Aβ) deposition status confirmed by amyloid positron emission tomography (PET). METHODS One hundred ninety-nine CSF samples from clinically diagnosed AD patients enrolled in a clinical study and who underwent amyloid PET were used for the measurement of CSF biomarkers Aβ 1-40 (Aβ40), Aβ 1-42 (Aβ42), total tau (t-Tau), and phosphorylated tau-181 (p-Tau181) using the LUMIPULSE system. These biomarkers and their combinations were compared to amyloid PET classification (negative or positive) using visual read assessments. Several combinations were also analyzed with a multivariable logistic regression model. RESULTS Aβ42, t-Tau, and p-Tau181, and the ratios of Aβ42 with other biomarkers had a good diagnostic agreement with amyloid PET imaging. The multivariable logistic regression analysis showed that amyloid PET status was associated with Aβ40 and Aβ42, but other factors, such as MMSE, sex, t-Tau, and p-Tau181, did not significantly add information to the model. CONCLUSIONS CSF biomarkers measured with the LUMIPULSE system showed good agreement with amyloid PET imaging. The ratio of Aβ42 with the other analyzed biomarkers showed a higher correlation with amyloid PET than Aβ42 alone, suggesting that the combinations of biomarkers could be useful in the diagnostic assessment in clinical research and potentially in routine clinical practice.
Collapse
Affiliation(s)
- Hisashi Nojima
- FUJIREBIO Inc.2‐1‐1, Nishishinjuku, Shinjuku‐kuTokyo163‐0410Japan
| | - Satoshi Ito
- Eisai Co., Ltd. 4‐6‐10 KoishikawaBunkyo‐kuTokyo112‐8088Japan,Eisai Inc.200 Metro BoulevardNutleyNew Jersey07110USA
| | - Akira Kushida
- FUJIREBIO Inc.2‐1‐1, Nishishinjuku, Shinjuku‐kuTokyo163‐0410Japan
| | - Aki Abe
- FUJIREBIO Inc.2‐1‐1, Nishishinjuku, Shinjuku‐kuTokyo163‐0410Japan
| | - Wataru Motsuchi
- FUJIREBIO Inc.2‐1‐1, Nishishinjuku, Shinjuku‐kuTokyo163‐0410Japan
| | - David Verbel
- Eisai Inc.200 Metro BoulevardNutleyNew Jersey07110USA
| | - Manu Vandijck
- Fujirebio‐Europe N.V.Technologiepark 69052GhentBelgium
| | - Geert Jannes
- Fujirebio‐Europe N.V.Technologiepark 69052GhentBelgium
| | | | - Katsumi Aoyagi
- FUJIREBIO Inc.2‐1‐1, Nishishinjuku, Shinjuku‐kuTokyo163‐0410Japan
| |
Collapse
|
49
|
Rezai AR, Ranjan M, Haut MW, Carpenter J, D’Haese PF, Mehta RI, Najib U, Wang P, Claassen DO, Chazen JL, Krishna V, Deib G, Zibly Z, Hodder SL, Wilhelmsen KC, Finomore V, Konrad PE, Kaplitt M, _ _. Focused ultrasound–mediated blood-brain barrier opening in Alzheimer’s disease: long-term safety, imaging, and cognitive outcomes. J Neurosurg 2022:1-9. [DOI: 10.3171/2022.9.jns221565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE
MRI-guided low-intensity focused ultrasound (FUS) has been shown to reversibly open the blood-brain barrier (BBB), with the potential to deliver therapeutic agents noninvasively to target brain regions in patients with Alzheimer’s disease (AD) and other neurodegenerative conditions. Previously, the authors reported the short-term safety and feasibility of FUS BBB opening of the hippocampus and entorhinal cortex (EC) in patients with AD. Given the need to treat larger brain regions beyond the hippocampus and EC, brain volumes and locations treated with FUS have now expanded. To evaluate any potential adverse consequences of BBB opening on disease progression, the authors report safety, imaging, and clinical outcomes among participants with mild AD at 6–12 months after FUS treatment targeted to the hippocampus, frontal lobe, and parietal lobe.
METHODS
In this open-label trial, participants with mild AD underwent MRI-guided FUS sonication to open the BBB in β-amyloid positive regions of the hippocampus, EC, frontal lobe, and parietal lobe. Participants underwent 3 separate FUS treatment sessions performed 2 weeks apart. Outcome assessments included safety, imaging, neurological, cognitive, and florbetaben β-amyloid PET.
RESULTS
Ten participants (range 55–76 years old) completed 30 separate FUS treatments at 2 participating institutions, with 6–12 months of follow-up. All participants had immediate BBB opening after FUS and BBB closure within 24–48 hours. All FUS treatments were well tolerated, with no serious adverse events related to the procedure. All 10 participants had a minimum of 6 months of follow-up, and 7 participants had a follow-up out to 1 year. Changes in the Alzheimer’s Disease Assessment Scale–cognitive and Mini-Mental State Examination scores were comparable to those in controls from the Alzheimer’s Disease Neuroimaging Initiative. PET scans demonstrated an average β-amyloid plaque of 14% in the Centiloid scale in the FUS-treated regions.
CONCLUSIONS
This study is the largest cohort of participants with mild AD who received FUS treatment, and has the longest follow-up to date. Safety was demonstrated in conjunction with reversible and repeated BBB opening in multiple cortical and deep brain locations, with a concomitant reduction of β-amyloid. There was no apparent cognitive worsening beyond expectations up to 1 year after FUS treatment, suggesting that the BBB opening treatment in multiple brain regions did not adversely influence AD progression. Further studies are needed to determine the clinical significance of these findings. FUS offers a unique opportunity to decrease amyloid plaque burden as well as the potential to deliver targeted therapeutics to multiple brain regions in patients with neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | - Marc W. Haut
- Behavioral Medicine and Psychiatry,
- Neurology, and
| | - Jeffrey Carpenter
- Neuroradiology, WVU Rockefeller Neuroscience Institute, Morgantown, West Virginia
| | | | - Rashi I. Mehta
- Neuroradiology, WVU Rockefeller Neuroscience Institute, Morgantown, West Virginia
| | | | - Peng Wang
- Neuroradiology, WVU Rockefeller Neuroscience Institute, Morgantown, West Virginia
| | | | | | - Vibhor Krishna
- Department of Neurosurgery, University of North Carolina, Chapel Hill, North Carolina
| | - Gerard Deib
- Neuroradiology, WVU Rockefeller Neuroscience Institute, Morgantown, West Virginia
| | - Zion Zibly
- Department of Neurosurgery, Sheba Medical Center, Ramat Gan, Israel; and
| | - Sally L. Hodder
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown, West Virginia
| | | | | | | | - Michael Kaplitt
- Neurological Surgery, Weill Cornell Medical College, New York, New York
| | | |
Collapse
|
50
|
Morrison MS, Aparicio HJ, Blennow K, Zetterberg H, Ashton NJ, Karikari TK, Tripodis Y, Martin B, Palmisano JN, Sugarman MA, Frank B, Steinberg EG, Turk KW, Budson AE, Au R, Goldstein LE, Jun GR, Kowall NW, Killiany R, Qiu WQ, Stern RA, Mez J, McKee AC, Stein TD, Alosco ML. Ante-mortem plasma phosphorylated tau (181) predicts Alzheimer's disease neuropathology and regional tau at autopsy. Brain 2022; 145:3546-3557. [PMID: 35554506 PMCID: PMC10233293 DOI: 10.1093/brain/awac175] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/07/2022] [Accepted: 05/02/2022] [Indexed: 11/14/2022] Open
Abstract
Blood-based biomarkers such as tau phosphorylated at threonine 181 (phosphorylated-tau181) represent an accessible, cost-effective and scalable approach for the in vivo detection of Alzheimer's disease pathophysiology. Plasma-pathological correlation studies are needed to validate plasma phosphorylated-tau181 as an accurate and reliable biomarker of Alzheimer's disease neuropathological changes. This plasma-to-autopsy correlation study included participants from the Boston University Alzheimer's Disease Research Center who had a plasma sample analysed for phosphorylated-tau181 between 2008 and 2018 and donated their brain for neuropathological examination. Plasma phosphorelated-tau181 was measured with single molecule array technology. Of 103 participants, 62 (60.2%) had autopsy-confirmed Alzheimer's disease. Average time between blood draw and death was 5.6 years (standard deviation = 3.1 years). Multivariable analyses showed higher plasma phosphorylated-tau181 concentrations were associated with increased odds for having autopsy-confirmed Alzheimer's disease [AUC = 0.82, OR = 1.07, 95% CI = 1.03-1.11, P < 0.01; phosphorylated-tau standardized (z-transformed): OR = 2.98, 95% CI = 1.50-5.93, P < 0.01]. Higher plasma phosphorylated-tau181 levels were associated with increased odds for having a higher Braak stage (OR = 1.06, 95% CI = 1.02-1.09, P < 0.01) and more severe phosphorylated-tau across six cortical and subcortical brain regions (ORs = 1.03-1.06, P < 0.05). The association between plasma phosphorylated-tau181 and Alzheimer's disease was strongest in those who were demented at time of blood draw (OR = 1.25, 95%CI = 1.02-1.53), but an effect existed among the non-demented (OR = 1.05, 95% CI = 1.01-1.10). There was higher discrimination accuracy for Alzheimer's disease when blood draw occurred in years closer to death; however, higher plasma phosphorylated-tau181 levels were associated with Alzheimer's disease even when blood draw occurred >5 years from death. Ante-mortem plasma phosphorylated-tau181 concentrations were associated with Alzheimer's disease neuropathology and accurately differentiated brain donors with and without autopsy-confirmed Alzheimer's disease. These findings support plasma phosphorylated-tau181 as a scalable biomarker for the detection of Alzheimer's disease.
Collapse
Affiliation(s)
- Madeline S Morrison
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Hugo J Aparicio
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Framingham Heart Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London WC1N 3BG, UK
| | - Nicholas J Ashton
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Thomas K Karikari
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Yorghos Tripodis
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Brett Martin
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA 02118, USA
| | - Joseph N Palmisano
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA 02118, USA
| | - Michael A Sugarman
- Department of Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Brandon Frank
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Eric G Steinberg
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Katherine W Turk
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, MA 02130, USA
| | - Andrew E Budson
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, MA 02130, USA
| | - Rhoda Au
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Framingham Heart Study, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
| | - Lee E Goldstein
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, Boston University College of Engineering, Boston, MA 02215, USA
| | - Gyungah R Jun
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Neil W Kowall
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, MA 02130, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ronald Killiany
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Biomedical Imaging, Boston University School of Medicine, Boston, MA 02118, USA
| | - Wei Qiao Qiu
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Robert A Stern
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurosurgery, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jesse Mez
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Framingham Heart Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ann C McKee
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, MA 02130, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- VA Bedford Healthcare System, U.S. Department of Veteran Affairs, Bedford, MA 01730, USA
| | - Thor D Stein
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, MA 02130, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- VA Bedford Healthcare System, U.S. Department of Veteran Affairs, Bedford, MA 01730, USA
| | - Michael L Alosco
- Boston University Alzheimer’s Disease Research Center and CTE Center, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|