51
|
Tolar M, Hey JA, Power A, Abushakra S. The Single Toxin Origin of Alzheimer's Disease and Other Neurodegenerative Disorders Enables Targeted Approach to Treatment and Prevention. Int J Mol Sci 2024; 25:2727. [PMID: 38473975 PMCID: PMC10932387 DOI: 10.3390/ijms25052727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
New data suggest that the aggregation of misfolded native proteins initiates and drives the pathogenic cascade that leads to Alzheimer's disease (AD) and other age-related neurodegenerative disorders. We propose a unifying single toxin theory of brain neurodegeneration that identifies new targets and approaches to the development of disease-modifying treatments. An extensive body of genetic evidence suggests soluble aggregates of beta-amyloid (Aβ) as the primary neurotoxin in the pathogenesis of AD. New insights from fluid biomarkers, imaging, and clinical studies provide further evidence for the decisive impact of toxic Aβ species in the initiation and progression of AD. Understanding the distinct roles of soluble and insoluble amyloid aggregates on AD pathogenesis has been the key missing piece of the Alzheimer's puzzle. Data from clinical trials with anti-amyloid agents and recent advances in the diagnosis of AD demonstrate that the driving insult in biologically defined AD is the neurotoxicity of soluble Aβ aggregates, called oligomers and protofibrils, rather than the relatively inert insoluble mature fibrils and amyloid plaques. Amyloid oligomers appear to be the primary factor causing the synaptic impairment, neuronal stress, spreading of tau pathology, and eventual cell death that lead to the clinical syndrome of AD dementia. All other biochemical effects and neurodegenerative changes in the brain that are observed in AD are a response to or a downstream effect of this initial toxic insult by oligomers. Other neurodegenerative disorders follow a similar pattern of pathogenesis, in which normal brain proteins with important biological functions become trapped in the aging brain due to impaired clearance and then misfold and aggregate into neurotoxic species that exhibit prion-like behavior. These aggregates then spread through the brain and cause disease-specific neurodegeneration. Targeting the inhibition of this initial step in neurodegeneration by blocking the misfolding and aggregation of healthy proteins has the potential to slow or arrest disease progression, and if treatment is administered early in the course of AD and other neurodegenerative disorders, it may delay or prevent the onset of clinical symptoms.
Collapse
|
52
|
Mravinacová S, Alanko V, Bergström S, Bridel C, Pijnenburg Y, Hagman G, Kivipelto M, Teunissen C, Nilsson P, Matton A, Månberg A. CSF protein ratios with enhanced potential to reflect Alzheimer's disease pathology and neurodegeneration. Mol Neurodegener 2024; 19:15. [PMID: 38350954 PMCID: PMC10863228 DOI: 10.1186/s13024-024-00705-z] [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: 08/31/2023] [Accepted: 01/23/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Amyloid and tau aggregates are considered to cause neurodegeneration and consequently cognitive decline in individuals with Alzheimer's disease (AD). Here, we explore the potential of cerebrospinal fluid (CSF) proteins to reflect AD pathology and cognitive decline, aiming to identify potential biomarkers for monitoring outcomes of disease-modifying therapies targeting these aggregates. METHOD We used a multiplex antibody-based suspension bead array to measure the levels of 49 proteins in CSF from the Swedish GEDOC memory clinic cohort at the Karolinska University Hospital. The cohort comprised 148 amyloid- and tau-negative individuals (A-T-) and 65 amyloid- and tau-positive individuals (A+T+). An independent sample set of 26 A-T- and 26 A+T+ individuals from the Amsterdam Dementia Cohort was used for validation. The measured proteins were clustered based on their correlation to CSF amyloid beta peptides, tau and NfL levels. Further, we used support vector machine modelling to identify protein pairs, matched based on their cluster origin, that reflect AD pathology and cognitive decline with improved performance compared to single proteins. RESULTS The protein-clustering revealed 11 proteins strongly correlated to t-tau and p-tau (tau-associated group), including mainly synaptic proteins previously found elevated in AD such as NRGN, GAP43 and SNCB. Another 16 proteins showed predominant correlation with Aβ42 (amyloid-associated group), including PTPRN2, NCAN and CHL1. Support vector machine modelling revealed that proteins from the two groups combined in pairs discriminated A-T- from A+T+ individuals with higher accuracy compared to single proteins, as well as compared to protein pairs composed of proteins originating from the same group. Moreover, combining the proteins from different groups in ratios (tau-associated protein/amyloid-associated protein) significantly increased their correlation to cognitive decline measured with cognitive scores. The results were validated in an independent cohort. CONCLUSIONS Combining brain-derived proteins in pairs largely enhanced their capacity to discriminate between AD pathology-affected and unaffected individuals and increased their correlation to cognitive decline, potentially due to adjustment of inter-individual variability. With these results, we highlight the potential of protein pairs to monitor neurodegeneration and thereby possibly the efficacy of AD disease-modifying therapies.
Collapse
Affiliation(s)
- Sára Mravinacová
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Vilma Alanko
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Sofia Bergström
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Claire Bridel
- Neurochemistry Lab, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Yolande Pijnenburg
- Department of Neurology, Alzheimer Centre, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Göran Hagman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology (AGE) Research Unit, Imperial College London, London, United Kingdom
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Charlotte Teunissen
- Neurochemistry Lab, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Peter Nilsson
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Anna Matton
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology (AGE) Research Unit, Imperial College London, London, United Kingdom
| | - Anna Månberg
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden.
| |
Collapse
|
53
|
Butts B, Huang H, Hu WT, Kehoe PG, Miners JS, Verble DD, Zetterberg H, Zhao L, Trotti LM, Benameur K, Scorr LM, Wharton W. sPDGFRβ and neuroinflammation are associated with AD biomarkers and differ by race: The ASCEND Study. Alzheimers Dement 2024; 20:1175-1189. [PMID: 37933404 PMCID: PMC10916968 DOI: 10.1002/alz.13457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 11/08/2023]
Abstract
INTRODUCTION There remains an urgent need to identify preclinical pathophysiological mechanisms of Alzheimer's disease (AD) development in high-risk, racially diverse populations. We explored the relationship between cerebrospinal fluid (CSF) markers of vascular injury and neuroinflammation with AD biomarkers in middle-aged Black/African American (B/AA) and non-Hispanic White (NHW) participants. METHODS Adults (45-65 years) with a parental history of AD were enrolled (n = 82). CSF and blood biomarkers were collected at baseline and year 2. RESULTS CSF total tau (t-tau), phosphorylated tau (p-tau), and amyloid beta (Aβ)40 were elevated at year 2 compared to baseline. CSF soluble platelet-derived growth factor receptor β (sPDGFRβ) levels, a marker of pericyte injury, correlated positively with t-tau, p-tau, Aβ40 markers of vascular injury, and cytokines at baseline and year 2. CSF sPDGFRβ and tau were significantly lower in B/AA than NHW. DISCUSSION Vascular dysfunction and neuroinflammation may precede cognitive decline and disease pathology in the very early preclinical stages of AD, and there are race-related differences in these relationships. HIGHLIGHTS Cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers changed over 2 years in high-risk middle-aged adults. Markers of vascular dysfunction were associated with the CSF biomarkers amyloid beta and tau. AD biomarkers were lower in Black compared to non-Hispanic White individuals. Markers of vascular dysfunction were lower among Black individuals.
Collapse
Affiliation(s)
- Brittany Butts
- Emory UniversityNell Hodgson Woodruff School of NursingAtlantaGeorgiaUSA
| | - Hanfeng Huang
- Georgetown University, School of MedicineWashingtonDistrict of ColumbiaUSA
| | - William T. Hu
- Rutgers UniversityInstitute for Health, Health Care Policy, and Aging ResearchNew BrunswickNew JerseyUSA
| | | | | | - Danielle D. Verble
- Emory UniversityNell Hodgson Woodruff School of NursingAtlantaGeorgiaUSA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water BayHong KongChina
| | - Liping Zhao
- Emory UniversityRollins School of Public HealthAtlantaGeorgiaUSA
| | | | | | | | - Whitney Wharton
- Emory UniversityNell Hodgson Woodruff School of NursingAtlantaGeorgiaUSA
| |
Collapse
|
54
|
Giuffrè GM, Quaranta D, Citro S, Morganti TG, Martellacci N, Vita MG, Rossini PM, Calabresi P, Marra C. Associations Between Free and Cued Selective Reminding Test and Cerebrospinal Fluid Biomarkers in Amnestic Mild Cognitive Impairment. J Alzheimers Dis 2024; 100:713-723. [PMID: 38905044 DOI: 10.3233/jad-240150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
Background The Free and Cued Selective Reminding Test (FCSRT), assessing verbal episodic memory with controlled learning and semantic cueing, has been recommended for detecting the genuine encoding and storage deficits characterizing AD-related memory disorders. Objective The present study aims at investigating the ability of FCSRT in predicting cerebrospinal fluid (CSF) evidence of amyloid-β positivity in subjects with amnestic mild cognitive impairment (aMCI) and exploring its associations with amyloidopathy, tauopathy and neurodegeneration biomarkers. Methods 120 aMCI subjects underwent comprehensive neurological and neuropsychological examinations, including the FCSRT assessment, and CSF collection; CSF Aβ42/40 ratio, p-tau181, and total-tau quantification were conducted by an automated CLEIA method on Lumipulse G1200. Based on the Aβ42/40 ratio value, subjects were classified as either A+ or A-. Results All FCSRT subitem scores were significantly lower in A+ group and significantly predicted the amyloid-β status, with Immediate Total Recall (ITR) being the best predictor. No significant correlations were found between FCSRT and CSF biomarkers in the A- aMCI group, while in the A+ aMCI group, all FCSRT subitem scores were negatively correlated with CSF p-tau181 and total-tau, but not with the Aβ42/40 ratio. Conclusions FCSRT confirms its validity as a tool for the diagnosis of AD, being able to predict the presence of amyloid-β deposition with high specificity. The associations between FCSRT subitem scores and CSF p-tau-181 and total-tau levels in aMCI due to AD could further encourage the clinical use of this simple and cost-effective test in the evaluation of individuals with aMCI.
Collapse
Affiliation(s)
- Guido Maria Giuffrè
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Davide Quaranta
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Salvatore Citro
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Tommaso Giuseppe Morganti
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Noemi Martellacci
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Gabriella Vita
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Paolo Maria Rossini
- Department of Neuroscience and Neurorehabilitation, Brain Connectivity Laboratory, IRCCS San Raffaele Roma, Rome, Italy
| | - Paolo Calabresi
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Camillo Marra
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
55
|
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
|
56
|
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
|
57
|
Sun CK, Guo F, Ou YN, Zhang MZ, Tan L, Tan MS. Association Between Carotid Plaque and Alzheimer's Disease Cerebrospinal Fluid Biomarkers and Cognitive Function in Cognitively Intact Adults: The CABLE Study. J Alzheimers Dis 2024; 100:207-217. [PMID: 38848186 DOI: 10.3233/jad-240131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Background The association between carotid plaque and cognitive decline has recently been reported. However, the current research evidence is insufficient, and the possible causes of cognitive changes are unknown. Objective This study aims to explore the relationships between carotid plaque and cognition functions, cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers in cognitively intact adults, and try to study the underlying mechanisms. Methods We enrolled 165 cognitively normal participants from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) study, who had CSF AD biomarker measurements and carotid ultrasound. Linear modeling was used to assess the association of carotid plaque with CSF biomarkers and cognition. Additionally, mediation analysis was conducted through 10,000 bootstrapped iterations to explore potential links between carotid plaque, AD pathology, and cognition. Results We found that carotid plaque exhibited significant correlations with Aβ42 (β = -1.173, p = 0.022), Aβ42/Aβ40 (β = -0.092, p < 0.001), P-tau/Aβ42 (β = 0.110, p = 0.045), and T-tau/Aβ42 (β = 0.451, p = 0.010). A significant correlation between carotid plaque and cognition decline was also found in men (β = -0.129, p = 0.021), and mediation analyses revealed that the effect of carotid plaque on cognitive function could be mediated by Aβ42/Aβ40 (proportion of mediation = 55.8%), P-tau/Aβ42 (proportion of mediation = 51.6%, p = 0.015) and T-tau/Aβ42 (proportion of mediation = 43.8%, p = 0.015) mediated. Conclusions This study demonstrated the link between carotid plaque and CSF AD biomarkers in cognitively intact adults, and the important role that AD pathology may play in the correlation between carotid plaque and cognitive changes.
Collapse
Affiliation(s)
- Cheng-Kun Sun
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Fan Guo
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ming-Zhan Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Department of Neurology, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao, China
| | - Meng-Shan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Department of Neurology, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao, China
| |
Collapse
|
58
|
Goodman MJ, Li XR, Livschitz J, Huang CC, Bendlin BB, Granadillo ED, for the Alzheimer’s Disease Neuroimaging Initiative. Comparing Symmetric Dimethylarginine and Amyloid-β42 as Predictors of Alzheimer's Disease Development. J Alzheimers Dis Rep 2023; 7:1427-1444. [PMID: 38225970 PMCID: PMC10789286 DOI: 10.3233/adr-230054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/15/2023] [Indexed: 01/17/2024] Open
Abstract
Background Physicians may soon be able to diagnose Alzheimer's disease (AD) in its early stages using fluid biomarkers like amyloid. However, it is acknowledged that additional biomarkers need to be characterized which would facilitate earlier monitoring of AD pathogenesis. Objective To determine if a potential novel inflammation biomarker for AD, symmetric dimethylarginine, has utility as a baseline serum biomarker for discriminating prodromal AD from cognitively unimpaired controls in comparison to cerebrospinal fluid amyloid-β42 (Aβ42). Methods Data including demographics, magnetic resonance imaging and fluorodeoxyglucose-positron emission tomography scans, Mini-Mental State Examination and Functional Activities Questionnaire scores, and biomarker concentrations were obtained from the Alzheimer's Disease Neuroimaging Initiative for a total of 146 prodromal AD participants and 108 cognitively unimpaired controls. Results Aβ42 (p = 0.65) and symmetric dimethylarginine (p = 0.45) were unable to predict age-matched cognitively unimpaired controls and prodromal AD participants. Aβ42 was negatively associated with regional brain atrophy and hypometabolism as well as cognitive and functional decline in cognitively unimpaired control participants (p < 0.05) that generally decreased in time. There were no significant associations between Aβ42 and symmetric dimethylarginine with imaging or neurocognitive biomarkers in prodromal AD patients. Conclusions Correlations were smaller between Aβ42 and neuropathological biomarkers over time and were absent in prodromal AD participants, suggesting a plateau effect dependent on age and disease stage. Evidence supporting symmetric dimethylarginine as a novel biomarker for AD as a single measurement was not found.
Collapse
Affiliation(s)
| | - Xin Ran Li
- Medical College of Wisconsin, Wauwatosa, WI, USA
| | | | | | | | - Elias D. Granadillo
- Medical College of Wisconsin, Wauwatosa, WI, USA
- University of Wisconsin, Madison, WI, USA
| | | |
Collapse
|
59
|
Cisterna-García A, Beric A, Ali M, Pardo JA, Chen HH, Fernandez MV, Norton J, Gentsch J, Bergmann K, Budde J, Perlmutter JS, Morris JC, Cruchaga C, Botia JA, Ibanez L. Cell-free RNA signatures predict Alzheimer's disease. iScience 2023; 26:108534. [PMID: 38089583 PMCID: PMC10711471 DOI: 10.1016/j.isci.2023.108534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 02/01/2024] Open
Abstract
There is a need for affordable, scalable, and specific blood-based biomarkers for Alzheimer's disease that can be applied to a population level. We have developed and validated disease-specific cell-free transcriptomic blood-based biomarkers composed by a scalable number of transcripts that capture AD pathobiology even in the presymptomatic stages of the disease. Accuracies are in the range of the current CSF and plasma biomarkers, and specificities are high against other neurodegenerative diseases.
Collapse
Affiliation(s)
- Alejandro Cisterna-García
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, Murcia, Spain
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Aleksandra Beric
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Jose Adrian Pardo
- Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, Murcia, Spain
| | - Hsiang-Han Chen
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Maria Victoria Fernandez
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Joanne Norton
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Jen Gentsch
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Kristy Bergmann
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - John Budde
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Joel S. Perlmutter
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Department of Radiology, Neuroscience, Physical Therapy, and Occupational Therapy, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - John C. Morris
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis, Saint Louis, MO, USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Department of Pathology and Immunology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis, Saint Louis, MO, USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Department of Genetics, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Juan A. Botia
- Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, Murcia, Spain
| | - Laura Ibanez
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
60
|
Mohammadi I, Adibparsa M, Najafi A, Sehat MS, Sadeghi M. A systematic review with meta-analysis to assess Alzheimer's disease biomarkers in adults with or without obstructive sleep apnoea. Int Orthod 2023; 21:100814. [PMID: 37776696 DOI: 10.1016/j.ortho.2023.100814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 10/02/2023]
Abstract
INTRODUCTION The aim was to design a meta-analysis evaluating the positron emission tomography (PET) uptake and cerebrospinal fluid (CSF), circulating levels of amyloid-β (Aβ), and tau proteins OSA group versus control group, as well as the association of these biomarkers with the severity of OSA. MATERIAL AND METHODS Four databases were searched until April 17, 2023, without any restrictions. The effect sizes were the standardized mean difference (SMD) along with a 95% confidence interval (CI). RESULTS A total of 21 articles were entered into the meta-analysis. The pooled SMDs of the CSF levels in OSA adults compared to controls were: -0.82 (P=0.004) for Aβ42, -1.13 (P<0.001) for Aβ40, 0.17 (P=0.23) for p-tau, 0.04 (P=0.65) for t-tau, 0.08 (P=0.89) for Aβ42/Aβ40 ratio, and 0.81 (P=0.001) for t-tau/Aβ42 ratio. The pooled SMD for the PET uptake of Aβ burden in OSA adults compared to controls was 0.30 (P=0.03). The pooled SMDs of the circulating levels in OSA adults compared to controls were: 0.67 (P=0.002) for Aβ42, 0.11 (P=0.82) for Aβ40, 0.35 (P=0.06) for p-tau, and 1.41(P=0.005) for t-tau. The pooled SMDs for levels of Aβ42, Aβ40, total Aβ, p-tau, t-tau, and Aβ42/Aβ40 ratio in severe OSA adults compared to mild/moderate OSA adults were -0.15 (P=0.33), 0.25 (P=35), 0.04 (P=87), -2.53 (P=0.24), -0.24 (P=0.52), and -0.28 (P=0.30), respectively. CONCLUSIONS The results indicated that CSF levels of Aβ42 and Aβ40 in OSA adults were significantly lower, but the CSF level of t-tau/Aβ42 ratio and PET Aβ burden uptake in OSA adults significantly were higher than in controls.
Collapse
Affiliation(s)
- Iman Mohammadi
- Oral and Maxillofacial Surgery Department, School of Dentistry, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran
| | - Mehrdad Adibparsa
- Department of Plastic Surgery, School of Medicine, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran
| | - Amir Najafi
- Oral and Maxillofacial Surgery Department, School of Dentistry, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran
| | - Mohammad Soroush Sehat
- Oral and Maxillofacial Surgery Department, School of Dentistry, Isfahan University of Medical Sciences, 81746-73461 Isfahan, Iran
| | - Masoud Sadeghi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, 67144-15185 Kermanshah, Iran.
| |
Collapse
|
61
|
Ye S, Wang P, Li Y, Wang W, Liu Q, Li Y. Halloysite nanotubes-loaded conductive polymer as substrate and label material for sensitive detection of amyloid-β protein by electrochemical immunosensor. Talanta 2023; 268:125345. [PMID: 39491948 DOI: 10.1016/j.talanta.2023.125345] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/05/2024]
Abstract
Amyloid-beta protein (Aβ) is a unique biomarker for Alzheimer's disease (AD). The sandwich-type electrochemical immunosensor, one of the key tools for detecting biomarkers, relies on a high-performance signal amplification approach to enhance its sensitivity. Ni/PdH nanodendrites (Ni/PdH NDs) have increased catalytic activity due to their unique interaction with palladium hydride and their nickel-rich surface, tunable shape and high specific surface area. Modified halloysite nanotubes (mHNT)-loaded with polypyrrole (PPy@mHNT) possess excellent dispersion and a large surface area. This enables the formation of a conductive network to prevent the accumulation of Ni/PdH NDs. Additionally, it exposes more electrocatalytic active centers, effectively amplifying electrical signals. By utilizing Ni/PdH@PPy@mHNT as the labeling material, it shows a consistent and remarkable electrocatalytic activity in H2O2 reduction, leading to signal amplification. The acid-etched HNT coated with polyaniline (PANI@eHNT) exhibits an exceptionally low background signal and outstanding conductivity. This not only accelerates electron transfer on the electrode surface, but also ensures the stable incubation of biomolecules post-amino grafting. Utilizing NH2-PANI@eHNT as a substrate material can guarantee stable biomolecule incubation, offer a stable sensing platform and enhance immunosensor performance. The signal can be amplified and the immunosensor's sensitivity can be raised through the efficient cooperation of the aforementioned nanomaterials. Under optimum circumstances, the electrochemical immunosensor had the lowest detection limit of 5.53 fg mL-1 and a linear range of 50 fg mL-1 to 100 ng mL-1. Based on the outstanding performance previously mentioned, this immunosensor is anticipated to aid in the early detection of AD.
Collapse
Affiliation(s)
- Sujie Ye
- School of Chemistry and Chemical Engineering, Shandong University of Technology, 255049, Zibo, PR China
| | - Ping Wang
- School of Chemistry and Chemical Engineering, Shandong University of Technology, 255049, Zibo, PR China.
| | - Yang Li
- School of Chemistry and Chemical Engineering, Shandong University of Technology, 255049, Zibo, PR China.
| | - Wenzhong Wang
- School of Chemistry and Chemical Engineering, Shandong University of Technology, 255049, Zibo, PR China
| | - Qing Liu
- School of Chemistry and Chemical Engineering, Shandong University of Technology, 255049, Zibo, PR China
| | - Yueyun Li
- School of Chemistry and Chemical Engineering, Shandong University of Technology, 255049, Zibo, PR China
| |
Collapse
|
62
|
Dubois B, von Arnim CAF, Burnie N, Bozeat S, Cummings J. Biomarkers in Alzheimer's disease: role in early and differential diagnosis and recognition of atypical variants. Alzheimers Res Ther 2023; 15:175. [PMID: 37833762 PMCID: PMC10571241 DOI: 10.1186/s13195-023-01314-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND Development of in vivo biomarkers has shifted the diagnosis of Alzheimer's disease (AD) from the later dementia stages of disease towards the earlier stages and has introduced the potential for pre-symptomatic diagnosis. The International Working Group recommends that AD diagnosis is restricted in the clinical setting to people with specific AD phenotypes and supportive biomarker findings. MAIN BODY In this review, we discuss the phenotypic presentation and use of biomarkers for the early diagnosis of typical and atypical AD and describe how this can support clinical decision making, benefit patient communication, and improve the patient journey. Early diagnosis is essential to optimize the benefits of available and emerging treatments. As atypical presentations of AD often mimic other dementias, differential diagnosis can be challenging and can be facilitated using AD biomarkers. However, AD biomarkers alone are not sufficient to confidently diagnose AD or predict disease progression and should be supplementary to clinical assessment to help inform the diagnosis of AD. CONCLUSIONS Use of AD biomarkers with incorporation of atypical AD phenotypes into diagnostic criteria will allow earlier diagnosis of patients with atypical clinical presentations that otherwise would have been misdiagnosed and treated inappropriately. Early diagnosis is essential to guide informed discussion, appropriate care and support, and individualized treatment. It is hoped that disease-modifying treatments will impact the underlying AD pathology; thus, determining the patient's AD phenotype will be a critical factor in guiding the therapeutic approach and the assessment of the effects of interventions.
Collapse
Affiliation(s)
- Bruno Dubois
- Assistance Publique-Hôpitaux de Paris (AP-HP), Memory and Alzheimer's Disease Institute, Sorbonne University, Paris, France
- Brain Institute, Sorbonne University, Paris, France
| | | | - Nerida Burnie
- General Practice, South West London CCG, London, UK
- London Dementia Clinical Network, London, UK
| | | | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| |
Collapse
|
63
|
Kwon HS, Kim JY, Koh SH, Choi SH, Lee EH, Jeong JH, Jang JW, Park KW, Kim EJ, Hong JY, Yoon SJ, Yoon B, Park HH, Han MH. Predicting cognitive stage transition using p-tau181, Centiloid, and other measures. Alzheimers Dement 2023; 19:4641-4650. [PMID: 36988152 PMCID: PMC12009171 DOI: 10.1002/alz.13054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/03/2023] [Accepted: 02/21/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND A combination of plasma phospho-tau (p-tau), amyloid beta (Aβ)-positron emission tomography (PET), brain magnetic resonance imaging, cognitive function tests, and other biomarkers might predict future cognitive decline. This study aimed to investigate the efficacy of combining these biomarkers in predicting future cognitive stage transitions within 3 years. METHODS Among the participants in the Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease (KBASE-V) study, 49 mild cognitive impairment (MCI) and 113 cognitively unimpaired (CU) participants with Aβ-PET and brain imaging data were analyzed. RESULTS Older age, increased plasma p-tau181, Aβ-PET positivity, and decreased semantic fluency were independently associated with cognitive stage transitions. Combining age, p-tau181, the Centiloid scale, semantic fluency, and hippocampal volume produced high predictive value in predicting future cognitive stage transition (area under the curve = 0.879). CONCLUSIONS Plasma p-tau181 and Centiloid scale alone or in combination with other biomarkers, might predict future cognitive stage transition in non-dementia patients. HIGHLIGHTS -Plasma p-tau181 and Centiloid scale might predict future cognitive stage transition. -Combining them or adding other biomarkers increased the predictive value. -Factors that independently associated with cognitive stage transition were demonstrated.
Collapse
Affiliation(s)
- Hyuk Sung Kwon
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Republic of Korea
| | - Ji Young Kim
- Department of Nuclear Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Republic of Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Republic of Korea
| | - Seong Hye Choi
- Department of Neurology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Eun-Hye Lee
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Republic of Korea
| | - Jee Hyang Jeong
- Department of Neurology, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Jae-Won Jang
- Department of Neurology, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Kyung Won Park
- Department of Neurology, Dong-A Medical Center, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - Jin Yong Hong
- Department of Neurology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Soo Jin Yoon
- Department of Neurology, Eulji University Hospital, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Bora Yoon
- Department of Neurology, Konyang University College of Medicine, Daejeon, Republic of Korea
| | - Hyun-Hee Park
- Department of Neurology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Republic of Korea
| | - Myung Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Republic of Korea
| |
Collapse
|
64
|
Petersen RC, Graf A, Brady C, De Santi S, Florian H, Landen J, Pontecorvo M, Randolph C, Sink KM, Carrillo MC, Weber CJ. Operationalizing selection criteria for clinical trials in Alzheimer's disease: Biomarker and clinical considerations. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12434. [PMID: 38023620 PMCID: PMC10655199 DOI: 10.1002/trc2.12434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023]
Abstract
Alzheimer's disease (AD) staging criteria lack standardized, empirical description. Well-defined AD staging criteria are an important consideration in protocol design, influencing a more standardized inclusion/exclusion criteria and defining what constitutes meaningful differentiation among the stages. However, many trials are being designed on the basis of biomarker features and the two need to be coordinated. The Alzheimer's Association Research Roundtable (AARR) Spring 2021 meeting discussed the implementation of preclinical AD staging criteria, and provided recommendations for how they may best be incorporated into clinical trials research. Discussion also included what currently available tools for global clinical trials may best define populations in preclinical AD trials, and if are we able to differentiate preclinical from clinical stages of the disease. Well-defined AD staging criteria are key to improving early detection, diagnostics, clinical trial enrollment, and identifying statistically significant clinical changes, and researchers discussed how emerging blood biomarkers may help with more efficient screening in preclinical stages.
Collapse
Affiliation(s)
| | - Ana Graf
- Novartis Pharma AGBaselSwitzerland
| | - Chris Brady
- WCG Clinical Endpoint Solutions, PrincetonNew JerseyUSA
| | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Giuffrè GM, Quaranta D, Costantini EM, Citro S, Martellacci N, De Ninno G, Vita MG, Guglielmi V, Rossini PM, Calabresi P, Marra C. Cerebrospinal fluid neurofilament light chain and total-tau as biomarkers of neurodegeneration in Alzheimer's disease and frontotemporal dementia. Neurobiol Dis 2023; 186:106267. [PMID: 37652185 DOI: 10.1016/j.nbd.2023.106267] [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: 04/02/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023] Open
Abstract
INTRODUCTION CSF Neurofilament light chain(NfL) is a promising biomarker of neurodegeneration, but its utility in discriminating between Alzheimer's disease(AD) and frontotemporal dementia(FTD) is limited. METHODS 105 patients with clinical-biological diagnosis of mild cognitive impairment(MCI) due to AD (N = 72) or clinical diagnosis of FTD (N = 33) underwent neuropsychological assessment and CSF Aβ42/40, p-tau181, total-tau and NfL quantification. Group comparisons, correlations between continuous variables and ROC curve analysis were carried out to assess NfL role in discriminating between MCI due to AD and FTD, exploring the associations between NfL, ATN biomarkers and neuropsychological measures. RESULTS NfL levels were significantly lower in the AD group, while levels of total-tau were higher. In the FTD group, significant correlations were found between NfL, p-tau181 and total-tau, and between NfL and cognitive performances. In the AD group, NfL levels were directly correlated with total-tau and p-tau181; Aβ42/40 ratio was inversely correlated with total-tau and p-tau181, but not with NfL. Moreover, p-tau181 and t-tau levels were found to be associated with episodic memory and lexical-semantic impairment. Total-tau/NfL ratio differentiated prodromal-AD from FTD with an AUC of 0.951, higher than the individual measures. DISCUSSION & CONCLUSIONS The results support that NfL and total-tau levels reflect distinct pathophysiological neurodegeneration mechanisms, independent and dependent of Aβ pathology, respectively, Combining them may enhance both markers reliability, their ratio showing high accuracy in distinguishing MCI due to AD from FTD. Moreover, our results revealed associations between NfL and disease severity in FTD and between tauopathy and episodic memory and lexical-semantic impairment in prodromal-AD.
Collapse
Affiliation(s)
- Guido Maria Giuffrè
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Davide Quaranta
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy.
| | | | - Salvatore Citro
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Noemi Martellacci
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Grazia De Ninno
- UOC of Chemistry, Biochemistry and Clinical Molecular Biology - Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Gabriella Vita
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Valeria Guglielmi
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Paolo Maria Rossini
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, Rome, Italy
| | - Paolo Calabresi
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Camillo Marra
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
66
|
Ferreira PCL, Zhang Y, Snitz B, Chang CCH, Bellaver B, Jacobsen E, Kamboh MI, Zetterberg H, Blennow K, Pascoal TA, Villemagne VL, Ganguli M, Karikari TK. Plasma biomarkers identify older adults at risk of Alzheimer's disease and related dementias in a real-world population-based cohort. Alzheimers Dement 2023; 19:4507-4519. [PMID: 36876954 PMCID: PMC10480336 DOI: 10.1002/alz.12986] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 03/07/2023]
Abstract
INTRODUCTION Plasma biomarkers-cost effective, non-invasive indicators of Alzheimer's disease (AD) and related disorders (ADRD)-have largely been studied in clinical research settings. Here, we examined plasma biomarker profiles and their associated factors in a population-based cohort to determine whether they could identify an at-risk group, independently of brain and cerebrospinal fluid biomarkers. METHODS We measured plasma phosphorylated tau181 (p-tau181), neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), and amyloid beta (Aβ)42/40 ratio in 847 participants from a population-based cohort in southwestern Pennsylvania. RESULTS K-medoids clustering identified two distinct plasma Aβ42/40 modes, further categorizable into three biomarker profile groups: normal, uncertain, and abnormal. In different groups, plasma p-tau181, NfL, and GFAP were inversely correlated with Aβ42/40, Clinical Dementia Rating, and memory composite score, with the strongest associations in the abnormal group. DISCUSSION Abnormal plasma Aβ42/40 ratio identified older adult groups with lower memory scores, higher dementia risks, and higher ADRD biomarker levels, with potential implications for population screening. HIGHLIGHTS Population-based plasma biomarker studies are lacking, particularly in cohorts without cerebrospinal fluid or neuroimaging data. In the Monongahela-Youghiogheny Healthy Aging Team study (n = 847), plasma biomarkers associated with worse memory and Clinical Dementia Rating (CDR), apolipoprotein E ε4, and greater age. Plasma amyloid beta (Aβ)42/40 ratio levels allowed clustering participants into abnormal, uncertain, and normal groups. Plasma Aβ42/40 correlated differently with neurofilament light chain, glial fibrillary acidic protein, phosphorylated tau181, memory composite, and CDR in each group. Plasma biomarkers can enable relatively affordable and non-invasive community screening for evidence of Alzheimer's disease and related disorders pathophysiology.
Collapse
Affiliation(s)
- Pamela C. L Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Yingjin Zhang
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Beth Snitz
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Chung-Chou H. Chang
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Erin Jacobsen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - M. Ilyas Kamboh
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1N 3BG, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, HKG, China
- UW Department of Medicine, School of Medicine and Public Health, Madison, WI, 53726, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, 431 41, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, 431 41, Sweden
| | - Tharick A. Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Victor L. Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Mary Ganguli
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Thomas K. Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, 431 41, Sweden
| |
Collapse
|
67
|
Hickman LB, Stern JM, Silverman DHS, Salamon N, Vossel K. Clinical, imaging, and biomarker evidence of amyloid- and tau-related neurodegeneration in late-onset epilepsy of unknown etiology. Front Neurol 2023; 14:1241638. [PMID: 37830092 PMCID: PMC10565489 DOI: 10.3389/fneur.2023.1241638] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/05/2023] [Indexed: 10/14/2023] Open
Abstract
Accumulating evidence suggests amyloid and tau-related neurodegeneration may play a role in development of late-onset epilepsy of unknown etiology (LOEU). In this article, we review recent evidence that epilepsy may be an initial manifestation of an amyloidopathy or tauopathy that precedes development of Alzheimer's disease (AD). Patients with LOEU demonstrate an increased risk of cognitive decline, and patients with AD have increased prevalence of preceding epilepsy. Moreover, investigations of LOEU that use CSF biomarkers and imaging techniques have identified preclinical neurodegeneration with evidence of amyloid and tau deposition. Overall, findings to date suggest a relationship between acquired, non-lesional late-onset epilepsy and amyloid and tau-related neurodegeneration, which supports that preclinical or prodromal AD is a distinct etiology of late-onset epilepsy. We propose criteria for assessing elevated risk of developing dementia in patients with late-onset epilepsy utilizing clinical features, available imaging techniques, and biomarker measurements. Further research is needed to validate these criteria and assess optimal treatment strategies for patients with probable epileptic preclinical AD and epileptic prodromal AD.
Collapse
Affiliation(s)
- L. Brian Hickman
- Mary S. Easton Center for Alzheimer’s Research and Care, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Neurology, UCLA Seizure Disorder Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - John M. Stern
- Department of Neurology, UCLA Seizure Disorder Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Daniel H. S. Silverman
- Mary S. Easton Center for Alzheimer’s Research and Care, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Noriko Salamon
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Keith Vossel
- Mary S. Easton Center for Alzheimer’s Research and Care, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
68
|
Teipel SJ, Dyrba M, Levin F, Altenstein S, Berger M, Beyle A, Brosseron F, Buerger K, Burow L, Dobisch L, Ewers M, Fliessbach K, Frommann I, Glanz W, Goerss D, Gref D, Hansen N, Heneka MT, Incesoy EI, Janowitz D, Keles D, Kilimann I, Laske C, Lohse A, Munk MH, Perneczky R, Peters O, Preis L, Priller J, Rostamzadeh A, Roy N, Schmid M, Schneider A, Spottke A, Spruth EJ, Wiltfang J, Düzel E, Jessen F, Kleineidam L, Wagner M, for the DELCODE study group and the Alzheimer’s Disease Neuroimaging Initiative. Cognitive Trajectories in Preclinical and Prodromal Alzheimer's Disease Related to Amyloid Status and Brain Atrophy: A Bayesian Approach. J Alzheimers Dis Rep 2023; 7:1055-1076. [PMID: 37849637 PMCID: PMC10578328 DOI: 10.3233/adr-230027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/22/2023] [Indexed: 10/19/2023] Open
Abstract
Background Cognitive decline is a key outcome of clinical studies in Alzheimer's disease (AD). Objective To determine effects of global amyloid load as well as hippocampus and basal forebrain volumes on longitudinal rates and practice effects from repeated testing of domain specific cognitive change in the AD spectrum, considering non-linear effects and heterogeneity across cohorts. Methods We included 1,514 cases from three cohorts, ADNI, AIBL, and DELCODE, spanning the range from cognitively normal people to people with subjective cognitive decline and mild cognitive impairment (MCI). We used generalized Bayesian mixed effects analysis of linear and polynomial models of amyloid and volume effects in time. Robustness of effects across cohorts was determined using Bayesian random effects meta-analysis. Results We found a consistent effect of amyloid and hippocampus volume, but not of basal forebrain volume, on rates of memory change across the three cohorts in the meta-analysis. Effects for amyloid and volumetric markers on executive function were more heterogeneous. We found practice effects in memory and executive performance in amyloid negative cognitively normal controls and MCI cases, but only to a smaller degree in amyloid positive controls and not at all in amyloid positive MCI cases. Conclusions We found heterogeneity between cohorts, particularly in effects on executive functions. Initial increases in cognitive performance in amyloid negative, but not in amyloid positive MCI cases and controls may reflect practice effects from repeated testing that are lost with higher levels of cerebral amyloid.
Collapse
Affiliation(s)
- Stefan J Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Martin Dyrba
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | - Fedor Levin
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Moritz Berger
- Institute for Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Aline Beyle
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Frederic Brosseron
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Lena Burow
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Klaus Fliessbach
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- University of Bonn Medical Center, Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
| | - Ingo Frommann
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- University of Bonn Medical Center, Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Doreen Goerss
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Daria Gref
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany
| | - Michael T. Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Enise I. Incesoy
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
- Department for Psychiatry and Psychotherapy, University Clinic Magdeburg, Magdeburg, Germany pGerman Center for Neurodegenerative Diseases (DZNE), T¨ubingen, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Deniz Keles
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), T¨ubingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of T¨ubingen, T¨ubingen, Germany
| | - Andrea Lohse
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Matthias H. Munk
- German Center for Neurodegenerative Diseases (DZNE), T¨ubingen, Germany
- Department of Psychiatry and Psychotherapy, University of T¨ubingen, T¨ubingen, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Lukas Preis
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
- School of Medicine, Technical University of Munich; Department of Psychiatry and Psychotherapy, Munich, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Ayda Rostamzadeh
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
| | - Nina Roy
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Matthias Schmid
- Institute for Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Anja Schneider
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- University of Bonn Medical Center, Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
| | - Annika Spottke
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Frank Jessen
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
| | - Luca Kleineidam
- University of Bonn Medical Center, Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
| | - Michael Wagner
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- University of Bonn Medical Center, Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
| | | |
Collapse
|
69
|
Jones KT, Gallen CL, Ostrand AE, Rojas JC, Wais P, Rini J, Chan B, Lago AL, Boxer A, Zhao M, Gazzaley A, Zanto TP. Gamma neuromodulation improves episodic memory and its associated network in amnestic mild cognitive impairment: a pilot study. Neurobiol Aging 2023; 129:72-88. [PMID: 37276822 PMCID: PMC10583532 DOI: 10.1016/j.neurobiolaging.2023.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 06/07/2023]
Abstract
Amnestic mild cognitive impairment (aMCI) is a predementia stage of Alzheimer's disease associated with dysfunctional episodic memory and limited treatment options. We aimed to characterize feasibility, clinical, and biomarker effects of noninvasive neurostimulation for aMCI. 13 individuals with aMCI received eight 60-minute sessions of 40-Hz (gamma) transcranial alternating current stimulation (tACS) targeting regions related to episodic memory processing. Feasibility, episodic memory, and plasma Alzheimer's disease biomarkers were assessed. Neuroplastic changes were characterized by resting-state functional connectivity (RSFC) and neuronal excitatory/inhibitory balance. Gamma tACS was feasible and aMCI participants demonstrated improvement in multiple metrics of episodic memory, but no changes in biomarkers. Improvements in episodic memory were most pronounced in participants who had the highest modeled tACS-induced electric fields and exhibited the greatest changes in RSFC. Increased RSFC was also associated with greater hippocampal excitability and higher baseline white matter integrity. This study highlights initial feasibility and the potential of gamma tACS to rescue episodic memory in an aMCI population by modulating connectivity and excitability within an episodic memory network.
Collapse
Affiliation(s)
- Kevin T Jones
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA.
| | - Courtney L Gallen
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - Avery E Ostrand
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - Julio C Rojas
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Peter Wais
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - James Rini
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA
| | - Brandon Chan
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Argentina Lario Lago
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Adam Boxer
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Weill Institute for Neurosciences, Memory and Aging Center, University of California-San Francisco, San Francisco, CA
| | - Min Zhao
- Departments of Ophthalmology and Vision Science and Dermatology, Institute for Regenerative Cures, University of California-Davis, Davis, CA
| | - Adam Gazzaley
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA; Departments of Physiology and Psychiatry, University of California-San Francisco, San Francisco, CA
| | - Theodore P Zanto
- Department of Neurology, University of California-San Francisco, San Francisco, CA; Neuroscape, University of California-San Francisco, San Francisco, CA.
| |
Collapse
|
70
|
Nisenbaum L, Martone R, Chen T, Rajagovindan R, Dent G, Beaver J, Rubel C, Racine A, He P, Harrison K, Dean R, Vandijck M, Haeberlein SB. CSF biomarker concordance with amyloid PET in Phase 3 studies of aducanumab. Alzheimers Dement 2023; 19:3379-3388. [PMID: 36795603 DOI: 10.1002/alz.12919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 02/17/2023]
Abstract
INTRODUCTION We assessed the use of cerebrospinal fluid (CSF) biomarkers as an alternative to positron emission tomography (PET) for brain amyloid beta (Aβ) pathology confirmation in the EMERGE and ENGAGE clinical trials. METHODS EMERGE and ENGAGE were randomized, placebo-controlled, Phase 3 trials of aducanumab in participants with early Alzheimer's disease. Concordance between CSF biomarkers (Aβ42, Aβ40, phosphorylated tau 181, and total tau) and amyloid PET status (visual read) at screening was examined. RESULTS Robust concordance between CSF biomarkers and amyloid PET visual status was observed (for Aβ42/Aβ40, AUC: 0.90; 95% CI: 0.83-0.97; p < 0.0001), confirming CSF biomarkers as a reliable alternative to amyloid PET in these studies. Compared with single CSF biomarkers, CSF biomarker ratios showed better agreement with amyloid PET visual reads, demonstrating high diagnostic accuracy. DISCUSSION These analyses add to the growing body of evidence supporting CSF biomarkers as reliable alternatives to amyloid PET imaging for brain Aβ pathology confirmation. HIGHLIGHTS CSF biomarkers and amyloid PET concordance were assessed in Ph3 aducanumab trials. Robust concordance between CSF biomarkers and amyloid PET was observed. CSF biomarker ratios increased diagnostic accuracy over single CSF biomarkers. CSF Aβ42/Aβ40 demonstrated high concordance with amyloid PET. Results support CSF biomarker testing as a reliable alternative to amyloid PET.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ping He
- Biogen, Cambridge, Massachusetts, USA
| | | | - Robert Dean
- Robert A. Dean Consulting, LLC, Indianapolis, Indiana, USA
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
71
|
Deming Y, Vasiljevic E, Morrow A, Miao J, Van Hulle C, Jonaitis E, Ma Y, Whitenack V, Kollmorgen G, Wild N, Suridjan I, Shaw LM, Asthana S, Carlsson CM, Johnson SC, Zetterberg H, Blennow K, Bendlin BB, Lu Q, Engelman CD. Neuropathology-based APOE genetic risk score better quantifies Alzheimer's risk. Alzheimers Dement 2023; 19:3406-3416. [PMID: 36795776 PMCID: PMC10427737 DOI: 10.1002/alz.12990] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 02/17/2023]
Abstract
INTRODUCTION Apolipoprotein E (APOE) ε4-carrier status or ε4 allele count are included in analyses to account for the APOE genetic effect on Alzheimer's disease (AD); however, this does not account for protective effects of APOE ε2 or heterogeneous effect of ε2, ε3, and ε4 haplotypes. METHODS We leveraged results from an autopsy-confirmed AD study to generate a weighted risk score for APOE (APOE-npscore). We regressed cerebrospinal fluid (CSF) amyloid and tau biomarkers on APOE variables from the Wisconsin Registry for Alzheimer's Prevention (WRAP), Wisconsin Alzheimer's Disease Research Center (WADRC), and Alzheimer's Disease Neuroimaging Initiative (ADNI). RESULTS The APOE-npscore explained more variance and provided a better model fit for all three CSF measures than APOE ε4-carrier status and ε4 allele count. These findings were replicated in ADNI and observed in subsets of cognitively unimpaired (CU) participants. DISCUSSION The APOE-npscore reflects the genetic effect on neuropathology and provides an improved method to account for APOE in AD-related analyses.
Collapse
Affiliation(s)
- Yuetiva Deming
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Eva Vasiljevic
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Autumn Morrow
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jiacheng Miao
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Carol Van Hulle
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Erin Jonaitis
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yue Ma
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Vanessa Whitenack
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sanjay Asthana
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Geriatric Research Education and Clinical Center, Madison, Wisconsin, USA
| | - Cynthia M Carlsson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Geriatric Research Education and Clinical Center, Madison, Wisconsin, USA
| | - Sterling C Johnson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Geriatric Research Education and Clinical Center, Madison, Wisconsin, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Barbara B Bendlin
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Qiongshi Lu
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Corinne D Engelman
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
72
|
Johnson ECB, Bian S, Haque RU, Carter EK, Watson CM, Gordon BA, Ping L, Duong DM, Epstein MP, McDade E, Barthélemy NR, Karch CM, Xiong C, Cruchaga C, Perrin RJ, Wingo AP, Wingo TS, Chhatwal JP, Day GS, Noble JM, Berman SB, Martins R, Graff-Radford NR, Schofield PR, Ikeuchi T, Mori H, Levin J, Farlow M, Lah JJ, Haass C, Jucker M, Morris JC, Benzinger TLS, Roberts BR, Bateman RJ, Fagan AM, Seyfried NT, Levey AI. Cerebrospinal fluid proteomics define the natural history of autosomal dominant Alzheimer's disease. Nat Med 2023; 29:1979-1988. [PMID: 37550416 PMCID: PMC10427428 DOI: 10.1038/s41591-023-02476-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/27/2023] [Indexed: 08/09/2023]
Abstract
Alzheimer's disease (AD) pathology develops many years before the onset of cognitive symptoms. Two pathological processes-aggregation of the amyloid-β (Aβ) peptide into plaques and the microtubule protein tau into neurofibrillary tangles (NFTs)-are hallmarks of the disease. However, other pathological brain processes are thought to be key disease mediators of Aβ plaque and NFT pathology. How these additional pathologies evolve over the course of the disease is currently unknown. Here we show that proteomic measurements in autosomal dominant AD cerebrospinal fluid (CSF) linked to brain protein coexpression can be used to characterize the evolution of AD pathology over a timescale spanning six decades. SMOC1 and SPON1 proteins associated with Aβ plaques were elevated in AD CSF nearly 30 years before the onset of symptoms, followed by changes in synaptic proteins, metabolic proteins, axonal proteins, inflammatory proteins and finally decreases in neurosecretory proteins. The proteome discriminated mutation carriers from noncarriers before symptom onset as well or better than Aβ and tau measures. Our results highlight the multifaceted landscape of AD pathophysiology and its temporal evolution. Such knowledge will be critical for developing precision therapeutic interventions and biomarkers for AD beyond those associated with Aβ and tau.
Collapse
Affiliation(s)
- Erik C B Johnson
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Shijia Bian
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Rafi U Haque
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - E Kathleen Carter
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Caroline M Watson
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Lingyan Ping
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Duc M Duong
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael P Epstein
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | | | - Celeste M Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Chengjie Xiong
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
- Division of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Richard J Perrin
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, USA
| | - Aliza P Wingo
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA
- Division of Mental Health, Atlanta VA Medical Center, Atlanta, GA, USA
| | - Thomas S Wingo
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jasmeer P Chhatwal
- Massachusetts General and Brigham & Women's Hospitals, Harvard Medical School, Boston, MA, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and GH Sergievsky Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah B Berman
- Departments of Neurology and Clinical and Translational Science, Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ralph Martins
- Edith Cowan University, Perth, Western Australia, Australia
| | | | - Peter R Schofield
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroshi Mori
- Osaka Metropolitan University Medical School, Nagaoka Sutoku University, Nagaoka, Japan
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - James J Lah
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Metabolic Biochemistry, Biomedical Center (BMC), Ludwig-Maximilians University, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - John C Morris
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Blaine R Roberts
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I Levey
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
73
|
Zhang S, Dong H, Bian J, Li D, Liu C. Targeting amyloid proteins for clinical diagnosis of neurodegenerative diseases. FUNDAMENTAL RESEARCH 2023; 3:505-519. [PMID: 38933553 PMCID: PMC11197785 DOI: 10.1016/j.fmre.2022.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
Abnormal aggregation and accumulation of pathological amyloid proteins such as amyloid-β, Tau, and α-synuclein play key pathological roles and serve as histological hallmarks in different neurodegenerative diseases (NDs) such as Alzheimer's disease (AD) and Parkinson's disease (PD). In addition, various post-translational modifications (PTMs) have been identified on pathological amyloid proteins and are subjected to change during disease progression. Given the central role of amyloid proteins in NDs, tremendous efforts have been made to develop amyloid-targeting strategies for clinical diagnosis and molecular classification of NDs. In this review, we summarize two major strategies for targeting amyloid aggregates, with a focus on the trials in AD diagnosis. The first strategy is a positron emission tomography (PET) scan of protein aggregation in the brain. We mainly focus on introducing the development of small-molecule PET tracers for specifically recognizing pathological amyloid fibrils. The second strategy is the detection of PTM biomarkers on amyloid proteins in cerebrospinal fluid and plasma. We discuss the pathological roles of different PTMs in diseases and how we can use the PTM profile of amyloid proteins for clinical diagnosis. Finally, we point out the potential technical challenges of these two strategies, and outline other potential strategies, as well as a combination of multiple strategies, for molecular diagnosis of NDs.
Collapse
Affiliation(s)
- Shenqing Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hui Dong
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Jiang Bian
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
74
|
Xu Y, Jiang H, Zhu B, Cao M, Feng T, Sun Z, Du G, Zhao Z. Advances and applications of fluids biomarkers in diagnosis and therapeutic targets of Alzheimer's disease. CNS Neurosci Ther 2023. [PMID: 37144603 DOI: 10.1111/cns.14238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/25/2023] [Accepted: 04/12/2023] [Indexed: 05/06/2023] Open
Abstract
AIMS Alzheimer's disease (AD) is a neurodegenerative disease with challenging early diagnosis and effective treatments due to its complex pathogenesis. AD patients are often diagnosed after the appearance of the typical symptoms, thereby delaying the best opportunity for effective measures. Biomarkers could be the key to resolving the challenge. This review aims to provide an overview of application and potential value of AD biomarkers in fluids, including cerebrospinal fluid, blood, and saliva, in diagnosis and treatment. METHODS A comprehensive search of the relevant literature was conducted to summarize potential biomarkers for AD in fluids. The paper further explored the biomarkers' utility in disease diagnosis and drug target development. RESULTS Research on biomarkers mainly focused on amyloid-β (Aβ) plaques, Tau protein abnormal phosphorylation, axon damage, synaptic dysfunction, inflammation, and related hypotheses associated with AD mechanisms. Aβ42 , total Tau (t-Tau), and phosphorylated Tau (p-Tau), have been endorsed for their diagnostic and predictive capability. However, other biomarkers remain controversial. Drugs targeting Aβ have shown some efficacy and those that target BACE1 and Tau are still undergoing development. CONCLUSION Fluid biomarkers hold considerable potential in the diagnosis and drug development of AD. However, improvements in sensitivity and specificity, and approaches for managing sample impurities, need to be addressed for better diagnosis.
Collapse
Affiliation(s)
- Yanan Xu
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- School of Pharmacy, Capital Medical University, Beijing, China
| | - Hailun Jiang
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhu
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mingnan Cao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Feng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhongshi Sun
- Department of Pharmacy, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhigang Zhao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- School of Pharmacy, Capital Medical University, Beijing, China
| |
Collapse
|
75
|
Hansson O, Blennow K, Zetterberg H, Dage J. Blood biomarkers for Alzheimer's disease in clinical practice and trials. NATURE AGING 2023; 3:506-519. [PMID: 37202517 PMCID: PMC10979350 DOI: 10.1038/s43587-023-00403-3] [Citation(s) in RCA: 183] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Blood-based biomarkers hold great promise to revolutionize the diagnostic and prognostic work-up of Alzheimer's disease (AD) in clinical practice. This is very timely, considering the recent development of anti-amyloid-β (Aβ) immunotherapies. Several assays for measuring phosphorylated tau (p-tau) in plasma exhibit high diagnostic accuracy in distinguishing AD from all other neurodegenerative diseases in patients with cognitive impairment. Prognostic models based on plasma p-tau levels can also predict future development of AD dementia in patients with mild cognitive complaints. The use of such high-performing plasma p-tau assays in the clinical practice of specialist memory clinics would reduce the need for more costly investigations involving cerebrospinal fluid samples or positron emission tomography. Indeed, blood-based biomarkers already facilitate identification of individuals with pre-symptomatic AD in the context of clinical trials. Longitudinal measurements of such biomarkers will also improve the detection of relevant disease-modifying effects of new drugs or lifestyle interventions.
Collapse
Affiliation(s)
- Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Lund, Sweden.
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for 27 Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeffrey Dage
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
76
|
Gertje EC, Janelidze S, van Westen D, Cullen N, Stomrud E, Palmqvist S, Hansson O, Mattsson-Carlgren N. Associations Between CSF Markers of Inflammation, White Matter Lesions, and Cognitive Decline in Individuals Without Dementia. Neurology 2023; 100:e1812-e1824. [PMID: 36882326 PMCID: PMC10136007 DOI: 10.1212/wnl.0000000000207113] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 01/11/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Small vessel disease (SVD) and neuroinflammation both occur in Alzheimer disease (AD) and other neurodegenerative diseases. It is unclear whether these processes are related or independent mechanisms in AD, especially in the early stages of disease. We therefore investigated the association between white matter lesions (WML; the most common manifestation of SVD) and CSF biomarkers of neuroinflammation and their effects on cognition in a population without dementia. METHODS Individuals without dementia from the Swedish BioFINDER study were included. The CSF was analyzed for proinflammatory markers (interleukin [IL]-6 and IL-8), cytokines (IL-7, IL-15, and IL-16), chemokines (interferon γ-induced protein 10, monocyte chemoattractant protein 1), markers of vascular injury (soluble intercellular adhesion molecule 1, soluble vascular adhesion molecule 1), and markers of angiogenesis (placental growth factor [PlGF], soluble fms-related tyrosine kinase 1 [sFlt-1], vascular endothelial growth factors [VEGF-A and VEFG-D]), and amyloid β (Aβ)42 Aβ40, and p-tau217. WML volumes were determined at baseline and longitudinally over 6 years. Cognition was measured at baseline and follow-up over 8 years. Linear regression models were used to test associations. RESULTS A total of 495 cognitively unimpaired (CU) elderly individuals and 247 patients with mild cognitive impairment (MCI) were included. There was significant worsening in cognition over time, measured by Mini-Mental State Examination, Clinical Dementia Rating, and modified preclinical Alzheimer composite score in CU individuals and patients with MCI, with more rapid worsening in MCI for all cognitive tests. At baseline, higher levels of PlGF (β = 0.156, p < 0.001), lower levels of sFlt-1 (β = -0.086, p = 0.003), and higher levels of IL-8 (β = 0.07, p = 0.030) were associated with more WML in CU individuals. In those with MCI, higher levels of PlGF (β = 0.172, p = 0.001), IL-16 (β = 0.125, p = 0.001), IL-8 (β = 0.096, p = 0.013), IL-6 (β = 0.088, p = 0.023), VEGF-A (β = 0.068, p = 0.028), and VEGF-D (β = 0.082, p = 0.028) were associated with more WML. PlGF was the only biomarker that was associated with WML independent of Aβ status and cognitive impairment. Longitudinal analyses of cognition showed independent effects of CSF inflammatory markers and WML on longitudinal cognition, especially in people without cognitive impairment at baseline. DISCUSSION Most neuroinflammatory CSF biomarkers were associated with WML in individuals without dementia. Our findings especially highlight a role for PlGF, which was associated with WML independent of Aβ status and cognitive impairment.
Collapse
Affiliation(s)
- Eske Christiane Gertje
- From the Clinical Memory Research Unit (E.C.G., S.J., N.C., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences Malmö, Lund University; Department of Internal Medicine (E.C.G.), Skåne University Hospital, Lund; Diagnostic Radiology (D.v.W.), Department of Clinical Sciences Lund, Lund University; Imaging and Function (D.v.W.), Skåne University Hospital, Lund; Memory Clinic (N.C., N.M.-C.), Skåne University Hospital, Malmö; Department of Clinical Sciences Lund, Neurology (E.S., S.P., O.H.), Lund University, Skåne University Hospital; and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden.
| | - Shorena Janelidze
- From the Clinical Memory Research Unit (E.C.G., S.J., N.C., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences Malmö, Lund University; Department of Internal Medicine (E.C.G.), Skåne University Hospital, Lund; Diagnostic Radiology (D.v.W.), Department of Clinical Sciences Lund, Lund University; Imaging and Function (D.v.W.), Skåne University Hospital, Lund; Memory Clinic (N.C., N.M.-C.), Skåne University Hospital, Malmö; Department of Clinical Sciences Lund, Neurology (E.S., S.P., O.H.), Lund University, Skåne University Hospital; and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Danielle van Westen
- From the Clinical Memory Research Unit (E.C.G., S.J., N.C., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences Malmö, Lund University; Department of Internal Medicine (E.C.G.), Skåne University Hospital, Lund; Diagnostic Radiology (D.v.W.), Department of Clinical Sciences Lund, Lund University; Imaging and Function (D.v.W.), Skåne University Hospital, Lund; Memory Clinic (N.C., N.M.-C.), Skåne University Hospital, Malmö; Department of Clinical Sciences Lund, Neurology (E.S., S.P., O.H.), Lund University, Skåne University Hospital; and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Nicholas Cullen
- From the Clinical Memory Research Unit (E.C.G., S.J., N.C., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences Malmö, Lund University; Department of Internal Medicine (E.C.G.), Skåne University Hospital, Lund; Diagnostic Radiology (D.v.W.), Department of Clinical Sciences Lund, Lund University; Imaging and Function (D.v.W.), Skåne University Hospital, Lund; Memory Clinic (N.C., N.M.-C.), Skåne University Hospital, Malmö; Department of Clinical Sciences Lund, Neurology (E.S., S.P., O.H.), Lund University, Skåne University Hospital; and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Erik Stomrud
- From the Clinical Memory Research Unit (E.C.G., S.J., N.C., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences Malmö, Lund University; Department of Internal Medicine (E.C.G.), Skåne University Hospital, Lund; Diagnostic Radiology (D.v.W.), Department of Clinical Sciences Lund, Lund University; Imaging and Function (D.v.W.), Skåne University Hospital, Lund; Memory Clinic (N.C., N.M.-C.), Skåne University Hospital, Malmö; Department of Clinical Sciences Lund, Neurology (E.S., S.P., O.H.), Lund University, Skåne University Hospital; and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Sebastian Palmqvist
- From the Clinical Memory Research Unit (E.C.G., S.J., N.C., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences Malmö, Lund University; Department of Internal Medicine (E.C.G.), Skåne University Hospital, Lund; Diagnostic Radiology (D.v.W.), Department of Clinical Sciences Lund, Lund University; Imaging and Function (D.v.W.), Skåne University Hospital, Lund; Memory Clinic (N.C., N.M.-C.), Skåne University Hospital, Malmö; Department of Clinical Sciences Lund, Neurology (E.S., S.P., O.H.), Lund University, Skåne University Hospital; and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Oskar Hansson
- From the Clinical Memory Research Unit (E.C.G., S.J., N.C., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences Malmö, Lund University; Department of Internal Medicine (E.C.G.), Skåne University Hospital, Lund; Diagnostic Radiology (D.v.W.), Department of Clinical Sciences Lund, Lund University; Imaging and Function (D.v.W.), Skåne University Hospital, Lund; Memory Clinic (N.C., N.M.-C.), Skåne University Hospital, Malmö; Department of Clinical Sciences Lund, Neurology (E.S., S.P., O.H.), Lund University, Skåne University Hospital; and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Niklas Mattsson-Carlgren
- From the Clinical Memory Research Unit (E.C.G., S.J., N.C., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences Malmö, Lund University; Department of Internal Medicine (E.C.G.), Skåne University Hospital, Lund; Diagnostic Radiology (D.v.W.), Department of Clinical Sciences Lund, Lund University; Imaging and Function (D.v.W.), Skåne University Hospital, Lund; Memory Clinic (N.C., N.M.-C.), Skåne University Hospital, Malmö; Department of Clinical Sciences Lund, Neurology (E.S., S.P., O.H.), Lund University, Skåne University Hospital; and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| |
Collapse
|
77
|
Honig LS, Kang MS, Lee AJ, Reyes-Dumeyer D, Piriz A, Soriano B, Franco Y, Coronado ZD, Recio P, Mejía DR, Medrano M, Lantigua RA, Teich AF, Dage JL, Mayeux R. Evaluation of Plasma Biomarkers for A/T/N Classification of Alzheimer Disease Among Adults of Caribbean Hispanic Ethnicity. JAMA Netw Open 2023; 6:e238214. [PMID: 37079306 PMCID: PMC10119732 DOI: 10.1001/jamanetworkopen.2023.8214] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/20/2023] [Indexed: 04/21/2023] Open
Abstract
Importance Cerebrospinal fluid (CSF) and plasma biomarkers can detect biological evidence of Alzheimer disease (AD), but their use in low-resource environments and among minority ethnic groups is limited. Objective To assess validated plasma biomarkers for AD among adults of Caribbean Hispanic ethnicity. Design, Setting, and Participants In this decision analytical modeling study, adults were recruited between January 1, 2018, and April 30, 2022, and underwent detailed clinical assessments and venipuncture. A subsample of participants also consented to lumbar puncture. Established CSF cut points were used to define AD biomarker-positive status, allowing determination of optimal cut points for plasma biomarkers in the same individuals. The performance of a panel of 6 plasma biomarkers was then assessed with respect to the entire group. Data analysis was performed in January 2023. Main Outcomes and Measures Main outcomes were the association of plasma biomarkers amyloid-β 1-42 (Aβ42), amyloid-β 1-40 (Aβ40), total tau (T-tau), phosphorylated tau181 (P-tau181), glial fibrillary acidic protein (GFAP), and neurofilament light chain (NfL) with AD diagnosis. These biomarkers allow assessment of amyloid (A), neurofibrillary degeneration (T), and neurodegeneration (N) aspects of AD. Statistical analyses performed included receiver operating characteristics, Pearson and Spearman correlations, t tests, and Wilcoxon rank-sum, chi-square, and Fisher exact tests. Exposures Exposures included age, sex, education, country of residence, apolipoprotein-ε4 (APOE-ε4) allele number, serum creatinine, blood urea nitrogen, and body mass index. Results This study included 746 adults. Participants had a mean (SD) age of 71.0 (7.8) years, 480 (64.3%) were women, and 154 (20.6%) met clinical criteria for AD. Associations were observed between CSF and plasma P-tau181 (r = .47 [95% CI, 0.32-0.60]), NfL (r = 0.57 [95% CI, 0.44-0.68]), and P-tau181/Aβ42 (r = 0.44 [95% CI, 0.29-0.58]). For AD defined by CSF biomarkers, plasma P-tau181 and P-tau181/Aβ42 provided biological evidence of AD. Among individuals judged to be clinically healthy without dementia, biomarker-positive status was determined by plasma P-tau181 for 133 (22.7%) and by plasma P-tau181/Aβ42 for 104 (17.7%). Among individuals with clinically diagnosed AD, 69 (45.4%) had plasma P-tau181 levels and 89 (58.9%) had P-tau181/Aβ42 levels that were inconsistent with AD. Individuals with biomarker-negative clinical AD status tended to have lower levels of education, were less likely to carry APOE-ε4 alleles, and had lower levels of GFAP and NfL than individuals with biomarker-positive clinical AD. Conclusions and Relevance In this cross-sectional study, plasma P-tau181 and P-tau181/Aβ42 measurements correctly classified Caribbean Hispanic individuals with and without AD. However, plasma biomarkers identified individuals without dementia with biological evidence of AD, and a portion of those with dementia whose AD biomarker profile was negative. These results suggest that plasma biomarkers can augment detection of preclinical AD among asymptomatic individuals and improve the specificity of AD diagnosis.
Collapse
Affiliation(s)
- Lawrence S. Honig
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- G. H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Universidad Pedro Henríquez Urena, Santo Domingo, Dominican Republic
| | - Min Suk Kang
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Annie J. Lee
- G. H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Universidad Pedro Henríquez Urena, Santo Domingo, Dominican Republic
| | - Dolly Reyes-Dumeyer
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- G. H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Angel Piriz
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Belisa Soriano
- Universidad Pedro Henríquez Urena, Santo Domingo, Dominican Republic
| | | | | | - Patricia Recio
- Center for Diagnosis, Advanced Medicine and Telemedicine, Santo Domingo, Dominican Republic
| | - Diones Rivera Mejía
- Universidad Pedro Henríquez Urena, Santo Domingo, Dominican Republic
- Center for Diagnosis, Advanced Medicine and Telemedicine, Santo Domingo, Dominican Republic
| | - Martin Medrano
- Pontíficia Universidad Católica Madre y Maestra, Santiago, Dominican Republic
| | - Rafael A. Lantigua
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Medicine, Vagelos College of Physicians and Surgeons, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Andrew F. Teich
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Neurology, Vagelos College of Physicians and Surgeons, New York Presbyterian Hospital, Columbia University, New York, New York
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Jeffrey L. Dage
- Department of Neurology, Indiana University School of Medicine, Indianapolis
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- G. H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Universidad Pedro Henríquez Urena, Santo Domingo, Dominican Republic
| |
Collapse
|
78
|
Andersson E, Schultz N, Saito T, Saido TC, Blennow K, Gouras GK, Zetterberg H, Hansson O. Cerebral Aβ deposition precedes reduced cerebrospinal fluid and serum Aβ42/Aβ40 ratios in the App NL-F/NL-F knock-in mouse model of Alzheimer's disease. Alzheimers Res Ther 2023; 15:64. [PMID: 36964585 PMCID: PMC10039589 DOI: 10.1186/s13195-023-01196-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/22/2023] [Indexed: 03/26/2023]
Abstract
BACKGROUND Aβ42/Aβ40 ratios in cerebrospinal fluid (CSF) and blood are reduced in preclinical Alzheimer's disease (AD), but their temporal and correlative relationship with cerebral Aβ pathology at this early disease stage is not well understood. In the present study, we aim to investigate such relationships using App knock-in mouse models of preclinical AD. METHODS CSF, serum, and brain tissue were collected from 3- to 18-month-old AppNL-F/NL-F knock-in mice (n = 48) and 2-18-month-old AppNL/NL knock-in mice (n = 35). The concentrations of Aβ42 and Aβ40 in CSF and serum were measured using Single molecule array (Simoa) immunoassays. Cerebral Aβ plaque burden was assessed in brain tissue sections by immunohistochemistry and thioflavin S staining. Furthermore, the concentrations of Aβ42 in soluble and insoluble fractions prepared from cortical tissue homogenates were measured using an electrochemiluminescence immunoassay. RESULTS In AppNL-F/NL-F knock-in mice, Aβ42/Aβ40 ratios in CSF and serum were significantly reduced from 12 and 16 months of age, respectively. The initial reduction of these biomarkers coincided with cerebral Aβ pathology, in which a more widespread Aβ plaque burden and increased levels of Aβ42 in the brain were observed from approximately 12 months of age. Accordingly, in the whole study population, Aβ42/Aβ40 ratios in CSF and serum showed a negative hyperbolic association with cerebral Aβ plaque burden as well as the levels of both soluble and insoluble Aβ42 in the brain. These associations tended to be stronger for the measures in CSF compared with serum. In contrast, no alterations in the investigated fluid biomarkers or apparent cerebral Aβ plaque pathology were found in AppNL/NL knock-in mice during the observation time. CONCLUSIONS Our findings suggest a temporal sequence of events in AppNL-F/NL-F knock-in mice, in which initial deposition of Aβ aggregates in the brain is followed by a decline of the Aβ42/Aβ40 ratio in CSF and serum once the cerebral Aβ pathology becomes significant. Our results also indicate that the investigated biomarkers were somewhat more strongly associated with measures of cerebral Aβ pathology when assessed in CSF compared with serum.
Collapse
Affiliation(s)
- Emelie Andersson
- Clinical Memory Research Unit, Lund University, 22184, Lund, Sweden.
| | - Nina Schultz
- Clinical Memory Research Unit, Lund University, 22184, Lund, Sweden
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-Shi, Saitama, Japan
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Gunnar K Gouras
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- 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, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, 22184, Lund, Sweden.
- Memory Clinic, SkåneUniversity Hospital, 20502, Malmö, Sweden.
| |
Collapse
|
79
|
Chun MY, Jang H, Kim HJ, Kim JP, Gallacher J, Allué JA, Sarasa L, Castillo S, Pascual-Lucas M, Na DL, Seo SW, on behalf of DPUK. 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: 3] [Impact Index Per Article: 1.5] [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
|
80
|
Salvadó G, Ossenkoppele R, Ashton NJ, Beach TG, Serrano GE, Reiman EM, Zetterberg H, Mattsson-Carlgren N, Janelidze S, Blennow K, Hansson O. Specific associations between plasma biomarkers and postmortem amyloid plaque and tau tangle loads. EMBO Mol Med 2023; 15:e17123. [PMID: 36912178 PMCID: PMC10165361 DOI: 10.15252/emmm.202217123] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
Several promising plasma biomarkers for Alzheimer's disease have been recently developed, but their neuropathological correlates have not yet been fully determined. To investigate and compare independent associations between multiple plasma biomarkers (p-tau181, p-tau217, p-tau231, Aβ42/40, GFAP, and NfL) and neuropathologic measures of amyloid and tau, we included 105 participants from the Arizona Study of Aging and Neurodegenerative Disorders (AZSAND) with antemortem plasma samples and a postmortem neuropathological exam, 48 of whom had longitudinal p-tau217 and p-tau181. When simultaneously including plaque and tangle loads, the Aβ42/40 ratio and p-tau231 were only associated with plaques (ρAβ42/40 [95%CI] = -0.53[-0.65, -0.35], ρp-tau231 [95%CI] = 0.28[0.10, 0.43]), GFAP was only associated with tangles (ρGFAP [95%CI] = 0.39[0.17, 0.57]), and p-tau217 and p-tau181 were associated with both plaques (ρp-tau217 [95%CI] = 0.40[0.21, 0.56], ρp-tau181 [95%CI] = 0.36[0.15, 0.50]) and tangles (ρp-tau217 [95%CI] = 0.52[0.34, 0.66]; ρp-tau181 [95%CI] = 0.36[0.17, 0.52]). A model combining p-tau217 and the Aβ42/40 ratio showed the highest accuracy for predicting the presence of Alzheimer's disease neuropathological change (ADNC, AUC[95%CI] = 0.89[0.82, 0.96]) and plaque load (R2 = 0.55), while p-tau217 alone was optimal for predicting tangle load (R2 = 0.45). Our results suggest that high-performing assays of plasma p-tau217 and Aβ42/40 might be an optimal combination to assess Alzheimer's-related pathology in vivo.
Collapse
Affiliation(s)
- Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley, NHS Foundation, London, UK
| | | | | | - Eric M Reiman
- Banner Alzheimer's Institute, Arizona State University and University of Arizona, Phoenix, AZ, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
81
|
Heinzinger N, Maass A, Berron D, Yakupov R, Peters O, Fiebach J, Villringer K, Preis L, Priller J, Spruth EJ, Altenstein S, Schneider A, Fliessbach K, Wiltfang J, Bartels C, Jessen F, Maier F, Glanz W, Buerger K, Janowitz D, Perneczky R, Rauchmann BS, Teipel S, Killimann I, Göerß D, Laske C, Munk MH, Spottke A, Roy N, Heneka MT, Brosseron F, Dobisch L, Ewers M, Dechent P, Haynes JD, Scheffler K, Wolfsgruber S, Kleineidam L, Schmid M, Berger M, Düzel E, Ziegler G. Exploring the ATN classification system using brain morphology. Alzheimers Res Ther 2023; 15:50. [PMID: 36915139 PMCID: PMC10009950 DOI: 10.1186/s13195-023-01185-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 02/08/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND The NIA-AA proposed amyloid-tau-neurodegeneration (ATN) as a classification system for AD biomarkers. The amyloid cascade hypothesis (ACH) implies a sequence across ATN groups that patients might undergo during transition from healthy towards AD: A-T-N-➔A+T-N-➔A+T+N-➔A+T+N+. Here we assess the evidence for monotonic brain volume decline for this particular (amyloid-conversion first, tau-conversion second, N-conversion last) and alternative progressions using voxel-based morphometry (VBM) in a large cross-sectional MRI cohort. METHODS We used baseline data of the DELCODE cohort of 437 subjects (127 controls, 168 SCD, 87 MCI, 55 AD patients) which underwent lumbar puncture, MRI scanning, and neuropsychological assessment. ATN classification was performed using CSF-Aβ42/Aβ40 (A+/-), CSF phospho-tau (T+/-), and adjusted hippocampal volume or CSF total-tau (N+/-). We compared voxel-wise model evidence for monotonic decline of gray matter volume across various sequences over ATN groups using the Bayesian Information Criterion (including also ROIs of Braak stages). First, face validity of the ACH transition sequence A-T-N-➔A+T-N-➔A+T+N-➔A+T+N+ was compared against biologically less plausible (permuted) sequences among AD continuum ATN groups. Second, we evaluated evidence for 6 monotonic brain volume progressions from A-T-N- towards A+T+N+ including also non-AD continuum ATN groups. RESULTS The ACH-based progression A-T-N-➔A+T-N-➔A+T+N-➔A+T+N+ was consistent with cognitive decline and clinical diagnosis. Using hippocampal volume for operationalization of neurodegeneration (N), ACH was most evident in 9% of gray matter predominantly in the medial temporal lobe. Many cortical regions suggested alternative non-monotonic volume progressions over ACH progression groups, which is compatible with an early amyloid-related tissue expansion or sampling effects, e.g., due to brain reserve. Volume decline in 65% of gray matter was consistent with a progression where A status converts before T or N status (i.e., ACH/ANT) when compared to alternative sequences (TAN/TNA/NAT/NTA). Brain regions earlier affected by tau tangle deposition (Braak stage I-IV, MTL, limbic system) present stronger evidence for volume decline than late Braak stage ROIs (V/VI, cortical regions). Similar findings were observed when using CSF total-tau for N instead. CONCLUSION Using the ATN classification system, early amyloid status conversion (before tau and neurodegeneration) is associated with brain volume loss observed during AD progression. The ATN system and the ACH are compatible with monotonic progression of MTL atrophy. TRIAL REGISTRATION DRKS00007966, 04/05/2015, retrospectively registered.
Collapse
Affiliation(s)
- Nils Heinzinger
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany. .,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - David Berron
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Renat Yakupov
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Department of Psychiatry, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Jochen Fiebach
- Center for Stroke Research Berlin, Charité-Universitätsmedizin, Berlin, Germany
| | - Kersten Villringer
- Center for Stroke Research Berlin, Charité-Universitätsmedizin, Berlin, Germany
| | - Lukas Preis
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany.,Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany.,University of Edinburgh and UK DRI, Edinburgh, UK
| | - Eike Jacob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.,Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany.,Department of Medical Sciences, Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Claudia Bartels
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Franziska Maier
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany.,Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Ingo Killimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany.,Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Doreen Göerß
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Matthias H Munk
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurology, University of Bonn, Bonn, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Frederic Brosseron
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Peter Dechent
- MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University Göttingen, Göttingen, Germany
| | - John Dylan Haynes
- Bernstein Center for Computational Neuroscience, Charité-Universitätsmedizin, Berlin, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Matthias Schmid
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute for Medical Biometry, University Hospital Bonn, Bonn, Germany
| | - Moritz Berger
- Institute for Medical Biometry, University Hospital Bonn, Bonn, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Gabriel Ziegler
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | | |
Collapse
|
82
|
Advances in sample preparation and HPLC-MS/MS methods for determining amyloid-β peptide in biological samples: a review. Anal Bioanal Chem 2023:10.1007/s00216-023-04631-9. [PMID: 36877264 DOI: 10.1007/s00216-023-04631-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/28/2023] [Accepted: 02/23/2023] [Indexed: 03/07/2023]
Abstract
Alzheimer's disease (AD), a neurological disorder, is a major public health concern and the most common form of dementia. Its typical symptoms include memory loss, confusion, changes in personality, and cognitive impairment, which result in patients gradually losing independence. Over the last decades, some studies have focused on searching for effective biomarkers as early diagnostic indicators of AD. Amyloid-β (Aβ) peptides have been consolidated as reliable AD biomarkers and have been incorporated into modern diagnostic research criteria. However, quantitative analysis of Aβ peptides in biological samples remains a challenge because both the sample and the physical-chemical properties of these peptides are complex. During clinical routine, Aβ peptides are measured in the cerebrospinal fluid by immunoassays, but the availability of a specific antibody is critical-in some cases, an antibody may not exist, or its specificity may be inadequate, leading to low sensitivity and false results. HPLC-MS/MS has been reported as a sensitive and selective method for determining different fragments of Aβ peptides in biological samples simultaneously. Developments in sample preparation techniques (preconcentration platforms) such as immunoprecipitation, 96-well plate SPME, online SPME, and fiber-in-tube SPME have enabled not only effective enrichment of Aβ peptides present at trace levels in biological samples, but also efficient exclusion of interferents from the sample matrix (sample cleanup). This high extraction efficiency has provided MS platforms with higher sensitivity. Recently, methods affording LLOQ values as low as 5 pg mL-1 have been reported. Such low LLOQ values are adequate for quantifying Aβ peptides in complex matrixes including cerebrospinal fluid (CSF) and plasma samples. This review summarizes the advances in mass spectrometry (MS)-based methods for quantifying Aβ peptides and covers the period 1992-2022. Important considerations regarding the development of the HPLC-MS/MS method such as the sample preparation step, optimization of the HPLC-MS/MS parameters, and matrix effects are described. Clinical applications, difficulties related to analysis of plasma samples, and future trends of these MS/MS-based methods are also discussed.
Collapse
|
83
|
Kasuga K, Tsukie T, Kikuchi M, Tokutake T, Washiyama K, Simizu S, Yoshizawa H, Kuroha Y, Yajima R, Mori H, Arakawa Y, Onda K, Miyashita A, Onodera O, Iwatsubo T, Ikeuchi T. The Clinical Application of Optimized AT(N) Classification in Alzheimer’s Clinical Syndrome (ACS) and non-ACS Conditions. Neurobiol Aging 2023; 127:23-32. [PMID: 37030016 DOI: 10.1016/j.neurobiolaging.2023.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
We aimed to assess the utility of AT(N) classification in clinical practice. We measured the cerebrospinal fluid levels of amyloid-β (Aβ) 42, Aβ40, phosphorylated tau, total tau, and neurofilament light chain (NfL) in samples from 230 patients with Alzheimer's clinical syndrome (ACS) and 328 patients with non-ACS. The concordance of two A-markers (i.e., Aβ42 alone and the Aβ42/Aβ40 ratio) was not significantly different between the ACS (87.4%) and non-ACS (74.1%) groups. However, the frequency of discordant cases with AAβ42-alone+/AAβ-ratio- was significantly higher in the non-ACS (23.8%) than in the ACS group (7.4%). The concordance of two N-markers (i.e., total tau and NfL) was 40.4% in the ACS group and 24.4% in the non-ACS group. In the ACS samples, the frequency of biological Alzheimer's disease (i.e., A+T+) in Ntau+ cases was 95% while that in NNfL+ cases was 65%. Reflecting Aβ deposition and neurodegeneration more accurately, we recommend the use of AT(N) classification defined by cerebrospinal fluid AAβ-ratioTNNfL in clinical practice.
Collapse
|
84
|
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: 9] [Impact Index Per Article: 4.5] [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
|
85
|
Johansson C, Thordardottir S, Laffita-Mesa J, Rodriguez-Vieitez E, Zetterberg H, Blennow K, Graff C. Plasma biomarker profiles in autosomal dominant Alzheimer's disease. Brain 2023; 146:1132-1140. [PMID: 36626935 PMCID: PMC9976964 DOI: 10.1093/brain/awac399] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/16/2022] [Accepted: 10/01/2022] [Indexed: 01/12/2023] Open
Abstract
Emerging plasma biomarkers of Alzheimer's disease might be non-invasive tools to trace early Alzheimer's disease-related abnormalities such as the accumulation of amyloid-beta peptides, neurofibrillary tau tangles, glial activation and neurodegeneration. It is, however, unclear which pathological processes in the CNS can be adequately detected by peripheral measurements and whether plasma biomarkers are equally applicable in both clinical and preclinical phases. Here we aimed to explore the timing and performance of plasma biomarkers in mutation carriers compared to non-carriers in autosomal dominant Alzheimer's disease. Samples (n = 164) from mutation carriers (n = 33) and non-carriers (n = 42) in a Swedish cohort of autosomal dominant Alzheimer's disease (APP p.KM670/671NL, APP p.E693G and PSEN1 p.H163Y) were included in explorative longitudinal analyses. Plasma phosphorylated tau (P-tau181), total tau (T-tau), neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) concentrations were measured with a single-molecule array method as previously described. Plasma biomarkers were additionally correlated to Alzheimer's disease core biomarkers in the CSF. Results from the longitudinal analyses confirmed that plasma P-tau181, NfL and GFAP concentrations were higher in mutation carriers compared to non-carriers. This change was observed in the presymptomatic phase and detectable first as an increase in GFAP approximately 10 years before estimated symptom onset, followed by increased levels of P-tau181 and NfL closer to expected onset. Plasma P-tau181 levels were correlated to levels of P-tau181 and T-tau in the CSF. Altogether, plasma P-tau181, GFAP and NfL seem to be feasible biomarkers to detect different Alzheimer's disease-related pathologies already in presymptomatic individuals. Interestingly, changes in plasma GFAP concentrations were detected prior to P-tau181 and NfL. Our results suggest that plasma GFAP might reflect Alzheimer's disease pathology upstream to accumulation of tangles and neurodegeneration. The implications of these findings need additional validation, in particular because of the limited sample size.
Collapse
Affiliation(s)
- Charlotte Johansson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Steinunn Thordardottir
- Memory Clinic, Department of Geriatrics, Landspitali University Hospital, Reykjavik, Iceland
| | - José Laffita-Mesa
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Elena Rodriguez-Vieitez
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Caroline Graff
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
86
|
Constantinides VC, Paraskevas GP, Boufidou F, Bourbouli M, Pyrgelis ES, Stefanis L, Kapaki E. CSF Aβ42 and Aβ42/Aβ40 Ratio in Alzheimer's Disease and Frontotemporal Dementias. Diagnostics (Basel) 2023; 13:diagnostics13040783. [PMID: 36832271 PMCID: PMC9955886 DOI: 10.3390/diagnostics13040783] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Alzheimer's disease dementia (ADD) may manifest with atypical phenotypes, resembling behavioral variant frontotemporal dementia (bvFTD) and corticobasal syndrome (CBS), phenotypes which typically have an underlying frontotemporal lobar degeneration with tau proteinopathy (FTLD-tau), such as Pick's disease, corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), or FTLD with TDP-43 proteinopathy (FTLD-TDP). CSF biomarkers total and phosphorylated tau (τT and τP-181), and amyloid beta with 42 and 40 amino acids (Aβ42 and Aβ40) are biomarkers of AD pathology. The primary aim of this study was to compare the diagnostic accuracy of Aβ42 to Aβ42/Aβ40 ratio in: (a) differentiating ADD vs. frontotemporal dementias; (b) patients with AD pathology vs. non-AD pathologies; (c) compare biomarker ratios and composite markers to single CSF biomarkers in the differentiation of AD from FTD; Methods: In total, 263 subjects were included (ADD: n = 98; bvFTD: n = 49; PSP: n = 50; CBD: n = 45; controls: n = 21). CSF biomarkers were measured by commercially available ELISAs (EUROIMMUN). Multiple biomarker ratios (Aβ42/Aβ40; τT/τP-181; τT/Aβ42; τP-181/Aβ42) and composite markers (t-tau: τT/(Aβ42/Aβ40); p-tau: τP-181/(Aβ42/Aβ40) were calculated. ROC curve analysis was performed to compare AUCs of Aβ42 and Aβ42/Aβ40 ratio and relevant composite markers between ADD and FTD, as defined clinically. BIOMARKAPD/ABSI criteria (abnormal τT, τP-181 Aβ42, and Aβ42/Aβ40 ratio) were used to re-classify all patients into AD pathology vs. non-AD pathologies, and ROC curve analysis was repeated to compare Aβ42 and Aβ42/Aβ40; Results: Aβ42 did not differ from Aβ42/Aβ40 ratio in the differentiation of ADD from FTD (AUCs 0.752 and 0.788 respectively; p = 0.212). The τT/Aβ42 ratio provided maximal discrimination between ADD and FTD (AUC:0.893; sensitivity 88.8%, specificity 80%). BIOMARKAPD/ABSI criteria classified 60 patients as having AD pathology and 211 as non-AD. A total of 22 had discrepant results and were excluded. Aβ42/Aβ40 ratio was superior to Aβ42 in the differentiation of AD pathology from non-AD pathology (AUCs: 0.939 and 0.831, respectively; p < 0.001). In general, biomarker ratios and composite markers were superior to single CSF biomarkers in both analyses. CONCLUSIONS Aβ42/Aβ40 ratio is superior to Aβ42 in identifying AD pathology, irrespective of the clinical phenotype. CSF biomarker ratios and composite markers provide higher diagnostic accuracy compared to single CSF biomarkers.
Collapse
Affiliation(s)
- Vasilios C. Constantinides
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Correspondence: ; Tel.: +30-21-0728-9285
| | - George P. Paraskevas
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Second Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, Rimini 1, 12462 Athens, Greece
| | - Fotini Boufidou
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Mara Bourbouli
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Efstratios-Stylianos Pyrgelis
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Leonidas Stefanis
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Elisabeth Kapaki
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| |
Collapse
|
87
|
Zou Y, Yu S, Ma X, Ma C, Mao C, Mu D, Li L, Gao J, Qiu L. How far is the goal of applying β-amyloid in cerebrospinal fluid for clinical diagnosis of Alzheimer's disease with standardization of measurements? Clin Biochem 2023; 112:33-42. [PMID: 36473516 DOI: 10.1016/j.clinbiochem.2022.11.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/02/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Cerebrospinal fluid (CSF) β-amyloid (Aβ) is important for early diagnosis of Alzheimer's disease (AD). However, the cohort distributions and cut-off values have large variation across different analytical assays, kits, and laboratories. In this review, we summarize the cut-off values and diagnostic performance for CSF Aβ1-42 and Aβ1-42/Aβ1-40, and explore the important effect factors. Based on the Alzheimer's Association external quality control program (AAQC program), the peer group coefficient of variation of manual ELISA assays for CSF Aβ1-42 was unsatisfied (>20%). Fully automated platforms with better performance have recently been developed, but still not widely applied. In 2020, the certified reference material (CRM) for CSF Aβ1-42 was launched; however, the AAQC 2021-round results did not show effective improvements. Thus, further development and popularization of CRM for CSF Aβ1-42 and Aβ1-40 are urgently required. Standardizing the diagnostic procedures of AD and related status and the pre-analytical protocols of CSF samples, improving detection performance of analytical assays, and popularizing the application of fully automated platforms are also important for the establishment of uniform cut-off values. Moreover, each laboratory should verify the applicability of uniform cut-off values, and evaluate whether it is necessary to establish its own population- and assay-specific cut-off values.
Collapse
Affiliation(s)
- Yutong Zou
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Songlin Yu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Xiaoli Ma
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China; Medical Science Research Center, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Chaochao Ma
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Chenhui Mao
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Danni Mu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Lei Li
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Jing Gao
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
88
|
Aarsland D, Khalifa K, Bergland AK, Soennesyn H, Oppedal K, Holteng LBA, Oesterhus R, Nakling A, Jarholm JA, de Lucia C, Fladby T, Brooker H, Dalen I, Ballard C. A Randomised Placebo-Controlled Study of Purified Anthocyanins on Cognition in Individuals at Increased Risk for Dementia. Am J Geriatr Psychiatry 2023; 31:141-151. [PMID: 36372613 DOI: 10.1016/j.jagp.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022]
Abstract
IMPORTANCE Identifying nutritional compounds which can reduce cognitive decline in older people is a hugely important topic. OBJECTIVE To study the safety and effect of anthocyanins in maintaining cognitive functioning in people at increased risk for dementia. DESIGN, SETTING, AND PARTICIPANTS Participants (206 individuals, aged 60-80 years) diagnosed with either mild cognitive impairment (MCI) or two or more cardiometabolic disorders (i.e., diabetes, hypertension, obesity) were enrolled at three different centres in Norway. INTERVENTION Participants were randomly assigned to four capsules with a total of 320 mg/d of naturally purified anthocyanins or placebo 1:1 for 24 weeks. MAIN OUTCOMES AND MEASURES The primary outcome was the Quality of Episodic Memory composite measure (0-100) from an online cognitive test battery CogTrack, which was administered at baseline and monthly for the next 24 weeks. Secondary outcomes included other cognitive scores from the CogTrack battery. We applied mixed effects models with a baseline test score, group, time and their interaction as fixed effects, as well as other predefined baseline covariates. The primary comparison was the group difference at week 24 based on a modified intention-to-treat principle. RESULTS The primary analysis did not show a significant group difference at 24 weeks (78.2 versus 76.8; adjusted mean difference 1.4 (95% confidence interval -0.9-3.7); effect size 0.15; p = 0.23). However, there was a significant difference in slopes during weeks 8-24 (p = 0.007); the anthocyanin group improved while the placebo group worsened. No differences were found for the secondary cognitive outcomes. Anthocyanin capsules were well-tolerated and safe to use. CONCLUSION Anthocyanin supplementation for 24 weeks was safe and well tolerated in people with MCI or cardiometabolic disorders. We found no significant group difference in episodic memory at the end of the study but statistically significant differences in slopes. Further studies are warranted to explore whether anthocyanins supplementation can reduce cognitive decline in people at increased risk of dementia. TRIAL REGISTRATION ClinicalTrials.gov, (Identifier NCT03419039). http://www. CLINICALTRIALS gov/, NCT03419039.
Collapse
Affiliation(s)
- Dag Aarsland
- Centre for Age-Related Medicine (DA, KK, AKB, HS, LBAH, RO, AN, CDL), Stavanger University Hospital, Stavanger, Norway; Department of Old Age Psychiatry (DA), King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Khadija Khalifa
- Centre for Age-Related Medicine (DA, KK, AKB, HS, LBAH, RO, AN, CDL), Stavanger University Hospital, Stavanger, Norway; Department of Old Age Psychiatry (DA), King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK; The Faculty of Health Sciences (KK), University of Stavanger, Stavanger, Norway.
| | - Anne K Bergland
- Centre for Age-Related Medicine (DA, KK, AKB, HS, LBAH, RO, AN, CDL), Stavanger University Hospital, Stavanger, Norway
| | - Hogne Soennesyn
- Centre for Age-Related Medicine (DA, KK, AKB, HS, LBAH, RO, AN, CDL), Stavanger University Hospital, Stavanger, Norway
| | - Ketil Oppedal
- Department of Electrical Engineering and Computer Science (KO), University of Stavanger, Stavanger, Norway
| | - Lise B A Holteng
- Centre for Age-Related Medicine (DA, KK, AKB, HS, LBAH, RO, AN, CDL), Stavanger University Hospital, Stavanger, Norway; Department of Clinical Medicine (LBAH, AN), University of Bergen, Bergen, Norway
| | - Ragnhild Oesterhus
- Centre for Age-Related Medicine (DA, KK, AKB, HS, LBAH, RO, AN, CDL), Stavanger University Hospital, Stavanger, Norway; The Hospital Pharmacy Enterprise of Western Norway (RO), Bergen, Norway
| | - Arne Nakling
- Centre for Age-Related Medicine (DA, KK, AKB, HS, LBAH, RO, AN, CDL), Stavanger University Hospital, Stavanger, Norway; Department of Clinical Medicine (LBAH, AN), University of Bergen, Bergen, Norway
| | - Jonas A Jarholm
- Department of Neurology (AJ, TF), Akershus University Hospital, Lørenskog, Norway
| | - Chiara de Lucia
- Centre for Age-Related Medicine (DA, KK, AKB, HS, LBAH, RO, AN, CDL), Stavanger University Hospital, Stavanger, Norway; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience (CDL), King's College London, London, UK
| | - Tormod Fladby
- Department of Neurology (AJ, TF), Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine (HB, TF), University of Oslo, Oslo, Norway
| | - Helen Brooker
- Medical School (HB), University of Exeter, Exeter, UK; Ecog Pro Ltd. (HB, CB), Bristol, UK
| | - Ingvild Dalen
- Section of Biostatistics, Department of Research (ID), Stavanger University Hospital, Stavanger, Norway
| | - Clive Ballard
- Medical School (HB), University of Exeter, Exeter, UK
| |
Collapse
|
89
|
Axelsson Andrén E, Kettunen P, Bjerke M, Rolstad S, Zetterberg H, Blennow K, Wallin A, Svensson J. Diagnostic Performance of Cerebrospinal Fluid Neurofilament Light Chain and Soluble Amyloid-β Protein Precursor β in the Subcortical Small Vessel Type of Dementia. J Alzheimers Dis 2023; 96:1515-1528. [PMID: 37980667 PMCID: PMC10741327 DOI: 10.3233/jad-230680] [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] [Accepted: 09/27/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND The subcortical small vessel type of dementia (SSVD) is a common subtype of vascular dementia, but there is a lack of disease-specific cerebrospinal fluid (CSF) biomarkers. OBJECTIVE We investigated whether CSF concentrations of neurofilament light chain (NFL), soluble amyloid-β protein precursor α (sAβPPα), sAβPPβ, and CSF/serum albumin ratio could separate SSVD from healthy controls, Alzheimer's disease (AD), and mixed dementia (combined AD and SSVD). METHODS This was a mono-center study of patients with SSVD (n = 38), AD (n = 121), mixed dementia (n = 62), and controls (n = 96). The CSF biomarkers were measured using immunoassays, and their independent contribution to the separation between groups were evaluated using the Wald test. Then, the area under the receiver operating characteristics curve (AUROC) and 95% confidence intervals (CIs) were calculated. RESULTS Elevated neurofilament light chain (NFL) and decreased sAβPPβ independently separated SSVD from controls, and sAβPPβ also distinguished SSVD from AD and mixed dementia. The combination of NFL and sAβPPβ discriminated SSVD from controls with high accuracy (AUROC 0.903, 95% CI: 0.834-0.972). Additionally, sAβPPβ combined with the core AD biomarkers (amyloid-β42, total tau, and phosphorylated tau181) had a high ability to separate SSVD from AD (AUROC 0.886, 95% CI: 0.830-0.942) and mixed dementia (AUROC 0.903, 95% CI: 0.838-0.968). CONCLUSIONS The high accuracy of NFL and sAβPPβ to separate SSVD from controls supports that SSVD is a specific diagnostic entity. Moreover, SSVD was distinguished from AD and mixed dementia using sAβPPβ in combination with the core AD biomarkers.
Collapse
Affiliation(s)
- Elin Axelsson Andrén
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Petronella Kettunen
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, Cognition and Old Age Psychiatry, Sahlgrenska University Hospital, Region Västra Götaland, Mölndal, Sweden
| | - Maria Bjerke
- Laboratory of Clinical Neurochemistry, Department of Clinical Biology, Universitair Ziekenhuis Brussel, and Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Biomedical Sciences and Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Sindre Rolstad
- Department of Psychology, Faculty of Social Science, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Labratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Labratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, Cognition and Old Age Psychiatry, Sahlgrenska University Hospital, Region Västra Götaland, Mölndal, Sweden
| | - Johan Svensson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Internal Medicine, Skaraborg Central Hospital, Region Västra Götaland, Skövde, Sweden
| |
Collapse
|
90
|
Zhou J, Wang ZB, Sun Y, Fu Y, Li D, Tan L. Cerebrospinal Fluid Complement 4 Levels Were Associated with Alzheimer's Disease Pathology and Cognition in Non-Demented Elderly. J Alzheimers Dis 2023; 96:1071-1081. [PMID: 38007670 DOI: 10.3233/jad-230513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
BACKGROUND Numerous studies have shown that the complement system plays an important role in Alzheimer's disease (AD). However, whether complement 4 (C4) protein in cerebrospinal fluid (CSF) was associated with AD pathology, especially in the early stage of AD, is still unclear. OBJECTIVE We aimed to explore the association of CSF C4 with AD pathology and cognition in the preclinical AD. METHODS The study included a total of 287 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. Based on the A/T scheme, they were divided into four groups to access the changes of CSF C4 in the preclinical AD. Linear regression models were used to test the associations between CSF C4 and AD core biomarkers, namely Aβ42, P-tau, and T-tau. RESULTS The level of CSF C4 decreased in the A + T- group compared with the A-T- group (p = 0.04) and it increased in the A-T+ group compared to the A + T- group (p = 0.01). In pooled samples, C4 was significantly associated with AD core biomarkers (all p < 0.05), but only in the A + group after stratification according to the A/T scheme. Furthermore, CSF C4 levels at baseline were associated with longitudinal cognitive changes. CONCLUSIONS Our results showed that CSF C4 levels changed dynamically in the preclinical AD, and that the responses of CSF C4 to brain Aβ pathology, tau pathology and neurodegeneration were found only in the presence of amyloid plaques, both of which indicates the complex link between C4 and AD.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Zhi-Bo Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Sun
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Da Li
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
91
|
Liu L, Lauro BM, He A, Lee H, Bhattarai S, Wolfe MS, Bennett DA, Karch CM, Young-Pearse T, Dominantly Inherited Alzheimer Network (DIAN), Selkoe DJ. Identification of the Aβ37/42 peptide ratio in CSF as an improved Aβ biomarker for Alzheimer's disease. Alzheimers Dement 2023; 19:79-96. [PMID: 35278341 PMCID: PMC9464800 DOI: 10.1002/alz.12646] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Identifying CSF-based biomarkers for the β-amyloidosis that initiates Alzheimer's disease (AD) could provide inexpensive and dynamic tests to distinguish AD from normal aging and predict future cognitive decline. METHODS We developed immunoassays specifically detecting all C-terminal variants of secreted amyloid β-protein and identified a novel biomarker, the Aβ 37/42 ratio, that outperforms the canonical Aβ42/40 ratio as a means to evaluate the γ-secretase activity and brain Aβ accumulation. RESULTS We show that Aβ 37/42 can distinguish physiological and pathological status in (1) presenilin-1 mutant vs wild-type cultured cells, (2) AD vs control brain tissue, and (3) AD versus cognitively normal (CN) subjects in CSF, where 37/42 (AUC 0.9622) outperformed 42/40 (AUC 0.8651) in distinguishing CN from AD. DISCUSSION We conclude that the Aβ 37/42 ratio sensitively detects presenilin/γ-secretase dysfunction and better distinguishes CN from AD than Aβ42/40 in CSF. Measuring this novel ratio alongside promising phospho-tau analytes may provide highly discriminatory fluid biomarkers for AD.
Collapse
Affiliation(s)
- Lei Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Bianca M. Lauro
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Amy He
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Hyo Lee
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Sanjay Bhattarai
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS USA
| | - Michael S. Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center Rush University Medical Center, Chicago, IL USA
| | - Celeste M. Karch
- Department of Psychiatry, Washington University in St Louis, St. Louis, MO USA
- Hope Center for Neurologic Disorders, St. Louis, MO USA
| | - Tracy Young-Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | | | - Dennis J. Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|
92
|
Shir D, Mielke MM, Hofrenning EI, Lesnick TG, Knopman DS, Petersen RC, Jack CR, Algeciras-Schimnich A, Vemuri P, Graff-Radford J. Associations of Neurodegeneration Biomarkers in Cerebrospinal Fluid with Markers of Alzheimer's Disease and Vascular Pathology. J Alzheimers Dis 2023; 92:887-898. [PMID: 36806507 PMCID: PMC10193844 DOI: 10.3233/jad-221015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
BACKGROUND The National Institute on Aging-Alzheimer's Association Research Framework proposes defining Alzheimer's disease by grouping imaging and fluid biomarkers by their respective pathologic processes. The AT(N) structure proposes several neurodegenerative fluid biomarkers (N) including total tau (t-tau), neurogranin (Ng), and neurofilament light chain (NfL). However, pathologic drivers influencing each biomarker remain unclear. OBJECTIVE To determine whether cerebrospinal fluid (CSF)-neurodegenerative biomarkers (N) map differentially to Alzheimer's disease pathology measured by Aβ42 (an indicator of amyloidosis, [A]), p-tau (an indicator of tau deposition, [T]), and MRI vascular pathology indicators (measured by white-matter integrity, infarcts, and microbleeds [V]). METHODS Participants were from Mayo Clinic Study of Aging (MCSA) with CSF measures of NfL, Ng, t-tau, Aβ42, and p-tau and available MRI brain imaging. Linear models assessed associations between CSF neurodegeneration (N) markers, amyloid markers (A), tau (T), and vascular pathology (V). RESULTS Participants (n = 408) had a mean age of 69.2±10.7; male, 217 (53.2%); cognitively unimpaired, 359 (88%). All three neurodegeneration biomarkers correlated with age (p < 0.001 for NfL and t-tau, p = 0.018 for Ng). Men had higher CSF-NfL levels; women had higher Ng (p < 0.001). NfL and t-tau levels correlated with infarcts (p = 0.009, p = 0.034 respectively); no biomarkers correlated with white-matter integrity. N biomarkers correlated with p-tau levels (T, p < 0.001). Higher Aβ42 levels associated with higher N-biomarker levels but only among cognitively unimpaired (A, p < 0.001). CONCLUSION The influence of vascular pathology in the general population on CSF (N) biomarkers is modest, with greater influence of infarcts than white-matter disruption. Neurodegeneration markers more closely correlated with tau than amyloid markers.
Collapse
Affiliation(s)
- Dror Shir
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Michelle M. Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota 55905, USA
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, 27101
| | | | - Timothy G. Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - David S. Knopman
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Ronald C. Petersen
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55905, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Clifford R. Jack
- Department of Radiology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | |
Collapse
|
93
|
Baumann K, Šneiderienė G, Sanguanini M, Schneider M, Rimon O, González Díaz A, Greer H, Thacker D, Linse S, Knowles TPJ, Vendruscolo M. A Kinetic Map of the Influence of Biomimetic Lipid Model Membranes on Aβ 42 Aggregation. ACS Chem Neurosci 2022; 14:323-329. [PMID: 36574473 PMCID: PMC9853501 DOI: 10.1021/acschemneuro.2c00765] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The aggregation of the amyloid β (Aβ) peptide is one of the molecular hallmarks of Alzheimer's disease (AD). Although Aβ deposits have mostly been observed extracellularly, various studies have also reported the presence of intracellular Aβ assemblies. Because these intracellular Aβ aggregates might play a role in the onset and progression of AD, it is important to investigate their possible origins at different locations of the cell along the secretory pathway of the amyloid precursor protein, from which Aβ is derived by proteolytic cleavage. Senile plaques found in AD are largely composed of the 42-residue form of Aβ (Aβ42). Intracellularly, Aβ42 is produced in the endoplasmatic reticulum (ER) and Golgi apparatus. Since lipid bilayers have been shown to promote the aggregation of Aβ, in this study, we measure the effects of the lipid membrane composition on the in vitro aggregation kinetics of Aβ42. By using large unilamellar vesicles to model cellular membranes at different locations, including the inner and outer leaflets of the plasma membrane, late endosomes, the ER, and the Golgi apparatus, we show that Aβ42 aggregation is inhibited by the ER and Golgi model membranes. These results provide a preliminary map of the possible effects of the membrane composition in different cellular locations on Aβ aggregation and suggest the presence of an evolutionary optimization of the lipid composition to prevent the intracellular aggregation of Aβ.
Collapse
Affiliation(s)
- Kevin
N. Baumann
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CambridgeCB2 1EW, U.K.
| | - Greta Šneiderienė
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CambridgeCB2 1EW, U.K.
| | - Michele Sanguanini
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CambridgeCB2 1EW, U.K.
| | - Matthias Schneider
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CambridgeCB2 1EW, U.K.
| | - Oded Rimon
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CambridgeCB2 1EW, U.K.
| | - Alicia González Díaz
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CambridgeCB2 1EW, U.K.
| | - Heather Greer
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CambridgeCB2 1EW, U.K.
| | - Dev Thacker
- Department
of Biochemistry and Structural Biology, Lund University, LundSE22100, Sweden
| | - Sara Linse
- Department
of Biochemistry and Structural Biology, Lund University, LundSE22100, Sweden
| | - Tuomas P. J. Knowles
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CambridgeCB2 1EW, U.K.,Cavendish
Laboratory, University of Cambridge, CambridgeCB3 0HE, U.K.
| | - Michele Vendruscolo
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CambridgeCB2 1EW, U.K.,
| |
Collapse
|
94
|
Yamashita K, Miura M, Watanabe S, Ishiki K, Arimatsu Y, Kawahira J, Kubo T, Sasaki K, Arai T, Hagino K, Irino Y, Nagai K, Verbel D, Koyama A, Dhadda S, Niiro H, Iwanaga S, Sato T, Yoshida T, Iwata A. Fully automated and highly specific plasma β-amyloid immunoassays predict β-amyloid status defined by amyloid positron emission tomography with high accuracy. Alzheimers Res Ther 2022; 14:86. [PMID: 35739591 PMCID: PMC9219197 DOI: 10.1186/s13195-022-01029-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/15/2022] [Indexed: 11/10/2022]
Abstract
Abstract
Background
Clinicians, researchers, and patients alike would greatly benefit from more accessible and inexpensive biomarkers for neural β-amyloid (Aβ). We aimed to assess the performance of fully automated plasma Aβ immunoassays, which correlate significantly with immunoprecipitation mass spectrometry assays, in predicting brain Aβ status as determined by visual read assessment of amyloid positron emission tomography (PET).
Methods
The plasma Aβ42/Aβ40 ratio was measured using a fully automated immunoassay platform (HISCL series) in two clinical studies (discovery and validation studies). The discovery and validation sample sets were retrospectively and randomly selected from participants with early Alzheimer’s disease (AD) identified during screening for the elenbecestat Phase 3 program.
Results
We included 197 participants in the discovery study (mean [SD] age 71.1 [8.5] years; 112 females) and 200 in the validation study (age 70.8 [7.9] years; 99 females). The plasma Aβ42/Aβ40 ratio predicted amyloid PET visual read status with areas under the receiver operating characteristic curves of 0.941 (95% confidence interval [CI] 0.910–0.973) and 0.868 (95% CI 0.816–0.920) in the discovery and validation studies, respectively. In the discovery study, a cutoff value of 0.102 was determined based on maximizing the Youden Index, and the sensitivity and specificity were calculated to be 96.0% (95% CI 90.1–98.9%) and 83.5% (95% CI 74.6–90.3%), respectively. Using the same cutoff value, the sensitivity and specificity in the validation study were calculated to be 88.0% (95% CI 80.0–93.6%) and 72.0% (95% CI 62.1–80.5%), respectively.
Conclusions
The plasma Aβ42/Aβ40 ratio measured using the HISCL series achieved high accuracy in predicting amyloid PET status. Since our blood-based immunoassay system is less invasive and more accessible than amyloid PET and cerebrospinal fluid testing, it may contribute to the diagnosis of AD in routine clinical practice.
Collapse
|
95
|
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, for the European Prevention of Alzheimer’s Disease (EPAD) Consortium. 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: 25] [Impact Index Per Article: 8.3] [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
|
96
|
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
|
97
|
Cerebrospinal Fluid Biomarker Profile in TDP-43-Related Genetic Frontotemporal Dementia. J Pers Med 2022; 12:jpm12101747. [PMID: 36294886 PMCID: PMC9605286 DOI: 10.3390/jpm12101747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/06/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Cerebrospinal fluid (CSF) biomarkers, namely total tau, phospho-tau and amyloid beta peptides, have received much attention specifically regarding Alzheimer’s disease (AD), since they can detect the biochemical fingerprint of AD and serve as a diagnostic tool for accurate and early diagnosis during life. In the same way, biomarkers for other neurodegenerative disease pathologies are also needed. We present a case series of six patients with genetic frontotemporal dementia (FTD), with TDP-43 underlying proteinopathy, in an attempt to assess TDP-43 as a novel biomarker alone and in combination with established AD biomarkers for this specific patient group, based on the principles of personalized and precision medicine. Our results indicate that genetic TDP-43-FTD is characterized by increased CSF TPD-43 and increased TDP-43 × τΤ/τP-181 combination. Hence, TDP-43 combined with tau proteins could be a useful tool for the diagnosis of genetic FTD with TDP-43 underling histopathology, supplementing clinical, neuropsychological and imaging data.
Collapse
|
98
|
Lynch M, Pham W, Sinclair B, O’Brien TJ, Law M, Vivash L. Perivascular spaces as a potential biomarker of Alzheimer's disease. Front Neurosci 2022; 16:1021131. [PMID: 36330347 PMCID: PMC9623161 DOI: 10.3389/fnins.2022.1021131] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/23/2022] [Indexed: 07/20/2023] Open
Abstract
Alzheimer's disease (AD) is a highly damaging disease that affects one's cognition and memory and presents an increasing societal and economic burden globally. Considerable research has gone into understanding AD; however, there is still a lack of effective biomarkers that aid in early diagnosis and intervention. The recent discovery of the glymphatic system and associated Perivascular Spaces (PVS) has led to the theory that enlarged PVS (ePVS) may be an indicator of AD progression and act as an early diagnostic marker. Visible on Magnetic Resonance Imaging (MRI), PVS appear to enlarge when known biomarkers of AD, amyloid-β and tau, accumulate. The central goal of ePVS and AD research is to determine when ePVS occurs in AD progression and if ePVS are causal or epiphenomena. Furthermore, if ePVS are indeed causative, interventions promoting glymphatic clearance are an attractive target for research. However, it is necessary first to ascertain where on the pathological progression of AD ePVS occurs. This review aims to examine the knowledge gap that exists in understanding the contribution of ePVS to AD. It is essential to understand whether ePVS in the brain correlate with increased regional tau distribution and global or regional Amyloid-β distribution and to determine if these spaces increase proportionally over time as individuals experience neurodegeneration. This review demonstrates that ePVS are associated with reduced glymphatic clearance and that this reduced clearance is associated with an increase in amyloid-β. However, it is not yet understood if ePVS are the outcome or driver of protein accumulation. Further, it is not yet clear if ePVS volume and number change longitudinally. Ultimately, it is vital to determine early diagnostic criteria and early interventions for AD to ease the burden it presents to the world; ePVS may be able to fulfill this role and therefore merit further research.
Collapse
Affiliation(s)
- Miranda Lynch
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - William Pham
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Benjamin Sinclair
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Terence J. O’Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
- Department of Neurology, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Meng Law
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Radiology, Alfred Health, Melbourne, VIC, Australia
- Department of Electrical and Computer Systems Engineering, Monash University, Melbourne, VIC, Australia
| | - Lucy Vivash
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
- Department of Neurology, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
99
|
Svenningsson AL, Stomrud E, Palmqvist S, Hansson O, Ossenkoppele R. Axonal degeneration and amyloid pathology predict cognitive decline beyond cortical atrophy. Alzheimers Res Ther 2022; 14:144. [PMID: 36192766 PMCID: PMC9531524 DOI: 10.1186/s13195-022-01081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 09/11/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND Cortical atrophy is associated with cognitive decline, but the association is not perfect. We aimed to identify factors explaining the discrepancy between the degree of cortical atrophy and cognitive decline in cognitively unimpaired elderly. METHODS The discrepancy between atrophy and cognitive decline was measured using the residuals from a linear regression analysis between change in whole brain cortical thickness over time and change in a cognitive composite measure over time in 395 cognitively unimpaired participants from the Swedish BioFINDER study. We tested for bivariate associations of this residual measure with demographic, imaging, and fluid biomarker variables using Pearson correlations and independent-samples t-tests, and for multivariate associations using linear regression models. Mediation analyses were performed to explore possible paths between the included variables. RESULTS In bivariate analyses, older age (r = -0.11, p = 0.029), male sex (t = -3.00, p = 0.003), larger intracranial volume (r = -0.17, p < 0.001), carrying an APOEe4 allele (t = -2.71, p = 0.007), larger white matter lesion volume (r = -0.16, p = 0.002), lower cerebrospinal fluid (CSF) β-amyloid (Aβ) 42/40 ratio (t = -4.05, p < 0.001), and higher CSF levels of phosphorylated tau (p-tau) 181 (r = -0.22, p < 0.001), glial fibrillary acidic protein (GFAP; r = -0.15, p = 0.003), and neurofilament light (NfL; r = -0.34, p < 0.001) were negatively associated with the residual measure, i.e., associated with worse than expected cognitive trajectory given the level of atrophy. In a multivariate analysis, only lower CSF Aβ42/40 ratio and higher CSF NfL levels explained cognition beyond brain atrophy. Mediation analyses showed that associations between the residual measure and APOEe4 allele, CSF Aβ42/40 ratio, and CSF GFAP and p-tau181 levels were mediated by levels of CSF NfL, as were the associations with the residual measure for age, sex, and WML volume. CONCLUSIONS Our results suggest that axonal degeneration and amyloid pathology independently affect the rate of cognitive decline beyond the degree of cortical atrophy. Furthermore, axonal degeneration mediated the negative effects of old age, male sex, and white matter lesions, and in part also amyloid and tau pathology, on cognition over time when accounting for cortical atrophy.
Collapse
Affiliation(s)
- Anna Linnéa Svenningsson
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.484519.5Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
100
|
Xu C, Zhao L, Dong C. A Review of Application of Aβ42/40 Ratio in Diagnosis and Prognosis of Alzheimer’s Disease. J Alzheimers Dis 2022; 90:495-512. [DOI: 10.3233/jad-220673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The number of patients with Alzheimer’s disease (AD) and non-Alzheimer’s disease (non-AD) has drastically increased over recent decades. The amyloid cascade hypothesis attributes a vital role to amyloid-β protein (Aβ) in the pathogenesis of AD. As the main pathological hallmark of AD, amyloid plaques consist of merely the 42 and 40 amino acid variants of Aβ (Aβ 42 and Aβ 40). The cerebrospinal fluid (CSF) biomarker Aβ 42/40 has been extensively investigated and eventually integrated into important diagnostic tools to support the clinical diagnosis of AD. With the development of highly sensitive assays and technologies, blood-based Aβ 42/40, which was obtained using a minimally invasive and cost-effective method, has been proven to be abnormal in synchrony with CSF biomarker values. This paper presents the recent progress of the CSF Aβ 42/40 ratio and plasma Aβ 42/40 for AD as well as their potential clinical application as diagnostic markers or screening tools for dementia.
Collapse
Affiliation(s)
- Chang Xu
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Li Zhao
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunbo Dong
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|