101
|
McKenzie C, Bucks RS, Weinborn M, Bourgeat P, Salvado O, Gavett BE. Residual reserve index modifies the effect of amyloid pathology on fluorodeoxyglucose metabolism: Implications for efficiency and capacity in cognitive reserve. Front Aging Neurosci 2022; 14:943823. [PMID: 36034126 PMCID: PMC9413056 DOI: 10.3389/fnagi.2022.943823] [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: 05/14/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background The residual approach to measuring cognitive reserve (using the residual reserve index) aims to capture cognitive resilience conferred by cognitive reserve, but may be confounded by factors representing brain resilience. We sought to distinguish between brain and cognitive resilience by comparing interactions between the residual reserve index and amyloid, tau, and neurodegeneration ["AT(N)"] biomarkers when predicting executive function. We hypothesized that the residual reserve index would moderate at least one path from an AT(N) biomarker to executive function (consistent with cognitive resilience), as opposed to moderating a path between two AT(N) biomarkers (suggestive of brain resilience). Methods Participants (N = 332) were from the Alzheimer's Disease Neuroimaging Initiative. The residual reserve index represented the difference between observed and predicted memory performance (a positive residual reserve index suggests higher cognitive reserve). AT(N) biomarkers were: CSF β-amyloid1-42/β-amyloid1-40 (A), plasma phosphorylated tau-181 (T), and FDG metabolism in AD-specific regions ([N]). AT(N) biomarkers (measured at consecutive time points) were entered in a sequential mediation model testing the indirect effects from baseline amyloid to executive function intercept (third annual follow-up) and slope (baseline to seventh follow-up), via tau and/or FDG metabolism. The baseline residual reserve index was entered as a moderator of paths between AT(N) biomarkers (e.g., amyloid-tau), and paths between AT(N) biomarkers and executive function. Results The residual reserve index interacted with amyloid pathology when predicting FDG metabolism: the indirect effect of amyloid → FDG metabolism → executive function intercept and slope varied as a function of the residual reserve index. With lower amyloid pathology, executive function performance was comparable at different levels of the residual reserve index, but a higher residual reserve index was associated with lower FDG metabolism. With higher amyloid pathology, a higher residual reserve index predicted better executive function via higher FDG metabolism. Conclusion The effect of the residual reserve index on executive function performance via FDG metabolism was consistent with cognitive resilience. This suggests the residual reserve index captures variation in cognitive reserve; specifically, neural efficiency, and neural capacity to upregulate metabolism to enhance cognitive resilience in the face of greater amyloid pathology. Implications for future research include the potential bidirectionality between neural efficiency and amyloid accumulation.
Collapse
Affiliation(s)
- Cathryn McKenzie
- School of Psychological Science, The University of Western Australia, Perth, WA, Australia
| | - Romola S. Bucks
- School of Psychological Science, The University of Western Australia, Perth, WA, Australia
| | - Michael Weinborn
- School of Psychological Science, The University of Western Australia, Perth, WA, Australia
| | - Pierrick Bourgeat
- Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation (CSIRO) Health and Biosecurity, Brisbane, QLD, Australia
| | - Olivier Salvado
- Data61, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Sydney, NSW, Australia
| | - Brandon E. Gavett
- School of Psychological Science, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
102
|
Gong X, Zhang H, Liu X, Liu Y, Liu J, Fapohunda FO, Lü P, Wang K, Tang M. Is liquid biopsy mature enough for the diagnosis of Alzheimer's disease? Front Aging Neurosci 2022; 14:977999. [PMID: 35992602 PMCID: PMC9389010 DOI: 10.3389/fnagi.2022.977999] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 07/18/2022] [Indexed: 01/10/2023] Open
Abstract
The preclinical diagnosis and clinical practice for Alzheimer's disease (AD) based on liquid biopsy have made great progress in recent years. As liquid biopsy is a fast, low-cost, and easy way to get the phase of AD, continual efforts from intense multidisciplinary studies have been made to move the research tools to routine clinical diagnostics. On one hand, technological breakthroughs have brought new detection methods to the outputs of liquid biopsy to stratify AD cases, resulting in higher accuracy and efficiency of diagnosis. On the other hand, diversiform biofluid biomarkers derived from cerebrospinal fluid (CSF), blood, urine, Saliva, and exosome were screened out and biologically verified. As a result, more detailed knowledge about the molecular pathogenesis of AD was discovered and elucidated. However, to date, how to weigh the reports derived from liquid biopsy for preclinical AD diagnosis is an ongoing question. In this review, we briefly introduce liquid biopsy and the role it plays in research and clinical practice. Then, we summarize the established fluid-based assays of the current state for AD diagnostic such as ELISA, single-molecule array (Simoa), Immunoprecipitation-Mass Spectrometry (IP-MS), liquid chromatography-MS, immunomagnetic reduction (IMR), multimer detection system (MDS). In addition, we give an updated list of fluid biomarkers in the AD research field. Lastly, the current outstanding challenges and the feasibility to use a stand-alone biomarker in the joint diagnostic strategy are discussed.
Collapse
Affiliation(s)
- Xun Gong
- Department of Rheumatology and Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Hantao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Xiaoyan Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yi Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Junlin Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | | | - Peng Lü
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Kun Wang
- Children’s Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
103
|
Kumar A, Janelidze S, Stomrud E, Palmqvist S, Hansson O, Mattsson-Carlgren N. β-Amyloid-Dependent and -Independent Genetic Pathways Regulating CSF Tau Biomarkers in Alzheimer Disease. Neurology 2022; 99:e476-e487. [PMID: 35641311 PMCID: PMC9421595 DOI: 10.1212/wnl.0000000000200605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/11/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Abnormal metabolism of β-amyloid (Aβ) and soluble phosphorylated tau (P-tau), as well as neurodegeneration, are key components of Alzheimer disease (AD), but it is unclear how these different processes are related to genetic risk factors for AD. METHODS In the Swedish BioFINDER study, we tested associations between a priori defined polygenic risk scores (PRSs) for AD (excluding single-nucleotide polymorphism [SNP] within the APOE region in the main analysis) and biomarkers in CSF (total tau [T-tau] and P-tau181; Aβ1-38, Aβ1-40, Aβ1-42, and Aβ1-42/1-40; and neurofilament light [NfL]) in cognitively unimpaired (CU) individuals (n = 751), and in patients with mild cognitive impairment (MCI) (n = 212) and AD dementia (n = 150). Results were validated in the Alzheimer's Disease Neuroimaging Initiative data set with 777 individuals (AD = 119, MCI = 442, and CU = 216). RESULTS PRSs with SNPs significant at p < 5e-03 (∼1,742 variants) were associated with higher CSF P-tau181 (β = 0.13, p = 5.6e-05) and T-tau (β = 0.12, p = 4.3e-04). The associations between PRS and tau measures were partly attenuated but remained significant after adjusting for Aβ status. Aβ pathology mediated 37% of the effect of this PRS on tau levels. Aβ-dependent and Aβ-independent subsets of the PRS were identified and characterized. There were also associations between PRSs and CSF Aβ biomarkers with nominal significance, but not when corrected for multiple comparisons. There were no associations between PRSs and CSF NfL. DISCUSSION Genetic pathways implicated in causing AD are related to altered levels of soluble tau through both Aβ-dependent and Aβ-independent mechanisms, which may have relevance for anti-tau drug development.
Collapse
Affiliation(s)
- Atul Kumar
- From the Clinical Memory Research Unit (A.K., S.J., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences, Lund University, Malmö; Memory Clinic (E.S., S.P., O.H.), Skåne University Hospital, Malmö; Department of Neurology (N.M.-C.), Skåne University Hospital, Lund; and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden.
| | - Shorena Janelidze
- From the Clinical Memory Research Unit (A.K., S.J., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences, Lund University, Malmö; Memory Clinic (E.S., S.P., O.H.), Skåne University Hospital, Malmö; Department of Neurology (N.M.-C.), Skåne University Hospital, Lund; and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Erik Stomrud
- From the Clinical Memory Research Unit (A.K., S.J., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences, Lund University, Malmö; Memory Clinic (E.S., S.P., O.H.), Skåne University Hospital, Malmö; Department of Neurology (N.M.-C.), Skåne University Hospital, Lund; and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Sebastian Palmqvist
- From the Clinical Memory Research Unit (A.K., S.J., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences, Lund University, Malmö; Memory Clinic (E.S., S.P., O.H.), Skåne University Hospital, Malmö; Department of Neurology (N.M.-C.), Skåne University Hospital, Lund; and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Oskar Hansson
- From the Clinical Memory Research Unit (A.K., S.J., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences, Lund University, Malmö; Memory Clinic (E.S., S.P., O.H.), Skåne University Hospital, Malmö; Department of Neurology (N.M.-C.), Skåne University Hospital, Lund; and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Niklas Mattsson-Carlgren
- From the Clinical Memory Research Unit (A.K., S.J., E.S., S.P., O.H., N.M.-C.), Department of Clinical Sciences, Lund University, Malmö; Memory Clinic (E.S., S.P., O.H.), Skåne University Hospital, Malmö; Department of Neurology (N.M.-C.), Skåne University Hospital, Lund; and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Sweden
| |
Collapse
|
104
|
Cummings J, Kinney J. Biomarkers for Alzheimer's Disease: Context of Use, Qualification, and Roadmap for Clinical Implementation. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:952. [PMID: 35888671 PMCID: PMC9318582 DOI: 10.3390/medicina58070952] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 12/30/2022]
Abstract
Background and Objectives: The US Food and Drug Administration (FDA) defines a biomarker as a characteristic that is measured as an indicator of normal biological processes, pathogenic processes, or responses to an exposure or intervention. Biomarkers may be used in clinical care or as drug development tools (DDTs) in clinical trials. The goal of this review and perspective is to provide insight into the regulatory guidance for the use of biomarkers in clinical trials and clinical care. Materials and Methods: We reviewed FDA guidances relevant to biomarker use in clinical trials and their transition to use in clinical care. We identified instructive examples of these biomarkers in Alzheimer's disease (AD) drug development and their application in clinical practice. Results: For use in clinical trials, biomarkers must have a defined context of use (COU) as a risk/susceptibility, diagnostic, monitoring, predictive, prognostic, pharmacodynamic, or safety biomarker. A four-stage process defines the pathway to establish the regulatory acceptance of the COU for a biomarker including submission of a letter of intent, description of the qualification plan, submission of a full qualification package, and acceptance through a qualification recommendation. Biomarkers used in clinical care may be companion biomarkers, in vitro diagnostic devices (IVDs), or laboratory developed tests (LDTs). A five-phase biomarker development process has been proposed to structure the biomarker development process. Conclusions: Biomarkers are increasingly important in drug development and clinical care. Adherence to regulatory guidance for biomarkers used in clinical trials and patient care is required to advance these important drug development and clinical tools.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Pam Quirk Brain Health and Biomarker Laboratory, Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
| | | |
Collapse
|
105
|
Faldu KG, Shah JS. Alzheimer's disease: a scoping review of biomarker research and development for effective disease diagnosis. Expert Rev Mol Diagn 2022; 22:681-703. [PMID: 35855631 DOI: 10.1080/14737159.2022.2104639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 07/19/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is regarded as the foremost reason for neurodegeneration that prominently affects the geriatric population. Characterized by extracellular accumulation of amyloid-beta (Aβ), intracellular aggregation of hyperphosphorylated tau (p-tau), and neuronal degeneration that causes impairment of memory and cognition. Amyloid/tau/neurodegeneration (ATN) classification is utilized for research purposes and involves amyloid, tau, and neuronal injury staging through MRI, PET scanning, and CSF protein concentration estimations. CSF sampling is invasive, and MRI and PET scanning requires sophisticated radiological facilities which limit its widespread diagnostic use. ATN classification lacks effectiveness in preclinical AD. AREAS COVERED This publication intends to collate and review the existing biomarker profile and the current research and development of a new arsenal of biomarkers for AD pathology from different biological samples, microRNA (miRNA), proteomics, metabolomics, artificial intelligence, and machine learning for AD screening, diagnosis, prognosis, and monitoring of AD treatments. EXPERT OPINION It is an accepted observation that AD-related pathological changes occur over a long period of time before the first symptoms are observed providing ample opportunity for detection of biological alterations in various biological samples that can aid in early diagnosis and modify treatment outcomes.
Collapse
Affiliation(s)
- Khushboo Govind Faldu
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Jigna Samir Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| |
Collapse
|
106
|
Chen YH, Lin RR, Huang HF, Xue YY, Tao QQ. Microglial Activation, Tau Pathology, and Neurodegeneration Biomarkers Predict Longitudinal Cognitive Decline in Alzheimer's Disease Continuum. Front Aging Neurosci 2022; 14:848180. [PMID: 35847667 PMCID: PMC9280990 DOI: 10.3389/fnagi.2022.848180] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/12/2022] [Indexed: 01/02/2023] Open
Abstract
Purpose Biomarkers used for predicting longitudinal cognitive change in Alzheimer's disease (AD) continuum are still elusive. Tau pathology, neuroinflammation, and neurodegeneration are the leading candidate predictors. We aimed to determine these three aspects of biomarkers in cerebrospinal fluid (CSF) and plasma to predict longitudinal cognition status using Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. Patients and Methods A total of 430 subjects including, 96 cognitive normal (CN) with amyloid β (Aβ)-negative, 54 CN with Aβ-positive, 195 mild cognitive impairment (MCI) with Aβ-positive, and 85 AD with amyloid-positive (Aβ-positive are identified by CSF Aβ42/Aβ40 < 0.138). Aβ burden was evaluated by CSF and plasma Aβ42/Aβ40 ratio; tau pathology was evaluated by CSF and plasma phosphorylated-tau (p-tau181); microglial activation was measured by CSF soluble TREM2 (sTREM2) and progranulin (PGRN); neurodegeneration was measured by CSF and plasma t-tau and structural magnetic resonance imaging (MRI); cognition was examined annually over the subsequent 8 years using the Alzheimer's Disease Assessment Scale Cognition 13-item scale (ADAS13) and Mini-Mental State Exam (MMSE). Linear mixed-effects models (LME) were applied to assess the correlation between biomarkers and longitudinal cognition decline, as well as their effect size on the prediction of longitudinal cognitive decline. Results Baseline CSF Aβ42/Aβ40 ratio was decreased in MCI and AD compared to CN, while CSF p-tau181 and t-tau increased. Baseline CSF sTREM2 and PGRN did not show any differences in MCI and AD compared to CN. Baseline brain volumes (including the hippocampal, entorhinal, middle temporal lobe, and whole-brain) decreased in MCI and AD groups. For the longitudinal study, there were significant interaction effects of CSF p-tau181 × time, plasma p-tau181 × time, CSF sTREM2 × time, and brain volumes × time, indicating CSF, and plasma p-tau181, CSF sTREM2, and brain volumes could predict longitudinal cognition deterioration rate. CSF sTREM2, CSF, and plasma p-tau181 had similar medium prediction effects, while brain volumes showed stronger effects in predicting cognition decline. Conclusion Our study reported that baseline CSF sTREM2, CSF, and plasma p-tau181, as well as structural MRI, could predict longitudinal cognitive decline in subjects with positive AD pathology. Plasma p-tau181 can be used as a relatively noninvasive reliable biomarker for AD longitudinal cognition decline prediction.
Collapse
Affiliation(s)
- Yi-He Chen
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Rong-Rong Lin
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui-Feng Huang
- Department of Neurology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Yan-Yan Xue
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Qing-Qing Tao
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
107
|
Pan R, Luo S, Huang Q, Li W, Cai T, Lai K, Shi X. The Associations of Cerebrospinal Fluid Ferritin with Neurodegeneration and Neuroinflammation Along the Alzheimer's Disease Continuum. J Alzheimers Dis 2022; 88:1115-1125. [PMID: 35754266 DOI: 10.3233/jad-220002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Increasing evidence has suggested that iron accumulation plays an important role in the onset and development of Alzheimer's disease (AD). However, the potential mechanism remains unclear. OBJECTIVE The present study investigated the associations of cerebrospinal fluid (CSF) ferritin, an indicator for brain iron load, with neurodegenerative and inflammatory changes in AD. METHODS The study involved 302 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI). They were classified as normal controls (A-T-N-, n = 48), AD continuum (A+TN-, n = 46; A+TN+, n = 166), and suspected non-AD pathology (A-TN+, n = 42), according to the amyloid/tau/neurodegeneration (ATN) system. Group comparisons of CSF ferritin among groups were performed using one-way ANOVA. Linear regression models were used to test the relationships between CSF ferritin and cognitive assessments, and the associations between CSF ferritin and other biomarkers, respectively. RESULTS We found that CSF ferritin showed significant differences among the ATN groups, with higher concentration in more advanced categories (A+TN+). Furthermore, CSF ferritin level was independently related to cognitive performance (MMSE, ADAS-Cog13, and ADNI-mem). Linear regression analysis indicated positive relationships between CSF ferritin and phosphorylated tau and total tau, rather than Aβ42. Significant associations were revealed between CSF ferritin and inflammatory proteins, including TNF-α, TNFR1, TNFR2, ICAM1, VCAM1, TGF-β1, IL-9, and IP-10, respectively. CONCLUSION Our results provide new insight into iron dysfunction in AD pathology and highlight elevated brain iron as a possible mechanism of neurodegeneration and neuroinflammation along AD continuum.
Collapse
Affiliation(s)
- Rui Pan
- School of Nursing, Huizhou Health Sciences Polytechnic, Huizhou, Guangdong Province, P. R. China
| | - Shuyi Luo
- Department of Cardiothoracic Surgery, The Third People's Hospital of Huizhou, Huizhou, Guangdong Province, P. R. China
| | - Qing Huang
- School of Foreign Languages, Huizhou University, Huizhou, Guangdong Province, P. R. China
| | - Weiwei Li
- School of Clinical Medicine, Huizhou Health Sciences Polytechnic, Huizhou, Guangdong Province, P. R. China
| | - Tianshu Cai
- School of Medicine and Medical Laboratory Science, Huizhou Health Sciences Polytechnic, Huizhou, Guangdong Province, P. R. China
| | - Kelin Lai
- School of Clinical Medicine, Huizhou Health Sciences Polytechnic, Huizhou, Guangdong Province, P. R. China
| | - Xiaolei Shi
- School of Clinical Medicine, Huizhou Health Sciences Polytechnic, Huizhou, Guangdong Province, P. R. China.,School of Clinical Medicine, Huizhou Health Sciences Polytechnic, Huizhou, Guangdong Province, P. R. China
| | | |
Collapse
|
108
|
Fernandez-Alvarez M, Atienza M, Zallo F, Matute C, Capetillo-Zarate E, Cantero JL. Linking Plasma Amyloid Beta and Neurofilament Light Chain to Intracortical Myelin Content in Cognitively Normal Older Adults. Front Aging Neurosci 2022; 14:896848. [PMID: 35783126 PMCID: PMC9247578 DOI: 10.3389/fnagi.2022.896848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022] Open
Abstract
Evidence suggests that lightly myelinated cortical regions are vulnerable to aging and Alzheimer’s disease (AD). However, it remains unknown whether plasma markers of amyloid and neurodegeneration are related to deficits in intracortical myelin content, and whether this relationship, in turn, is associated with altered patterns of resting-state functional connectivity (rs-FC). To shed light into these questions, plasma levels of amyloid-β fragment 1–42 (Aβ1–42) and neurofilament light chain (NfL) were measured using ultra-sensitive single-molecule array (Simoa) assays, and the intracortical myelin content was estimated with the ratio T1-weigthed/T2-weighted (T1w/T2w) in 133 cognitively normal older adults. We assessed: (i) whether plasma Aβ1–42 and/or NfL levels were associated with intracortical myelin content at different cortical depths and (ii) whether cortical regions showing myelin reductions also exhibited altered rs-FC patterns. Surface-based multiple regression analyses revealed that lower plasma Aβ1–42 and higher plasma NfL were associated with lower myelin content in temporo-parietal-occipital regions and the insular cortex, respectively. Whereas the association with Aβ1–42 decreased with depth, the NfL-myelin relationship was most evident in the innermost layer. Older individuals with higher plasma NfL levels also exhibited altered rs-FC between the insula and medial orbitofrontal cortex. Together, these findings establish a link between plasma markers of amyloid/neurodegeneration and intracortical myelin content in cognitively normal older adults, and support the role of plasma NfL in boosting aberrant FC patterns of the insular cortex, a central brain hub highly vulnerable to aging and neurodegeneration.
Collapse
Affiliation(s)
- Marina Fernandez-Alvarez
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Fatima Zallo
- Departamento de Neurociencias, Achucarro Basque Center for Neuroscience, Universidad del País Vasco, Leioa, Spain
| | - Carlos Matute
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Departamento de Neurociencias, Achucarro Basque Center for Neuroscience, Universidad del País Vasco, Leioa, Spain
| | - Estibaliz Capetillo-Zarate
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Departamento de Neurociencias, Achucarro Basque Center for Neuroscience, Universidad del País Vasco, Leioa, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Jose L. Cantero
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- *Correspondence: Jose L. Cantero,
| |
Collapse
|
109
|
Tang X, Yv H, Wang F, Wang J, Liu S, Wu X, Dong R, Lin X, Wang B, Bi Y. The Relationship Between Suboptimal Social Networks and Postoperative Delirium: The PNDABLE Study. Front Aging Neurosci 2022; 14:851368. [PMID: 35769605 PMCID: PMC9235411 DOI: 10.3389/fnagi.2022.851368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 05/25/2022] [Indexed: 11/19/2022] Open
Abstract
Background Although it has been proven that social networks are related to cognition, studies are conducted to characterize the correlation between social networks and postoperative delirium (POD). Objective We investigated whether suboptimal social networks are a risk factor for POD, and to verify whether different levels of intimacy in the same social relationship can affect the concentration of cerebrospinal fluid (CSF) biomarkers, such as amyloid-β (Aβ42), total tau (T-tau), and phosphorylated tau (P-tau), and the mediating role of CSF biomarkers between social network and POD in middle-aged and elderly Han people. Methods Our study recruited 743 participants from The Perioperative Neurocognitive Disorder and Biomarker Lifestyle (PNDABLE) study. Confusion Assessment Method (CAM) was used to evaluate the incidence of POD and its severity was measured using the Memorial Delirium Assessment Scale (MDAS). The social networks were measured using self-reported questionnaires about social ties. Mann–Whitney U test, Logistic Regression and Independent-samples test were used for Statistical Analysis. Results The incidence of POD was 20.7%. Mann–Whitney U test showed that the total score of the social network was associated with POD (P < 0.001). Independent-samples test showed that different levels of intimacy in the same social relationship were significantly associated with CSF POD biomarkers, and mediation analyses revealed that the association between suboptimal social networks and POD was partially mediated by T-tau (proportion: 20%), P-tau (proportion: 33%), Aβ42/T-tau (proportion: 14%), and Aβ42/P-tau (proportion: 15%). Conclusion Having suboptimal social networks is a risk factor for POD in middle-aged and elderly Han people. CSF POD biomarkers can mediate the correlation between suboptimal social networks and POD, which is mainly mediated by tau protein. Clinical Trial Registration www.chictr.org.cn, identifier ChiCTR2000033439.
Collapse
Affiliation(s)
- Xinhui Tang
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Hui Yv
- Department of Anesthesiology, Qingdao Eye Hospital of Shandong First Medical University, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Fei Wang
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Jiahan Wang
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Siyv Liu
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xiaoyue Wu
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Rui Dong
- Department of Anesthesiology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Xu Lin
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Bin Wang
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Yanlin Bi
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
- *Correspondence: Yanlin Bi,
| |
Collapse
|
110
|
Haußmann R, Homeyer P, Brandt MD, Donix M. [Prognostic and diagnostic value of cerebrospinal fluid analysis in neurodegenerative dementia diseases]. DER NERVENARZT 2022; 93:1236-1242. [PMID: 35670835 DOI: 10.1007/s00115-022-01339-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 11/25/2022]
Abstract
Cerebrospinal fluid (CSF) analysis is an important diagnostic tool in the assessment of dementia. For the differentiation of Alzheimer's disease from other etiologies of dementia syndromes, established biological markers could be helpful to confirm a distinctive neuropathology. Whereas negative CSF findings can rule out the majority of primarily neurodegenerative disorders, overlapping biomarker profiles remain a diagnostic challenge. Therefore, it is important to interpret CSF results within a specific clinical context. Furthermore, atypical CSF data can be challenging and require profound knowledge of preanalytics, biomarker profiles and the broad spectrum of diseases associated with cognitive decline. Beyond the Alzheimer's disease clinical spectrum, current studies aim at investigating CSF biomarkers to better differentiate tauopathies, TDP43(Transactive response DNA binding protein 43 kDa)-proteinopathies and synucleinopathies.
Collapse
Affiliation(s)
- R Haußmann
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.
| | - P Homeyer
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland
| | - M D Brandt
- Klinik und Poliklinik für Neurologie, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.,DZNE, Deutsches Zentrum für Neurodegenerative Erkrankungen, Dresden, Deutschland
| | - M Donix
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.,DZNE, Deutsches Zentrum für Neurodegenerative Erkrankungen, Dresden, Deutschland
| |
Collapse
|
111
|
Sawa M, Overk C, Becker A, Derse D, Albay R, Weldy K, Salehi A, Beach TG, Doran E, Head E, Yu YE, Mobley WC. Impact of increased APP gene dose in Down syndrome and the Dp16 mouse model. Alzheimers Dement 2022; 18:1203-1234. [PMID: 34757693 PMCID: PMC9085977 DOI: 10.1002/alz.12463] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/21/2021] [Accepted: 07/29/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION People with Down syndrome (DS) are predisposed to Alzheimer's disease (AD). The amyloid hypothesis informs studies of AD. In AD-DS, but not sporadic AD, increased APP copy number is necessary, defining the APP gene dose hypothesis. Which amyloid precursor protein (APP) products contribute needs to be determined. METHODS Brain levels of full-length protein (fl-hAPP), C-terminal fragments (hCTFs), and amyloid beta (Aβ) peptides were measured in DS, AD-DS, non-demented controls (ND), and sporadic AD cases. The APP gene-dose hypothesis was evaluated in the Dp16 model. RESULTS DS and AD-DS differed from ND and AD for all APP products. In AD-DS, Aβ42 and Aβ40 levels exceeded AD. APP products were increased in the Dp16 model; increased APP gene dose was necessary for loss of vulnerable neurons, tau pathology, and activation of astrocytes and microglia. DISCUSSION Increases in APP products other than Aβ distinguished AD-DS from AD. Deciphering AD-DS pathogenesis necessitates deciphering which APP products contribute and how.
Collapse
Affiliation(s)
- Mariko Sawa
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Cassia Overk
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ann Becker
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Dominique Derse
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ricardo Albay
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Kim Weldy
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ahmad Salehi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305
| | - Thomas G. Beach
- Brain and Body Donation Program, Banner Sun Health Research Institute, Sun City, AZ 85351
| | - Eric Doran
- Department of Pediatrics, University of California, Irvine, CA, 92697
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, 92697
| | - Y. Eugene Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - William C Mobley
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624,Correspondence to: William Mobley M.D., Department of Neurosciences, UCSD School of Medicine, 9500 Gilman Drive, GPL 355, La Jolla, CA 92093-0624;
| |
Collapse
|
112
|
Soo SA, Zailan FZ, Tan JY, Sandhu GK, Wong BYX, Wang BZ, Ng ASL, Chiew HJ, Ng KP, Kandiah N. Safety and Usefulness of Lumbar Puncture for the Diagnosis and Management of Young-Onset Cognitive Disorders. J Alzheimers Dis 2022; 87:479-488. [PMID: 35275537 DOI: 10.3233/jad-215453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Young-onset cognitive disorders (YOCD) often manifests with complex and atypical presentations due to underlying heterogenous pathologies. Therefore, a biomarker-based evaluation will allow for timely diagnosis and definitive management. OBJECTIVE Here, we evaluated the safety and usefulness of cerebrospinal fluid (CSF) sampling through lumbar puncture (LP) in YOCD patients in a tertiary clinical setting. METHODS Patients with mild cognitive impairment (MCI) and mild dementia with age of onset between 45-64 years were evaluated. Patients underwent magnetic resonance imaging and their medial temporal lobe atrophy (MTA) was rated. LP side-effects and the impact of the CSF findings on diagnosis and management were analyzed. RESULTS 142 patients (53 (37.32%) MCI, 51 (35.92%) dementia of the Alzheimer's disease [DAT] type, and 38 (26.76%) non-AD type dementia) who underwent LP between 2015 to 2021 were analyzed. Using post-LP results and MTA ratings, 74 (52.11%) patients met the AT(N) criteria for AD. 56 (39.44%) patients (28 out of 53 (50.0%) MCI, 12 out of 51 (21.43%) DAT, and 16 out of 38 (28.57%) non-AD dementia) had a change in diagnosis following LP. 13 (9.15%) patients developed side-effects post-LP (11 (84.62%) patients had headache, 1 (7.69%) patient had backache, and 1 (7.69%) patient had headache and backache). 32 (22.54%) patients had a change in management post-LP, 24 (75.0%) had medication changes, 10 (31.30%) had referrals to other specialists, and 3 (9.40%) was referred for clinical trial with disease modifying interventions. CONCLUSION LP is well-tolerated in YOCD and can bring about relevant clinical decisions with regards to the diagnosis and management of this complex clinical condition.
Collapse
Affiliation(s)
- See Ann Soo
- Department of Neurology, National Neuroscience Institute, Singapore
| | | | - Jayne Yi Tan
- Department of Neurology, National Neuroscience Institute, Singapore
| | | | | | | | | | - Hui Jin Chiew
- Department of Neurology, National Neuroscience Institute, Singapore
| | - Kok Pin Ng
- Department of Neurology, National Neuroscience Institute, Singapore.,Duke NUS Medical School, Singapore.,Lee Kong Chian School of Medicine-NTU, Singapore
| | - Nagaendran Kandiah
- Department of Neurology, National Neuroscience Institute, Singapore.,Duke NUS Medical School, Singapore.,Lee Kong Chian School of Medicine-NTU, Singapore
| |
Collapse
|
113
|
Kang MS, Shin M, Ottoy J, Aliaga AA, Mathotaarachchi S, Quispialaya K, Pascoal TA, Collins DL, Chakravarty MM, Mathieu A, Sandelius Å, Blennow K, Zetterberg H, Massarweh G, Soucy JP, Cuello AC, Gauthier S, Waterston M, Yoganathan N, Lessard E, Haqqani A, Rennie K, Stanimirovic D, Chakravarthy B, Rosa-Neto P. Preclinical in vivo longitudinal assessment of KG207-M as a disease-modifying Alzheimer's disease therapeutic. J Cereb Blood Flow Metab 2022; 42:788-801. [PMID: 34378436 PMCID: PMC9014686 DOI: 10.1177/0271678x211035625] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In vivo biomarker abnormalities provide measures to monitor therapeutic interventions targeting amyloid-β pathology as well as its effects on downstream processes associated with Alzheimer's disease pathophysiology. Here, we applied an in vivo longitudinal study design combined with imaging and cerebrospinal fluid biomarkers, mirroring those used in human clinical trials to assess the efficacy of a novel brain-penetrating anti-amyloid fusion protein treatment in the McGill-R-Thy1-APP transgenic rat model. The bi-functional fusion protein consisted of a blood-brain barrier crossing single domain antibody (FC5) fused to an amyloid-β oligomer-binding peptide (ABP) via Fc fragment of mouse IgG (FC5-mFc2a-ABP). A five-week treatment with FC5-mFc2a-ABP (loading dose of 30 mg/Kg/iv followed by 15 mg/Kg/week/iv for four weeks) substantially reduced brain amyloid-β levels as measured by positron emission tomography and increased the cerebrospinal fluid amyloid-β42/40 ratio. In addition, the 5-week treatment rectified the cerebrospinal fluid neurofilament light chain concentrations, resting-state functional connectivity, and hippocampal atrophy measured using magnetic resonance imaging. Finally, FC5-mFc2a-ABP (referred to as KG207-M) treatment did not induce amyloid-related imaging abnormalities such as microhemorrhage. Together, this study demonstrates the translational values of the designed preclinical studies for the assessment of novel therapies based on the clinical biomarkers providing tangible metrics for designing early-stage clinical trials.
Collapse
Affiliation(s)
- Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada.,McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | - Monica Shin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada
| | - Julie Ottoy
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada
| | - Arturo Aliaga Aliaga
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada.,McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada
| | - Kely Quispialaya
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada
| | - D Louis Collins
- McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | | | - Axel Mathieu
- Douglas Mental Health University Institute, Montreal, Canada
| | - Åsa Sandelius
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, 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.,UK Dementia Research Institute at UCL, London, UK.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Gassan Massarweh
- McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | - Jean-Paul Soucy
- McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada.,McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | | | | | - Etienne Lessard
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Arsalan Haqqani
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Kerry Rennie
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Danica Stanimirovic
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Balu Chakravarthy
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada.,McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
114
|
Amft M, Ortner M, Eichenlaub U, Goldhardt O, Diehl-Schmid J, Hedderich DM, Yakushev I, Grimmer T. The cerebrospinal fluid biomarker ratio Aβ42/40 identifies amyloid positron emission tomography positivity better than Aβ42 alone in a heterogeneous memory clinic cohort. Alzheimers Res Ther 2022; 14:60. [PMID: 35473631 PMCID: PMC9044878 DOI: 10.1186/s13195-022-01003-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/08/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) analysis for detecting amyloid positivity may be as reliable as positron emission tomography (PET). We evaluated the performance of the amyloid beta (Aβ)42/40 ratio for predicting amyloid positivity by PET, compared with Aβ42 alone, and phosphorylated tau 181 (pTau181)/Aβ42 and total tau (tTau)/Aβ42 ratios, using fully automated CSF immunoassays (Roche Diagnostics International Ltd, Rotkreuz, Switzerland) in a heterogeneous cohort of patients with a range of cognitive disorders reflecting the typical population of a memory clinic. METHODS CSF samples from 103 patients with known amyloid PET status (PET positive = 54; PET negative = 49) were retrospectively selected from one site in Germany; 71 patients were undergoing treatment for mild cognitive impairment (n = 44) or mild-to-moderate dementia (n = 27) due to Alzheimer's disease (AD), and 32 patients were undergoing treatment for non-AD-related cognitive disorders. Aβ42, pTau181, and tTau concentrations were measured in CSF samples using the respective Elecsys® CSF immunoassays modified for use on the cobas e 411 analyzer; Aβ40 concentrations were measured using a non-commercially available robust prototype assay. Sensitivities/specificities for amyloid positivity cut-offs (Youden-derived and pre-defined) were calculated, and receiver operating characteristic analyses determined area under the curve (AUC) versus amyloid PET status. Limitations include a small sample size, use of a pre-analytical protocol not in accordance with the Elecsys CSF immunoassay method sheets, and the lack of a pre-defined cut-off for Aβ42/40. RESULTS Point estimates for sensitivity and specificity of CSF biomarkers and biomarker ratios versus amyloid PET were 0.93 and 0.57 for Aβ42, 0.96 and 0.69 for pTau181/Aβ42, 0.92 and 0.69 for tTau/Aβ42, and 0.94 and 0.82 for Aβ42/40. For AUCs, point estimates (95% confidence intervals) versus amyloid PET were 0.78 (0.68-0.88) for Aβ42, 0.88 (0.81-0.95) for pTau181/Aβ42, 0.87 (0.80-0.95) for tTau/Aβ42, and 0.90 (0.83-0.97) for Aβ42/40. CONCLUSIONS CSF Aβ42/40 ratio can predict PET amyloid positivity with high accuracy in patients with a range of cognitive disorders when evaluating Aβ pathology independent of tau and neurodegeneration for research purposes. The performance of Aβ42/40 was comparable with pTau181/Aβ42 and tTau/Aβ42 used in clinical practice and better than Aβ42 alone.
Collapse
Affiliation(s)
- Michaela Amft
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Marion Ortner
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Udo Eichenlaub
- Clinical Development Medical Affairs, Roche Diagnostic Solutions, Roche Diagnostics GmbH, Penzberg, Germany
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Dennis M Hedderich
- Department of Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Igor Yakushev
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.
| |
Collapse
|
115
|
Mielke MM, Aakre JA, Algeciras-Schimnich A, Proctor NK, Machulda MM, Eichenlaub U, Knopman DS, Vemuri P, Graff-Radford J, Jack CR, Petersen RC, Dage JL. Comparison of CSF phosphorylated tau 181 and 217 for cognitive decline. Alzheimers Dement 2022; 18:602-611. [PMID: 34310832 PMCID: PMC8789950 DOI: 10.1002/alz.12415] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 11/08/2022]
Abstract
INTRODUCTION The prognostic utility of cerebrospinal fluid (CSF) phosphorylated tau 217 (p-tau217) and p-tau181 is not understood. METHODS Analyses included 753 Mayo Clinic Study on Aging participants (median age = 71.6; 57% male). CSF amyloid beta (Aβ)42 and p-tau181 were measured with Elecsys immunoassays. CSF p-tau181 and p-tau217 were also measured with Meso Scale Discovery (MSD). We used Cox proportional hazards models for risk of mild cognitive impairment (MCI) and linear mixed models for risk of global and domain-specific cognitive decline and cortical thickness. Analyses were stratified by elevated brain amyloid based on CSF Aβ42 or amyloid positron emission tomography for those with imaging. RESULTS CSF p-tau217 was superior to p-tau181 for the diagnosis of Alzheimer's disease (AD) pathology. CSF MSD p-tau181 and p-tau217 were associated with risk of MCI among amyloid-positive individuals. Differences between CSF p-tau measures predicting cortical thickness were subtle. DISCUSSION There are subtle differences for CSF p-tau217 and p-tau181 as prognostic AD markers.
Collapse
Affiliation(s)
- Michelle M. Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jeremiah A. Aakre
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Mary M. Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | - Ronald C. Petersen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
116
|
Kettunen P, Bjerke M, Eckerström C, Jonsson M, Zetterberg H, Blennow K, Svensson J, Wallin A. Blood-brain barrier dysfunction and reduced cerebrospinal fluid levels of soluble amyloid precursor protein-β in patients with subcortical small-vessel disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12296. [PMID: 35356486 PMCID: PMC8949877 DOI: 10.1002/dad2.12296] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/13/2022] [Accepted: 01/30/2022] [Indexed: 11/30/2022]
Abstract
Introduction Subcortical small-vessel disease (SSVD) is the most common vascular cognitive disorder. However, because no disease-specific cerebrospinal fluid (CSF) biomarkers are available for SSVD, our aim was to identify such markers. Methods We included 170 healthy controls and patients from the Gothenburg Mild Cognitive Impairment (MCI) study clinically diagnosed with SSVD dementia, Alzheimer's disease (AD), or mixed AD/SSVD. We quantified CSF levels of amyloid-β (Aβ)x-38, Aβx-40, Aβx-42, as well as soluble amyloid precursor protein (sAPP)-α and sAPP-β. Results sAPP-β was lower in SSVD patients than in AD patients and controls. Receiver-operating characteristic (ROC) analyses showed that sAPP-β moderately separated SSVD from AD and controls. Moreover, the CSF/serum albumin ratio was elevated exclusively in SSVD and could moderately separate SSVD from the other groups in ROC analyses. Discussion SSVD has a biomarker profile that differs from that of AD and controls, and to some extent also from mixed AD/SSVD, suggesting that signs of blood-brain barrier (BBB) dysfunction and sAPP-β could be additional tools to diagnose SSVD. Highlights Patients with subcortical small-vessel disease (SSVD) exhibited reduced levels of sAPP-β and disturbances of the blood-brain barrier (BBB).This biochemical pattern is different from that of Alzheimer's disease (AD) and to some degree from that of mixed AD/SSVD.Our findings are speaking in favor of the concept that SSVD is a distinct vascular cognitive disorder (VCD) form.
Collapse
Affiliation(s)
- Petronella Kettunen
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Maria Bjerke
- Laboratory of Neurochemistry, Department of Clinical Biology and Center for NeurosciencesUniversitair Ziekenhuis BrusselBrusselsBelgium
- Department of Biomedical SciencesInstitute Born‐BungeUniversity of AntwerpAntwerpBelgium
| | - Carl Eckerström
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Immunology and Transfusion MedicineSahlgrenska University HospitalGothenburgSweden
| | - Michael Jonsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Johan Svensson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Region Västra Götaland, Department of Internal MedicineSkaraborg Central HospitalSkövdeSweden
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
117
|
Alegret M, Sotolongo-Grau O, de Antonio EE, Pérez-Cordón A, Orellana A, Espinosa A, Gil S, Jiménez D, Ortega G, Sanabria A, Roberto N, Hernández I, Rosende-Roca M, Tartari JP, Alarcon-Martin E, de Rojas I, Montrreal L, Morató X, Cano A, Rentz DM, Tárraga L, Ruiz A, Valero S, Marquié M, Boada M. Automatized FACEmemory® scoring is related to Alzheimer's disease phenotype and biomarkers in early-onset mild cognitive impairment: the BIOFACE cohort. Alzheimers Res Ther 2022; 14:43. [PMID: 35303916 PMCID: PMC8933921 DOI: 10.1186/s13195-022-00988-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/10/2022] [Indexed: 11/13/2022]
Abstract
Background FACEmemory® is the first computerized, self-administered verbal episodic memory test with voice recognition. It can be conducted under minimal supervision and contains an automatic scoring system to avoid administrator errors. Moreover, it is suitable for discriminating between cognitively healthy and amnestic mild cognitive impairment (MCI) individuals, and it is associated with Alzheimer’s disease (AD) cerebrospinal fluid (CSF) biomarkers. This study aimed to determine whether FACEmemory scoring is related to performance on classical memory tests and to AD biomarkers of brain magnetic resonance imaging (MRI) and CSF in patients with early-onset MCI (EOMCI). Methods Ninety-four patients with EOMCI from the BIOFACE study completed FACEmemory, classical memory tests (the Spanish version of the Word Free and Cued Selective Reminding Test -FCSRT-, the Word List from the Wechsler Memory Scale, third edition, and the Spanish version of the Rey–Osterrieth Complex Figure Test), and a brain MRI. Eighty-two individuals also underwent a lumbar puncture. Results FACEmemory scoring was moderately correlated with FCSRT scoring. With regard to neuroimaging MRI results, worse execution on FACEmemory was associated with lower cortical volume in the right prefrontal and inferior parietal areas, along with the left temporal and associative occipital areas. Moreover, the total FACEmemory score correlated with CSF AD biomarkers (Aβ1-42/Aβ1-40 ratio, p181-tau, and Aβ1-42/p181-tau ratio). When performance on FACEmemory was compared among the ATN classification groups, significant differences between the AD group and normal and SNAP groups were found. Conclusions FACEmemory is a promising tool for detecting memory deficits sensitive to early-onset AD, but it also allows the detection of memory-impaired cases due to other etiologies. Our findings suggest that FACEmemory scoring can detect the AD endophenotype and that it is also associated with AD-related changes in MRI and CSF in patients with EOMCI. The computerized FACEmemory tool might be an opportunity to facilitate early detection of MCI in younger people than 65, who have a growing interest in new technologies.
Collapse
Affiliation(s)
- Montserrat Alegret
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain. .,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| | - Oscar Sotolongo-Grau
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ester Esteban de Antonio
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Alba Pérez-Cordón
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Adelina Orellana
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Espinosa
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Gil
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Jiménez
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Gemma Ortega
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Angela Sanabria
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Natalia Roberto
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Hernández
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Maitee Rosende-Roca
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Emilio Alarcon-Martin
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Montrreal
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Xavier Morató
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Amanda Cano
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Dorene M Rentz
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
118
|
Cullen N, Janelidze S, Palmqvist S, Stomrud E, Mattsson-Carlgren N, Hansson O. Association of CSF Aβ 38 Levels With Risk of Alzheimer Disease-Related Decline. Neurology 2022; 98:e958-e967. [PMID: 34937781 PMCID: PMC8901176 DOI: 10.1212/wnl.0000000000013228] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/08/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Experimental studies suggest that the balance between short and long β-amyloid (Aβ) species might modulate the toxic effects of Aβ in Alzheimer disease (AD), but clinical evidence is lacking. We studied whether Aβ38 levels in CSF relate to risk of AD dementia and cognitive decline. METHODS CSF Aβ38 levels were measured in 656 individuals across 2 clinical cohorts: the Swedish BioFINDER study and the Alzheimer's Disease Neuroimaging Initiative (ADNI). Cox regression models were used to evaluate the association between baseline Aβ38 levels and risk of AD dementia in AD biomarker-positive individuals (AD+; determined by CSF phosphorylated tau [P-tau]/Aβ42 ratio) with subjective cognitive decline (SCD) or mild cognitive impairment (MCI). Linear mixed-effects models were used to evaluate the association between baseline Aβ38 levels and cognitive decline as measured by the Mini-Mental State Examination (MMSE) in AD+ participants with SCD, MCI, or AD dementia. RESULTS In the BioFINDER cohort, high Aβ38 levels were associated with slower decline in MMSE score (β = 0.30 points per SD, p = 0.001) and with lower risk of conversion to AD dementia (hazard ratio 0.83 per SD, p = 0.03). In the ADNI cohort, higher Aβ38 levels were associated with less decline in MMSE score (β = 0.27, p = 0.01) but not risk of conversion to AD dementia (p = 0.66). Aβ38 levels in both cohorts were significantly associated with both cognitive and clinical outcomes when further adjusted for CSF P-tau or CSF Aβ42 levels. DISCUSSION Higher CSF Aβ38 levels are associated with lower risk of AD-related changes in 2 independent clinical cohorts. These findings suggest that γ-secretase modulators could be effective as disease-altering therapy. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov Identifier: NCT03174938.
Collapse
Affiliation(s)
- Nicholas Cullen
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden.
| | - Shorena Janelidze
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden
| | - Sebastian Palmqvist
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden
| | - Erik Stomrud
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden
| | - Oskar Hansson
- From the Clinical Memory Research Unit (N.C., S.J., S.P., E.S., N.M.-C., O.H.), Department of Clinical Sciences Malmö, Faculty of Medicine, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University; Memory Clinic (S.P., E.S., O.H.), Skåne University Hospital, Malmö; and Department of Neurology (N.M.-C.), Skåne University Hospital, Lund, Sweden
| |
Collapse
|
119
|
Margraf NG, Jensen-Kondering U, Weiler C, Leypoldt F, Maetzler W, Philippen S, Bartsch T, Flüh C, Röcken C, Möller B, Royl G, Neumann A, Brüggemann N, Roeben B, Schulte C, Bender B, Berg D, Kuhlenbäumer G. Cerebrospinal Fluid Biomarkers in Cerebral Amyloid Angiopathy: New Data and Quantitative Meta-Analysis. Front Aging Neurosci 2022; 14:783996. [PMID: 35237145 PMCID: PMC8884145 DOI: 10.3389/fnagi.2022.783996] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/03/2022] [Indexed: 01/31/2023] Open
Abstract
Background To evaluate the diagnostic accuracy of cerebrospinal fluid (CSF) biomarkers in patients with probable cerebral amyloid angiopathy (CAA) according to the modified Boston criteria in a retrospective multicentric cohort. Methods Beta-amyloid 1-40 (Aβ40), beta-amyloid 1-42 (Aβ42), total tau (t-tau), and phosphorylated tau 181 (p-tau181) were measured in 31 patients with probable CAA, 28 patients with Alzheimer’s disease (AD), and 30 controls. Receiver-operating characteristics (ROC) analyses were performed for the measured parameters as well as the Aβ42/40 ratio to estimate diagnostic parameters. A meta-analysis of all amenable published studies was conducted. Results In our data Aβ42/40 (AUC 0.88) discriminated best between CAA and controls while Aβ40 did not perform well (AUC 0.63). Differentiating between CAA and AD, p-tau181 (AUC 0.75) discriminated best in this study while Aβ40 (AUC 0.58) and Aβ42 (AUC 0.54) provided no discrimination. In the meta-analysis, Aβ42/40 (AUC 0.90) showed the best discrimination between CAA and controls followed by t-tau (AUC 0.79), Aβ40 (AUC 0.76), and p-tau181 (AUC 0.71). P-tau181 (AUC 0.76), Aβ40 (AUC 0.73), and t-tau (AUC 0.71) differentiated comparably between AD and CAA while Aβ42 (AUC 0.54) did not. In agreement with studies examining AD biomarkers, Aβ42/40 discriminated excellently between AD and controls (AUC 0.92–0.96) in this study as well as the meta-analysis. Conclusion The analyzed parameters differentiate between controls and CAA with clinically useful accuracy (AUC > ∼0.85) but not between CAA and AD. Since there is a neuropathological, clinical and diagnostic continuum between CAA and AD, other diagnostic markers, e.g., novel CSF biomarkers or other parameters might be more successful.
Collapse
Affiliation(s)
- Nils G. Margraf
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
- *Correspondence: Nils G. Margraf,
| | - Ulf Jensen-Kondering
- Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
- Department of Neuroradiology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Caroline Weiler
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
| | - Frank Leypoldt
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
- Institute of Clinical Chemistry, University Medical Center Schleswig-Holstein, Kiel/Lübeck, Germany
| | - Walter Maetzler
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
| | - Sarah Philippen
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
| | - Thorsten Bartsch
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
| | - Charlotte Flüh
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
| | - Christoph Röcken
- Department of Pathology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
| | - Bettina Möller
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
| | - Georg Royl
- Department of Neurology, University Medical Center Schleswig Holstein, Campus Lübeck, Lübeck, Germany
| | - Alexander Neumann
- Department of Neuroradiology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Norbert Brüggemann
- Department of Neurology, University Medical Center Schleswig Holstein, Campus Lübeck, Lübeck, Germany
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Benjamin Roeben
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Claudia Schulte
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Benjamin Bender
- Department of Neuroradiology, Diagnostical and Interventional Neuroradiology, University Hospital of Tübingen, Tübingen, Germany
| | - Daniela Berg
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Gregor Kuhlenbäumer
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany
| |
Collapse
|
120
|
Clinical usefulness of the CSF β-amyloid Aβ1-42/Aβ1-40 ratio for Alzheimer's disease diagnosis: a retrospective study in a Belgian academic hospital. Acta Neurol Belg 2022; 122:245-247. [PMID: 35037226 DOI: 10.1007/s13760-021-01846-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/06/2021] [Indexed: 11/01/2022]
|
121
|
Mahaman YAR, Embaye KS, Huang F, Li L, Zhu F, Wang JZ, Liu R, Feng J, Wang X. Biomarkers used in Alzheimer's disease diagnosis, treatment, and prevention. Ageing Res Rev 2022; 74:101544. [PMID: 34933129 DOI: 10.1016/j.arr.2021.101544] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD), being the number one in terms of dementia burden, is an insidious age-related neurodegenerative disease and is presently considered a global public health threat. Its main histological hallmarks are the Aβ senile plaques and the P-tau neurofibrillary tangles, while clinically it is marked by a progressive cognitive decline that reflects the underlying synaptic loss and neurodegeneration. Many of the drug therapies targeting the two pathological hallmarks namely Aβ and P-tau have been proven futile. This is probably attributed to the initiation of therapy at a stage where cognitive alterations are already obvious. In other words, the underlying neuropathological changes are at a stage where these drugs lack any therapeutic value in reversing the damage. Therefore, there is an urgent need to start treatment in the very early stage where these changes can be reversed, and hence, early diagnosis is of primordial importance. To this aim, the use of robust and informative biomarkers that could provide accurate diagnosis preferably at an earlier phase of the disease is of the essence. To date, several biomarkers have been established that, to a different extent, allow researchers and clinicians to evaluate, diagnose, and more specially exclude other related pathologies. In this study, we extensively reviewed data on the currently explored biomarkers in terms of AD pathology-specific and non-specific biomarkers and highlighted the recent developments in the diagnostic and theragnostic domains. In the end, we have presented a separate elaboration on aspects of future perspectives and concluding remarks.
Collapse
|
122
|
Morozova A, Zorkina Y, Abramova O, Pavlova O, Pavlov K, Soloveva K, Volkova M, Alekseeva P, Andryshchenko A, Kostyuk G, Gurina O, Chekhonin V. Neurobiological Highlights of Cognitive Impairment in Psychiatric Disorders. Int J Mol Sci 2022; 23:1217. [PMID: 35163141 PMCID: PMC8835608 DOI: 10.3390/ijms23031217] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
This review is focused on several psychiatric disorders in which cognitive impairment is a major component of the disease, influencing life quality. There are plenty of data proving that cognitive impairment accompanies and even underlies some psychiatric disorders. In addition, sources provide information on the biological background of cognitive problems associated with mental illness. This scientific review aims to summarize the current knowledge about neurobiological mechanisms of cognitive impairment in people with schizophrenia, depression, mild cognitive impairment and dementia (including Alzheimer's disease).The review provides data about the prevalence of cognitive impairment in people with mental illness and associated biological markers.
Collapse
Affiliation(s)
- Anna Morozova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Yana Zorkina
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Olga Abramova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Olga Pavlova
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Konstantin Pavlov
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Kristina Soloveva
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Maria Volkova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Polina Alekseeva
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Alisa Andryshchenko
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Georgiy Kostyuk
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Olga Gurina
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Vladimir Chekhonin
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
123
|
Teipel SJ, Dyrba M, Vergallo A, Lista S, Habert MO, Potier MC, Lamari F, Dubois B, Hampel H, Grothe MJ. Partial Volume Correction Increases the Sensitivity of 18F-Florbetapir-Positron Emission Tomography for the Detection of Early Stage Amyloidosis. Front Aging Neurosci 2022; 13:748198. [PMID: 35002673 PMCID: PMC8729321 DOI: 10.3389/fnagi.2021.748198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/05/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose: To test whether correcting for unspecific signal from the cerebral white matter increases the sensitivity of amyloid-PET for early stages of cerebral amyloidosis. Methods: We analyzed 18F-Florbetapir-PET and cerebrospinal fluid (CSF) Aβ42 data from 600 older individuals enrolled in the Alzheimer’s Disease Neuroimaging Initiative (ADNI), including people with normal cognition, mild cognitive impairment (MCI), and Alzheimer’s disease (AD) dementia. We determined whether three compartmental partial volume correction (PVC-3), explicitly modeling signal spill-in from white matter, significantly improved the association of CSF Aβ42 levels with global 18F-Florbetapir-PET values compared with standard processing without PVC (non-PVC) and a widely used two-compartmental PVC method (PVC-2). In additional voxel-wise analyses, we determined the sensitivity of PVC-3 compared with non-PVC and PVC-2 for detecting early regional amyloid build-up as modeled by decreasing CSF Aβ42 levels. For replication, we included an independent sample of 43 older individuals with subjective memory complaints from the INveStIGation of AlzHeimer’s PredicTors cohort (INSIGHT-preAD study). Results: In the ADNI sample, PVC-3 18F-Florbetapir-PET values normalized to whole cerebellum signal showed significantly stronger associations with CSF Aβ42 levels than non-PVC or PVC-2, particularly in the lower range of amyloid levels. These effects were replicated in the INSIGHT-preAD sample. PVC-3 18F-Florbetapir-PET data detected regional amyloid build-up already at higher (less abnormal) CSF Aβ42 levels than non-PVC or PVC-2 data. Conclusion: A PVC approach that explicitly models unspecific white matter binding improves the sensitivity of amyloid-PET for identifying the earliest stages of cerebral amyloid pathology which has implications for future primary prevention trials.
Collapse
Affiliation(s)
- Stefan J Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany.,Department of Psychosomatic Medicine, University Medicine Rostock, Rostock, Germany
| | - Martin Dyrba
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | - Andrea Vergallo
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'Hôpital, Paris, France
| | - Simone Lista
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'Hôpital, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'Hôpital, Paris, France.,Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'Hôpital, Paris, France
| | - Marie Odile Habert
- Laboratoire d'Imagerie Biomédicale, CNRS, INSERM, LIB, Sorbonne University, Paris, France.,Department of Nuclear Medicine, Pitié-Salpêtrière Hospital, AP-HP, Paris, France.,Centre d'Acquisition et Traitement des Images (CATI platform), Paris, France
| | - Marie-Claude Potier
- ICM Institut du Cerveau et de la Moelle Épinière, CNRS UMR 7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, 47 Bd de l'Hôpital, Paris, France
| | - Foudil Lamari
- UF Biochimie des Maladies Neurométaboliques, Service de Biochimie Métabolique, Hôpital Pitié-Salpêtrière, Paris, France
| | - Bruno Dubois
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'Hôpital, Paris, France
| | - Harald Hampel
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'Hôpital, Paris, France
| | - Michel J Grothe
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany.,Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| |
Collapse
|
124
|
Morrow A, Panyard DJ, Deming YK, Jonaitis E, Dong R, Vasiljevic E, Betthauser TJ, Kollmorgen G, Suridjan I, Bayfield A, Van Hulle CA, Zetterberg H, Blennow K, Carlsson CM, Asthana S, Johnson SC, Engelman CD. Cerebrospinal Fluid Sphingomyelins in Alzheimer's Disease, Neurodegeneration, and Neuroinflammation. J Alzheimers Dis 2022; 90:667-680. [PMID: 36155504 PMCID: PMC9809197 DOI: 10.3233/jad-220349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Sphingomyelin (SM) levels have been associated with Alzheimer's disease (AD), but the association direction has been inconsistent and research on cerebrospinal fluid (CSF) SMs has been limited by sample size, breadth of SMs examined, and diversity of biomarkers available. OBJECTIVE Here, we seek to build on our understanding of the role of SM metabolites in AD by studying a broad range of CSF SMs and biomarkers of AD, neurodegeneration, and neuroinflammation. METHODS Leveraging two longitudinal AD cohorts with metabolome-wide CSF metabolomics data (n = 502), we analyzed the relationship between the levels of 12 CSF SMs, and AD diagnosis and biomarkers of pathology, neurodegeneration, and neuroinflammation using logistic, linear, and linear mixed effects models. RESULTS No SMs were significantly associated with AD diagnosis, mild cognitive impairment, or amyloid biomarkers. Phosphorylated tau, neurofilament light, α-synuclein, neurogranin, soluble triggering receptor expressed on myeloid cells 2, and chitinase-3-like-protein 1 were each significantly, positively associated with at least 5 of the SMs. CONCLUSION The associations between SMs and biomarkers of neurodegeneration and neuroinflammation, but not biomarkers of amyloid or diagnosis of AD, point to SMs as potential biomarkers for neurodegeneration and neuroinflammation that may not be AD-specific.
Collapse
Affiliation(s)
- Autumn Morrow
- Department of Population Health Sciences, University of Wisconsin-Madison, 610 Walnut Street, 707 WARF Building, Madison, WI 53726, United States of America
| | - Daniel J. Panyard
- Department of Genetics, School of Medicine, Stanford University, 291 Campus Drive, Stanford, CA 94305, United States of America
| | - Yuetiva K. Deming
- Department of Population Health Sciences, University of Wisconsin-Madison, 610 Walnut Street, 707 WARF Building, Madison, WI 53726, United States of America
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
| | - Erin Jonaitis
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Wisconsin Alzheimer’s Institute, UW School of Medicine and Public Health, 610 Walnut Street, 9th Floor, Madison, WI 53726
| | - Ruocheng Dong
- Department of Population Health Sciences, University of Wisconsin-Madison, 610 Walnut Street, 707 WARF Building, Madison, WI 53726, United States of America
| | - Eva Vasiljevic
- Department of Population Health Sciences, University of Wisconsin-Madison, 610 Walnut Street, 707 WARF Building, Madison, WI 53726, United States of America
- Center for Demography of Health and Aging, University of Wisconsin-Madison, 1180 Observatory Drive, Madison, WI 53706
| | - Tobey J Betthauser
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
| | | | - Ivonne Suridjan
- Roche Diagnostics International Ltd, Forrenstrasse 2, 6343 Rotkreuz, Switzerland
| | - Anna Bayfield
- Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Carol A. Van Hulle
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, 43180 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180 Mölndal, Sweden
- United Kingdom Dementia Research Institute at UCL, London, WC1E6BT, United Kingdom
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1H0AL, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, 43180 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180 Mölndal, Sweden
| | - Cynthia M. Carlsson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
- William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, United States of America
| | - Sanjay Asthana
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
- William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, United States of America
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
- William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, United States of America
| | - Corinne D. Engelman
- Department of Population Health Sciences, University of Wisconsin-Madison, 610 Walnut Street, 707 WARF Building, Madison, WI 53726, United States of America
| |
Collapse
|
125
|
Zhan Y, Fei R, Lu Y, Wan Y, Wu X, Dong J, Meng D, Ge Q, Zhao X. Ultrasensitive detection of multiple Alzheimer's disease biomarkers by SERS-LFA. Analyst 2022; 147:4124-4131. [DOI: 10.1039/d2an00717g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A lateral flow assay, based on Surface-enhanced Raman scattering nanotags (SERS-LFA), is proposed for simultaneous quantification of multiple AD biomarkers with high sensitivity.
Collapse
Affiliation(s)
- Yuanbao Zhan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou 215163, China
- Southeast University Shenzhen Research Institute, Shenzhen 518000, China
| | - Ruihua Fei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou 215163, China
- Southeast University Shenzhen Research Institute, Shenzhen 518000, China
| | - Yu Lu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou 215163, China
- Southeast University Shenzhen Research Institute, Shenzhen 518000, China
| | - Yu Wan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Jiangsu Simcere Pharmaceutical Co., Ltd, Nanjing 210042, China
| | - Xuming Wu
- Nantong Fourth People's Hospital, Jiangsu 226005, China
| | - Jian Dong
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen 518000, China
| | - Dianhuai Meng
- Rehabilitation Medical Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qinyu Ge
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen 518000, China
| | - Xiangwei Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou 215163, China
- Southeast University Shenzhen Research Institute, Shenzhen 518000, China
| |
Collapse
|
126
|
Jiang C, Wang Q, Xie S, Chen Z, Fu L, Peng Q, Liang Y, Guo H, Guo T, for the Alzheimer’s Disease Neuroimaging Initiative. OUP accepted manuscript. Brain Commun 2022; 4:fcac084. [PMID: 35441134 PMCID: PMC9014538 DOI: 10.1093/braincomms/fcac084] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/21/2021] [Accepted: 03/29/2022] [Indexed: 11/14/2022] Open
Affiliation(s)
- Chenyang Jiang
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Qingyong Wang
- Department of Neurology, University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen 518107, China
| | - Siwei Xie
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Zhicheng Chen
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Liping Fu
- Department of Nuclear Medicine, China-Japan Friendship Hospital, 2 Yinghuayuan Dongjie, Beijing 100029, China
| | - Qiyu Peng
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Ying Liang
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Hongbo Guo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Correspondence to: Tengfei Guo, PhD Institute of Biomedical Engineering Shenzhen Bay Laboratory, No.5 Kelian Road Shenzhen 518132, China E-mail:
| | | |
Collapse
|
127
|
Lewczuk P, Wiltfang J, Kornhuber J, Verhasselt A. Distributions of Aβ42 and Aβ42/40 in the Cerebrospinal Fluid in View of the Probability Theory. Diagnostics (Basel) 2021; 11:diagnostics11122372. [PMID: 34943609 PMCID: PMC8700661 DOI: 10.3390/diagnostics11122372] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022] Open
Abstract
Amyloid β 42/40 concentration quotient has been empirically shown to improve accuracy of the neurochemical diagnostics of Alzheimer’s Disease (AD) compared to the Aβ42 concentration alone, but this improvement in diagnostic performance has not been backed up by a theoretical argumentation so far. In this report we show that better accuracy of Aβ42/40 compared to Aβ1-42 is granted by fundamental laws of probability. In particular, it can be shown that the dispersion of a distribution of a quotient of two random variables (Aβ42/40) is smaller than the dispersion of the random variable in the numerator (Aβ42), provided that the two variables are proportional. Further, this concept predicts and explains presence of outlying observations, i.e., AD patients with falsely negatively high Aβ42/40 ratio, and non-AD subjects with extremely low, falsely positive, Aβ42/40 ratio.
Collapse
Affiliation(s)
- Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
- Department of Neurodegeneration Diagnostics, Medical University of Białystok and Department of Biochemical Diagnostics, University Hospital of Białystok, 15-269 Białystok, Poland
- Correspondence:
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center of Göttingen (UMG), 37075 Göttingen, Germany;
- German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany
- Neurosciences and Signaling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Anneleen Verhasselt
- Center for Statistics, Data Science Institute, Hasselt University, 3590 Hasselt, Belgium;
| |
Collapse
|
128
|
Ma LZ, Hu H, Wang ZT, Ou YN, Dong Q, Tan L, Yu JT. P-tau and neurodegeneration mediate the effect of β-amyloid on cognition in non-demented elders. Alzheimers Res Ther 2021; 13:200. [PMID: 34911582 PMCID: PMC8675473 DOI: 10.1186/s13195-021-00943-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/05/2021] [Indexed: 04/12/2023]
Abstract
BACKGROUND There are many pathological changes in the brains of Alzheimer's disease (AD) patients. For many years, the mainstream view on the pathogenesis of AD believes that β-amyloid (Aβ) usually acts independently in addition to triggering functions. However, the evidence now accumulating indicates another case that these pathological types have synergies. The objective of this study was to investigate whether effects of Aβ pathology on cognition were mediated by AD pathologies, including tau-related pathology (p-tau), neurodegeneration (t-tau, MRI measurements), axonal injury (NFL), synaptic dysfunction (neurogranin), and neuroinflammation (sTREM2, YKL-40). METHODS Three hundred seventy normal controls (CN) and 623 MCI patients from the ADNI (Alzheimer's Disease Neuroimaging Initiative) database were recruited in this research. Linear mixed-effects models were used to evaluate the associations of baseline Aβ with cognitive decline and biomarkers of several pathophysiological pathways. Causal mediation analyses with 10,000 bootstrapped iterations were conducted to explore the mediation effects of AD pathologies on cognition. RESULTS Tau-related pathology, neurodegeneration, neuroinflammation are correlated with the concentration of Aβ, even in CN participants. The results show that age, gender, and APOE ε4 carrier status have a moderating influence on some of these relationships. There is a stronger association of Aβ with biomarkers and cognitive changes in the elderly and females. In CN group, Aβ pathology is directly related to poor cognition and has no mediating effect (p < 0.05). In mild cognitive impairment, tau-related pathology (26.15% of total effect) and neurodegeneration (14.8% to 47.0% of total effect) mediate the impact of Aβ on cognition. CONCLUSIONS In conclusion, early Aβ accumulation has an independent effect on cognitive decline in CN and a tau, neurodegeneration-dependent effect in the subsequent cognitive decline in MCI patients.
Collapse
Affiliation(s)
- Ling-Zhi Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China.
| |
Collapse
|
129
|
Sacchi L, Carandini T, Fumagalli GG, Pietroboni AM, Contarino VE, Siggillino S, Arcaro M, Fenoglio C, Zito F, Marotta G, Castellani M, Triulzi F, Galimberti D, Scarpini E, Arighi A. Unravelling the Association Between Amyloid-PET and Cerebrospinal Fluid Biomarkers in the Alzheimer's Disease Spectrum: Who Really Deserves an A+? J Alzheimers Dis 2021; 85:1009-1020. [PMID: 34897084 DOI: 10.3233/jad-210593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Association between cerebrospinal fluid (CSF)-amyloid-β (Aβ)42 and amyloid-PET measures is inconstant across the Alzheimer's disease (AD) spectrum. However, they are considered interchangeable, along with Aβ 42/40 ratio, for defining 'Alzheimer's Disease pathologic change' (A+). OBJECTIVE Herein, we further characterized the association between amyloid-PET and CSF biomarkers and tested their agreement in a cohort of AD spectrum patients. METHODS We include ed 23 patients who underwent amyloid-PET, MRI, and CSF analysis showing reduced levels of Aβ 42 within a 365-days interval. Thresholds used for dichotomization were: Aβ 42 < 640 pg/mL (Aβ 42+); pTau > 61 pg/mL (pTau+); and Aβ 42/40 < 0.069 (ADratio+). Amyloid-PET scans were visually assessed and processed by four pipelines (SPMCL, SPMAAL, FSGM, FSWC). RESULTS Different pipelines gave highly inter-correlated standardized uptake value ratios (SUVRs) (rho = 0.93-0.99). The most significant findings were: pTau positive correlation with SPMCL SUVR (rho = 0.56, p = 0.0063) and Aβ 42/40 negative correlation with SPMCL and SPMAAL SUVRs (rho = -0.56, p = 0.0058; rho = -0.52, p = 0.0117 respectively). No correlations between CSF-Aβ 42 and global SUVRs were observed. In subregion analysis, both pTau and Aβ 42/40 values significantly correlated with cingulate SUVRs from any pipeline (R2 = 0.55-0.59, p < 0.0083), with the strongest associations observed for the posterior/isthmus cingulate areas. However, only associations observed for Aβ 42/40 ratio were still significant in linear regression models. Moreover, combining pTau with Aβ 42 or using Aβ 42/40, instead of Aβ 42 alone, increased concordance with amyloid-PET status from 74% to 91% based on visual reads and from 78% to 96% based on Centiloids. CONCLUSION We confirmed that, in the AD spectrum, amyloid-PET measures show a stronger association and a better agreement with CSF-Aβ 42/40 and secondarily pTau rather than Aβ 42 levels.
Collapse
Affiliation(s)
- Luca Sacchi
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Tiziana Carandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Anna Margherita Pietroboni
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Silvia Siggillino
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marina Arcaro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Felicia Zito
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio Marotta
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Castellani
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio Triulzi
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Galimberti
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elio Scarpini
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Arighi
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
130
|
Han HF, Yen HC, Wu HC, Tan HY, Xu W, Jiang HS, Tsai PJ, Qian K, Wu YC, Chen CC. Ultrasensitive Detection of Alzheimer's Amyloids on a Plasmonic-Gold Platform. ACS APPLIED MATERIALS & INTERFACES 2021; 13:57036-57042. [PMID: 34843217 DOI: 10.1021/acsami.1c19157] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
More than 55 million people live with dementia worldwide in 2021, and there are nearly 10 million new cases every year. Alzheimer's disease (AD) is the most common cause of dementia. Despite urgent need, early detection of AD and long-term monitoring of AD progression have been challenging. This is due to the limited availability of brain imaging facilities and the highly invasive procedure with the cerebrospinal fluid assay to assess the level of AD biomarkers, such as beta-amyloid (Aβ). Reliable measurements of AD biomarkers in blood samples are still difficult because of their very low abundance. Here, we develop a rapid, specific, and ultrasensitive immunoassay using plasmonic-gold nanoisland (pGOLD) chips with near-infrared fluorescence-enhanced detection for Aβ1-40 and Aβ1-42. We show step-by-step processes and results during the platform establishment, including antibody specificity and sensitivity tests, antibody pair examination, condition optimization, and procedure refinement. Finally, we demonstrate the platform performance with detection sensitivity at the subpicogram per milliliter level. This platform, therefore, has a great application potential for early detection of AD using blood samples.
Collapse
Affiliation(s)
- Hsiao-Fen Han
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 106, Taiwan
| | - Hung-Chi Yen
- Department of Chemistry, National Taiwan Normal University, Taipei 116, Taiwan
| | - Hsiu-Chuan Wu
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Branch, and College of Medicine, Chang Gung University, Taoyuan City 333, Taiwan
| | - Hsin-Yuan Tan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou Branch, and College of Medicine, Chang Gung University, Taoyuan City 333, Taiwan
| | - Wei Xu
- School of Biomedical Engineering, Shanghai Chest Hospital and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hang-Shiang Jiang
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 106, Taiwan
| | - Ping-Jui Tsai
- Department of Orthopedics, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan City 333, Taiwan
| | - Kun Qian
- School of Biomedical Engineering, Shanghai Chest Hospital and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yi-Chun Wu
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 106, Taiwan
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Chia-Chun Chen
- Department of Chemistry, National Taiwan Normal University, Taipei 116, Taiwan
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| |
Collapse
|
131
|
Kuroda T, Honma M, Mori Y, Futamura A, Sugimoto A, Kasai H, Yano S, Hieda S, Kasuga K, Ikeuchi T, Ono K. White Matter Lesions May Aid in Differentiating Idiopathic Normal Pressure Hydrocephalus and Alzheimer's Disease. J Alzheimers Dis 2021; 85:851-862. [PMID: 34864676 DOI: 10.3233/jad-215187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Idiopathic normal pressure hydrocephalus (iNPH) is often misdiagnosed as Alzheimer's disease (AD) due to overlapping pathophysiology and similar imaging characteristics, including ventricular enlargement and increased white matter lesions (WMLs). OBJECTIVE To compare the extent and distribution of WMLs directly between iNPH and AD and examine the association with underlying pathophysiology. METHODS Twelve patients with iNPH (mean age: 78.08 years; 5 females), 20 with AD (mean age: 75.40 years; 13 females), and 10 normal cognition (NC) participants (mean age: 76.60 years; 7 females) were recruited. The extent and distribution of WMLs and the lateral ventricular volume (LV-V) were evaluated on MRI using voxel-based morphometry analysis. Concentrations of cerebrospinal fluid biomarkers, such as amyloid-β protein (Aβ)42, Aβ 40, Aβ 38, and tau species, were also measured. Risk factors for small vessel disease (SVD) were assessed by blood examination and medical records. RESULTS The periventricular WML volume (PWML-V) and deep WML volume (DWML-V) were significantly larger in iNPH than in AD and NC. The DWML-V was dominant in iNPH, while the PWML-V was dominant in AD and NC. GM-V was significantly smaller in AD than in iNPH and NC. The LV-V positively correlated with WML-V in all participants. There was a significant negative correlation between LV-V and Aβ 38 in iNPH. Furthermore, there was no significant difference in SVD risk factors between the groups. CONCLUSION The differences in the extent and distribution of WMLs between iNPH and AD, especially predominance of DWML-V over PWML-V in iNPH, may reflect decreased fluid and Aβ clearance.
Collapse
Affiliation(s)
- Takeshi Kuroda
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Motoyasu Honma
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| | - Yukiko Mori
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Akinori Futamura
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Azusa Sugimoto
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hideyo Kasai
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Satoshi Yano
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Sotaro Hieda
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Chuo-ku, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Chuo-ku, Niigata, Japan
| | - Kenjiro Ono
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.,Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
132
|
Gobom J, Parnetti L, Rosa-Neto P, Vyhnalek M, Gauthier S, Cataldi S, Lerch O, Laczo J, Cechova K, Clarin M, Benet AI, Pascoal TA, Rahmouni N, Vandijck M, Huyck E, Le Bastard N, Stevenson J, Chamoun M, Alcolea D, Lleó A, Andreasson U, Verbeek MM, Bellomo G, Rinaldi R, Ashton N, Zetterberg H, Sheardova K, Hort J, Blennow K. Validation of the LUMIPULSE automated immunoassay for the measurement of core AD biomarkers in cerebrospinal fluid. Clin Chem Lab Med 2021; 60:207-219. [PMID: 34773730 DOI: 10.1515/cclm-2021-0651] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 11/02/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVES The core cerebrospinal fluid (CSF) biomarkers; total tau (tTau), phospho-tau (pTau), amyloid β 1-42 (Aβ 1-42), and the Aβ 1-42/Aβ 1-40 ratio have transformed Alzheimer's disease (AD) research and are today increasingly used in clinical routine laboratories as diagnostic tools. Fully automated immunoassay instruments with ready-to-use assay kits and calibrators has simplified their analysis and improved reproducibility of measurements. We evaluated the analytical performance of the fully automated immunoassay instrument LUMIPULSE G (Fujirebio) for measurement of the four core AD CSF biomarkers and determined cutpoints for AD diagnosis. METHODS Comparison of the LUMIPULSE G assays was performed with the established INNOTEST ELISAs (Fujirebio) for hTau Ag, pTau 181, β-amyloid 1-42, and with V-PLEX Plus Aβ Peptide Panel 1 (6E10) (Meso Scale Discovery) for Aβ 1-42/Aβ 1-40, as well as with a LC-MS reference method for Aβ 1-42. Intra- and inter-laboratory reproducibility was evaluated for all assays. Clinical cutpoints for Aβ 1-42, tTau, and pTau was determined by analysis of three cohorts of clinically diagnosed patients, comprising 651 CSF samples. For the Aβ 1-42/Aβ 1-40 ratio, the cutpoint was determined by mixture model analysis of 2,782 CSF samples. RESULTS The LUMIPULSE G assays showed strong correlation to all other immunoassays (r>0.93 for all assays). The repeatability (intra-laboratory) CVs ranged between 2.0 and 5.6%, with the highest variation observed for β-amyloid 1-40. The reproducibility (inter-laboratory) CVs ranged between 2.1 and 6.5%, with the highest variation observed for β-amyloid 1-42. The clinical cutpoints for AD were determined to be 409 ng/L for total tau, 50.2 ng/L for pTau 181, 526 ng/L for β-amyloid 1-42, and 0.072 for the Aβ 1-42/Aβ 1-40 ratio. CONCLUSIONS Our results suggest that the LUMIPULSE G assays for the CSF AD biomarkers are fit for purpose in clinical laboratory practice. Further, they corroborate earlier presented reference limits for the biomarkers.
Collapse
Affiliation(s)
- Johan Gobom
- 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
| | - Lucilla Parnetti
- Laboratory of Clinical Neurochemistry, Section of Neurology, University of Perugia, Perugia, Italy
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Montreal Neurological Institute, Montreal, QC, Canada
| | - Martin Vyhnalek
- Department of Neurology, Second Medical Faculty, Charles University, Prague, Czech Republic.,Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Serge Gauthier
- Department of Neurology and Neurosurgery, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Montreal Neurological Institute, Montreal, QC, Canada
| | - Samuela Cataldi
- Laboratory of Clinical Neurochemistry, Section of Neurology, University of Perugia, Perugia, Italy
| | - Ondrej Lerch
- Department of Neurology, Second Medical Faculty, Charles University, Prague, Czech Republic.,Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Jan Laczo
- Department of Neurology, Second Medical Faculty, Charles University, Prague, Czech Republic.,Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Katerina Cechova
- Department of Neurology, Second Medical Faculty, Charles University, Prague, Czech Republic.,Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Marcus Clarin
- 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
| | - Andrea I Benet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Neserine Rahmouni
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | | | | | | | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Daniel Alcolea
- Department of Neurology, Memory Unit, Hospital de la Santa Creu i Sant Pau- Biomedical Research Institute Sant Pau-Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alberto Lleó
- Department of Neurology, Memory Unit, Hospital de la Santa Creu i Sant Pau- Biomedical Research Institute Sant Pau-Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ulf Andreasson
- 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
| | - Marcel M Verbeek
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Neurology, Radboud Alzheimer Centre, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Giovanni Bellomo
- Laboratory of Clinical Neurochemistry, Section of Neurology, University of Perugia, Perugia, Italy
| | - Roberta Rinaldi
- Laboratory of Clinical Neurochemistry, Section of Neurology, University of Perugia, Perugia, Italy
| | - Nicholas Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - 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, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Katerina Sheardova
- Department of Neurology, Second Medical Faculty, Charles University, Prague, Czech Republic.,First Department of Neurology, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic
| | - Jakub Hort
- Department of Neurology, Second Medical Faculty, Charles University, Prague, Czech Republic.,Motol University Hospital, Prague, Czech Republic.,First Department of Neurology, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic
| | - 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
| |
Collapse
|
133
|
Extra Virgin Olive Oil consumption from Mild Cognitive Impairment patients attenuates oxidative and nitrative stress reflecting on the reduction of the PARP levels and DNA damage. Exp Gerontol 2021; 156:111621. [PMID: 34748951 DOI: 10.1016/j.exger.2021.111621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/24/2021] [Accepted: 11/01/2021] [Indexed: 11/23/2022]
Abstract
Oxidative/nitrative stress that results from the unbalance of the overproduction/clearance of reactive oxygen/nitrogen species (ROS/NOS), originated from a variety of endo- and/or exo-genous sources, can have detrimental effects on DNA and is involved in Alzheimer's disease (AD) pathology. An excellent marker of oxidative DNA lesions is 8-hydroxy-2'-deoxyguanosine (8-OHdG) while of nitrative stress the enzyme NOS2 (Nitric oxide synthase 2). Under massive oxidative stress, poly(ADP-ribose)polymerase 1 (PARP-1) enzyme activity, responsible for restoration of DNA damage, is augmented, DNA repair enzymes are recruited, and cell survival/or death is ensued through PARP-1 activation, which is correlated positively with neurodegenerative diseases. In this biochemical study the levels of PARP-1, 8-oxo-dG, and NOS2, Aβ1-42, and p-tau in their sera determined using Enzyme-Linked Immunosorbent Assay (ELISA). Patients diagnosed with Mild Cognitive Impairment participated in MICOIL clinical trial, were daily administered with 50 ml Extra Virgin Olive Oil (EVOO) for one year. All MCI patients' biomarkers that had consumed EVOO were tantamount to those of healthy participants, contrary to MCI patients who were not administered. EVOO administration in MCI patients resulted in the restoration of DNA damage and of the well-established "hallmarks" AD biomarkers, thanks probably to its antioxidant properties exhibiting a therapeutic potentiality against AD. Molecular docking simulations of the EVOO constituents on the crystal structure of PARP-1 and NOS-2 target enzymes were also employed, to study in silico the ability of the compounds to bind to these enzymes and explain the observed in vitro activity. In silico analysis has proved the binding of EVOO constituents on PARP-1and NOS-2 enzymes and their interaction with crucial amino acids of the active sites. CLINICAL TRIAL REGISTRATION: https://clinicaltrials.gov/ct2/show/NCT03362996. MICOIL GOV IDENTIFIER: NCT03362996.
Collapse
|
134
|
Nerattini M, Rubino F, Arnone A, Polito C, Mazzeo S, Lombardi G, Puccini G, Nacmias B, De Cristofaro MT, Sorbi S, Pupi A, Sciagrà R, Bessi V, Berti V. Cerebral amyloid load determination in a clinical setting: interpretation of amyloid biomarker discordances aided by tau and neurodegeneration measurements. Neurol Sci 2021; 43:2469-2480. [PMID: 34739618 DOI: 10.1007/s10072-021-05704-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/26/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) diagnosis can be hindered by amyloid biomarkers discordances. OBJECTIVE We aim to interpret discordances between amyloid positron emission tomography (Amy-PET) and cerebrospinal fluid (CSF) (Aβ42 and Aβ42/40), using Amy-PET semiquantitative analysis, [18F]fluorodeoxyglucose (FDG)-PET pattern, and CSF assays. METHOD Thirty-six subjects with dementia or mild cognitive impairment, assessed by neuropsychological tests, structural and functional imaging, and CSF assays (Aβ42, Aβ42/40, p-tau, t-tau), were retrospectively examined. Amy-PET and FDG-PET scans were analyzed by visual assessment and voxel-based analysis. SUVR were calculated on Amy-PET scans. RESULTS Groups were defined basing on the agreement among CSF Aβ42 (A), CSF Aβ42/40 Ratio (R), and Amy-PET (P) dichotomic results ( ±). In discordant groups, CSF assays, Amy-PET semiquantification, and FDG-PET patterns supported the diagnosis suggested by any two agreeing amyloid biomarkers. In groups with discordant CSF Aβ42, the ratio always agrees with Amy-PET results, solving both false-negative and false-positive Aβ42 results, with Aβ42 levels close to the cut-off in A + R-P- subjects. The A + R + P- group presented high amyloid deposition in relevant areas, such as precuneus, posterior cingulate cortex (PCC) and dorsolateral frontal inferior cortex at semiquantitative analysis. CONCLUSION The amyloid discordant cases could be overcome by combining CSF Aβ42, CSF ratio, and Amy-PET results. The concordance of any 2 out of the 3 biomarkers seems to reveal the remaining one as a false result. A cut-off point review could avoid CSF Aβ42 false-negative results. The regional semiquantitative Amy-PET analysis in AD areas, such as precuneus and PCC, could increase the accuracy in AD diagnosis.
Collapse
Affiliation(s)
- Matilde Nerattini
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy.
| | - Federica Rubino
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Annachiara Arnone
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Cristina Polito
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - Salvatore Mazzeo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - Gemma Lombardi
- IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143, Florence, Italy
| | - Giulia Puccini
- Department of Nuclear Medicine, Hospital of Prato, Via Suor Niccolina Infermiera, 20/22, 59100, Prato, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143, Florence, Italy
| | - Maria Teresa De Cristofaro
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143, Florence, Italy
| | - Alberto Pupi
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Roberto Sciagrà
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - Valentina Berti
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| |
Collapse
|
135
|
Chong JR, Ashton NJ, Karikari TK, Tanaka T, Schöll M, Zetterberg H, Blennow K, Chen CP, Lai MKP. Blood-based high sensitivity measurements of beta-amyloid and phosphorylated tau as biomarkers of Alzheimer's disease: a focused review on recent advances. J Neurol Neurosurg Psychiatry 2021; 92:1231-1241. [PMID: 34510001 DOI: 10.1136/jnnp-2021-327370] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/30/2021] [Indexed: 01/11/2023]
Abstract
Discovery and development of clinically useful biomarkers for Alzheimer's disease (AD) and related dementias have been the focus of recent research efforts. While cerebrospinal fluid and positron emission tomography or MRI-based neuroimaging markers have made the in vivo detection of AD pathology and its consequences possible, the high cost and invasiveness have limited their widespread use in the clinical setting. On the other hand, advances in potentially more accessible blood-based biomarkers had been impeded by lack of sensitivity in detecting changes in markers of the hallmarks of AD, including amyloid-β (Aβ) peptides and phosphorylated tau (P-tau). More recently, however, emerging technologies with superior sensitivity and specificity for measuring Aβ and P-tau have reported high concordances with AD severity. In this focused review, we describe several emerging technologies, including immunoprecipitation-mass spectrometry (IP-MS), single molecule array and Meso Scale Discovery immunoassay platforms, and appraise the current literature arising from their use to identify plaques, tangles and other AD-associated pathology. While there is potential clinical utility in adopting these technologies, we also highlight the further studies needed to establish Aβ and P-tau as blood-based biomarkers for AD, including validation with existing large sample sets, new independent cohorts from diverse backgrounds as well as population-based longitudinal studies. In conclusion, the availability of sensitive and reliable measurements of Aβ peptides and P-tau species in blood holds promise for the diagnosis, prognosis and outcome assessments in clinical trials for AD.
Collapse
Affiliation(s)
- Joyce R Chong
- Memory, Aging and Cognition Centre, National University Health Systems, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nicholas J Ashton
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Psychology and Neuroscience, King's College London, Institute of Psychiatry, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK.,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia, South London and Maudsley NHS Foundation, London, UK.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tomotaka Tanaka
- Memory, Aging and Cognition Centre, National University Health Systems, Singapore.,Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden.,UK Dementia Research Institute at UCL, University College London, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,UK Dementia Research Institute at UCL, University College London, London, UK
| | - Christopher P Chen
- Memory, Aging and Cognition Centre, National University Health Systems, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mitchell K P Lai
- Memory, Aging and Cognition Centre, National University Health Systems, Singapore .,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
136
|
Genetic effects on longitudinal cognitive decline during the early stages of Alzheimer's disease. Sci Rep 2021; 11:19853. [PMID: 34615922 PMCID: PMC8494841 DOI: 10.1038/s41598-021-99310-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 09/22/2021] [Indexed: 11/08/2022] Open
Abstract
Cognitive decline in early-stage Alzheimer's disease (AD) may depend on genetic variability. In the Swedish BioFINDER study, we used polygenic scores (PGS) (for AD, intelligence, and educational attainment) to predict longitudinal cognitive change (measured by mini-mental state examination (MMSE) [primary outcome] and other cognitive tests) over a mean of 4.2 years. We included 260 β-amyloid (Aβ) negative cognitively unimpaired (CU) individuals, 121 Aβ-positive CU (preclinical AD), 50 Aβ-negative mild cognitive impairment (MCI) patients, and 127 Aβ-positive MCI patients (prodromal AD). Statistical significance was determined at Bonferroni corrected p value < 0.05. The PGS for intelligence (beta = 0.1, p = 2.9e-02) was protective against decline in MMSE in CU and MCI participants regardless of Aβ status. The polygenic risk score for AD (beta = - 0.12, p = 9.4e-03) was correlated with the rate of change in MMSE and was partially mediated by Aβ-pathology (mediation effect 20%). There was no effect of education PGS on cognitive measures. Genetic variants associated with intelligence mitigate cognitive decline independent of Aβ-pathology, while effects of genetic variants associated with AD are partly mediated by Aβ-pathology.
Collapse
|
137
|
Soliman HM, Ghonaim GA, Gharib SM, Chopra H, Farag AK, Hassanin MH, Nagah A, Emad-Eldin M, Hashem NE, Yahya G, Emam SE, Hassan AEA, Attia MS. Exosomes in Alzheimer's Disease: From Being Pathological Players to Potential Diagnostics and Therapeutics. Int J Mol Sci 2021; 22:10794. [PMID: 34639135 PMCID: PMC8509246 DOI: 10.3390/ijms221910794] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 12/15/2022] Open
Abstract
Exosomes (EXOs) were given attention as an extracellular vesicle (EV) with a pivotal pathophysiological role in the development of certain neurodegenerative disorders (NDD), such as Parkinson's and Alzheimer's disease (AD). EXOs have shown the potential to carry pathological and therapeutic cargo; thus, researchers have harnessed EXOs in drug delivery applications. EXOs have shown low immunogenicity as natural drug delivery vehicles, thus ensuring efficient drug delivery without causing significant adverse reactions. Recently, EXOs provided potential drug delivery opportunities in AD and promising future clinical applications with the diagnosis of NDD and were studied for their usefulness in disease detection and prediction prior to the emergence of symptoms. In the future, the microfluidics technique will play an essential role in isolating and detecting EXOs to diagnose AD before the development of advanced symptoms. This review is not reiterative literature but will discuss why EXOs have strong potential in treating AD and how they can be used as a tool to predict and diagnose this disorder.
Collapse
Affiliation(s)
- Hagar M. Soliman
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Ghada A. Ghonaim
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Shaza M. Gharib
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Aya K. Farag
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Mohamed H. Hassanin
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Abdalrazeq Nagah
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Mahmoud Emad-Eldin
- Department of Clinical, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Nevertary E. Hashem
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Sherif E. Emam
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Abdalla E. A. Hassan
- Applied Nucleic Acids Research Center & Chemistry, Faculty of Science, Zagazig 44519, Egypt;
| | - Mohamed S. Attia
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| |
Collapse
|
138
|
Korecka M, Shaw LM. Mass spectrometry-based methods for robust measurement of Alzheimer's disease biomarkers in biological fluids. J Neurochem 2021; 159:211-233. [PMID: 34244999 PMCID: PMC9057379 DOI: 10.1111/jnc.15465] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/11/2021] [Accepted: 07/06/2021] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia affecting 60%-70% of people afflicted with this disease. Accurate antemortem diagnosis is urgently needed for early detection of AD to enable reliable estimation of prognosis, intervention, and monitoring of the disease. The National Institute on Aging/Alzheimer's Association sponsored the 'Research Framework: towards a biological definition of AD', which recommends using different biomarkers in living persons for a biomarker-based definition of AD regardless of clinical status. Fluid biomarkers represent one of key groups of them. Since cerebrospinal fluid (CSF) is in direct contact with brain and many proteins present in the brain can be detected in CSF, this fluid has been regarded as the best biofluid in which to measure AD biomarkers. Recently, technological advancements in protein detection made possible the effective study of plasma AD biomarkers despite their significantly lower concentrations versus to that in CSF. This and other challenges that face plasma-based biomarker measurements can be overcome by using mass spectrometry. In this review, we discuss AD biomarkers which can be reliably measured in CSF and plasma using targeted mass spectrometry coupled to liquid chromatography (LC/MS/MS). We describe progress in LC/MS/MS methods' development, emphasize the challenges, and summarize major findings. We also highlight the role of mass spectrometry and progress made in the process of global standardization of the measurement of Aβ42/Aβ40. Finally, we briefly describe exploratory proteomics which seek to identify new biomarkers that can contribute to detection of co-pathological processes that are common in sporadic AD.
Collapse
Affiliation(s)
- Magdalena Korecka
- Department of Pathology and Laboratory Medicine Perlman School of Medicine University of Pennsylvania Philadelphia PA USA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine Perlman School of Medicine University of Pennsylvania Philadelphia PA USA
| |
Collapse
|
139
|
Roos TT, Garcia MG, Martinsson I, Mabrouk R, Israelsson B, Deierborg T, Kobro-Flatmoen A, Tanila H, Gouras GK. Neuronal spreading and plaque induction of intracellular Aβ and its disruption of Aβ homeostasis. Acta Neuropathol 2021; 142:669-687. [PMID: 34272583 PMCID: PMC8423700 DOI: 10.1007/s00401-021-02345-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/18/2021] [Accepted: 07/07/2021] [Indexed: 01/11/2023]
Abstract
The amyloid-beta peptide (Aβ) is thought to have prion-like properties promoting its spread throughout the brain in Alzheimer’s disease (AD). However, the cellular mechanism(s) of this spread remains unclear. Here, we show an important role of intracellular Aβ in its prion-like spread. We demonstrate that an intracellular source of Aβ can induce amyloid plaques in vivo via hippocampal injection. We show that hippocampal injection of mouse AD brain homogenate not only induces plaques, but also damages interneurons and affects intracellular Aβ levels in synaptically connected brain areas, paralleling cellular changes seen in AD. Furthermore, in a primary neuron AD model, exposure of picomolar amounts of brain-derived Aβ leads to an apparent redistribution of Aβ from soma to processes and dystrophic neurites. We also observe that such neuritic dystrophies associate with plaque formation in AD-transgenic mice. Finally, using cellular models, we propose a mechanism for how intracellular accumulation of Aβ disturbs homeostatic control of Aβ levels and can contribute to the up to 10,000-fold increase of Aβ in the AD brain. Our data indicate an essential role for intracellular prion-like Aβ and its synaptic spread in the pathogenesis of AD.
Collapse
Affiliation(s)
- Tomas T Roos
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Megg G Garcia
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Isak Martinsson
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Rana Mabrouk
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Bodil Israelsson
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Heikki Tanila
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Gunnar K Gouras
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
140
|
The Neurobiological Basis of Cognitive Side Effects of Electroconvulsive Therapy: A Systematic Review. Brain Sci 2021; 11:brainsci11101273. [PMID: 34679338 PMCID: PMC8534116 DOI: 10.3390/brainsci11101273] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 11/26/2022] Open
Abstract
Decades of research have consistently demonstrated the efficacy of electroconvulsive therapy (ECT) for the treatment of major depressive disorder (MDD), but its clinical use remains somewhat restricted because of its cognitive side effects. The aim of this systematic review is to comprehensively summarize current evidence assessing potential biomarkers of ECT-related cognitive side effects. Based on our systematic search of human studies indexed in PubMed, Scopus, and Web of Knowledge, a total of 29 studies evaluating patients with MDD undergoing ECT were reviewed. Molecular biomarkers studies did not consistently identify concentration changes in plasma S-100 protein, neuron-specific enolase (NSE), or Aβ peptides significantly associated with cognitive performance after ECT. Importantly, these findings suggest that ECT-related cognitive side effects cannot be explained by mechanisms of neural cell damage. Notwithstanding, S-100b protein and Aβ40 peptide concentrations, as well as brain-derived neurotrophic factor (BDNF) polymorphisms, have been suggested as potential predictive biomarkers of cognitive dysfunction after ECT. In addition, recent advances in brain imaging have allowed us to identify ECT-induced volumetric and functional changes in several brain structures closely related to memory performance such as the hippocampus. We provide a preliminary framework to further evaluate neurobiological cognitive vulnerability profiles of patients with MDD treated with ECT.
Collapse
|
141
|
Wang YL, Chen J, Du ZL, Weng H, Zhang Y, Li R, Jia Z, Sun M, Jiang J, Wang FZ, Xu J. Plasma p-tau181 Level Predicts Neurodegeneration and Progression to Alzheimer's Dementia: A Longitudinal Study. Front Neurol 2021; 12:695696. [PMID: 34557143 PMCID: PMC8452983 DOI: 10.3389/fneur.2021.695696] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/09/2021] [Indexed: 11/30/2022] Open
Abstract
Background: Plasma-based biomarkers would be potential biomarkers for early diagnosis of Alzheimer's disease (AD) because they are more available and cost-effective than cerebrospinal fluid (CSF) or neuroimaging. Therefore, we aimed to evaluate whether phosphorylated tau181 (p-tau181) in plasma could be an accurate AD predictor. Methods: Participants from the ADNI database included 185 cognitively unimpaired subjects with negative Aβ (CU–), 66 subjects with pre-clinical AD (CU with positive Aβ), 164 subjects with mild cognitive impairment with negative Aβ (MCI–), 254 subjects with prodromal AD (MCI with positive Aβ), and 98 subjects with dementia. Multiple linear regression models, linear mixed-effects models, and local regression were used to explore cross-sectional and longitudinal associations of plasma p-tau181 with cognition, neuroimaging, or CSF biomarkers adjusted for age, sex, education, and APOE genotype. Besides, Kaplan–Meier and adjusted Cox-regression model were performed to predict the risk of progression to dementia. Receiver operating characteristic analyses were performed to evaluate the predictive value of p-tau181. Results: Plasma p-tau181 level was highest in AD dementia, followed by prodromal AD and pre-clinical AD. In pre-clinical AD, plasma p-tau181 was negatively associated with hippocampal volume (β = −0.031, p-value = 0.017). In prodromal AD, plasma p-tau181 was associated with decreased global cognition, executive function, memory, language, and visuospatial functioning (β range −0.119 to −0.273, p-value < 0.05) and correlated with hippocampal volume (β = −0.028, p-value < 0.005) and white matter hyperintensity volume (WMH) volume (β = 0.02, p-value = 0.01). In AD dementia, increased plasma p-tau181 was associated with worse memory. In the whole group, baseline plasma p-tau181 was significantly associated with longitudinal increases in multiple neuropsychological test z-scores and correlated with AD-related CSF biomarkers and hippocampal volume (p-value < 0.05). Meanwhile, CU or MCI with high plasma p-tau181 carried a higher risk of progression to dementia. The area under the curve (AUC) of the adjusted model (age, sex, education, APOE genotype, and plasma p-tau181) was 0.78; that of additionally included CSF biomarkers was 0.84. Conclusions: Plasma p-tau181 level is related to multiple AD-associated cognitive domains and AD-related CSF biomarkers at the clinical stages of AD. Moreover, plasma p-tau181 level is related to the change rates of cognitive decline and hippocampal atrophy. Thus, this study confirms the utility of plasma p-tau181 as a non-invasive biomarker for early detection and prediction of AD.
Collapse
Affiliation(s)
- Yan-Li Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinglong Chen
- Department of Geriatric Medicine, China National Clinical Key Specialty, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhong-Li Du
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China.,Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Haoyi Weng
- Shenzhen WeGene Clinical Laboratory, Shenzhen, China.,WeGene, Shenzhen Zaozhidao Technology Co. Ltd., Shenzhen, China.,Hunan Provincial Key Lab on Bioinformatics, School of Science and Engineering, Central South University, Shenzhen, China
| | - Yuan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Runzhi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ziyan Jia
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mengfan Sun
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiwei Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fang-Ze Wang
- Department of Cardiology, Weifang People's Hospital, Weifang, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | | |
Collapse
|
142
|
Insulin and Insulin Resistance in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22189987. [PMID: 34576151 PMCID: PMC8472298 DOI: 10.3390/ijms22189987] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023] Open
Abstract
Insulin plays a range of roles as an anabolic hormone in peripheral tissues. It regulates glucose metabolism, stimulates glucose transport into cells and suppresses hepatic glucose production. Insulin influences cell growth, differentiation and protein synthesis, and inhibits catabolic processes such as glycolysis, lipolysis and proteolysis. Insulin and insulin-like growth factor-1 receptors are expressed on all cell types in the central nervous system. Widespread distribution in the brain confirms that insulin signaling plays important and diverse roles in this organ. Insulin is known to regulate glucose metabolism, support cognition, enhance the outgrowth of neurons, modulate the release and uptake of catecholamine, and regulate the expression and localization of gamma-aminobutyric acid (GABA). Insulin is also able to freely cross the blood–brain barrier from the circulation. In addition, changes in insulin signaling, caused inter alia insulin resistance, may accelerate brain aging, and affect plasticity and possibly neurodegeneration. There are two significant insulin signal transduction pathways: the PBK/AKT pathway which is responsible for metabolic effects, and the MAPK pathway which influences cell growth, survival and gene expression. The aim of this study is to describe the role played by insulin in the CNS, in both healthy people and those with pathologies such as insulin resistance and Alzheimer’s disease.
Collapse
|
143
|
Erhardt EB, Adair JC, Knoefel JE, Caprihan A, Prestopnik J, Thompson J, Hobson S, Siegel D, Rosenberg GA. Inflammatory Biomarkers Aid in Diagnosis of Dementia. Front Aging Neurosci 2021; 13:717344. [PMID: 34489684 PMCID: PMC8416621 DOI: 10.3389/fnagi.2021.717344] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/22/2021] [Indexed: 11/17/2022] Open
Abstract
Dual pathology of Alzheimer's disease (AD) and vascular cognitive impairment and dementia (VCID) commonly are found together at autopsy, but mixed dementia (MX) is difficult to diagnose during life. Biological criteria to diagnose AD have been defined, but are not available for vascular disease. We used the biological criteria for AD and white matter injury based on MRI to diagnose MX. Then we measured multiple biomarkers in CSF and blood with multiplex biomarker kits for proteases, angiogenic factors, and cytokines to explore pathophysiology in each group. Finally, we used machine learning with the Random forest algorithm to select the biomarkers of maximal importance; that analysis identified three proteases, matrix metalloproteinase-10 (MMP-10), MMP-3 and MMP-1; three angiogenic factors, VEGF-C, Tie-2 and PLGF, and three cytokines interleukin-2 (IL-2), IL-6, IL-13. To confirm the clinical importance of the variables, we showed that they correlated with results of neuropsychological testing.
Collapse
Affiliation(s)
- Erik B Erhardt
- Department of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, United States
| | - John C Adair
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States.,Center for Memory and Aging, Albuquerque, NM, United States
| | - Janice E Knoefel
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States.,Center for Memory and Aging, Albuquerque, NM, United States
| | | | | | | | - Sasha Hobson
- Center for Memory and Aging, Albuquerque, NM, United States
| | - David Siegel
- Department of Anesthesiology, University of New Mexico, Albuquerque, NM, United States
| | - Gary A Rosenberg
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States.,Center for Memory and Aging, Albuquerque, NM, United States
| |
Collapse
|
144
|
Lombardi G, Pupi A, Bessi V, Polito C, Padiglioni S, Ferrari C, Lucidi G, Berti V, De Cristofaro MT, Piaceri I, Bagnoli S, Nacmias B, Sorbi S. Challenges in Alzheimer's Disease Diagnostic Work-Up: Amyloid Biomarker Incongruences. J Alzheimers Dis 2021; 77:203-217. [PMID: 32716357 DOI: 10.3233/jad-200119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Discordance among amyloid biomarkers is a challenge to overcome in order to increase diagnostic accuracy in dementia. OBJECTIVES 1) To verify that cerebrospinal fluid (CSF) Aβ42/Aβ40 ratio (AβR) better agrees with Amyloid PET (Amy-PET) results compared to CSF Aβ42; 2) to detect differences among concordant positive, concordant negative, and discordant cases, basing the concordance definition on the agreement between CSF AβR and Amy-PET results; 3) to define the suspected underlying pathology of discordant cases using in vivo biomarkers. METHOD We retrospectively enrolled 39 cognitively impaired participants in which neuropsychological tests, apolipoprotein E genotype determination, TC/MRI, FDG-PET, Amy-PET, and CSF analysis had been performed. In all cases, CSF analysis was repeated using the automated Lumipulse method. In discordant cases, FDG-PET scans were evaluated visually and using automated classifiers. RESULTS CSF AβR better agreed with Amy-PET compared to CSF Aβ42 (Cohen's K 0.431 versus 0.05). Comparisons among groups did not show any difference in clinical characteristics except for age at symptoms onset that was higher in the 6 discordant cases with abnormal CSF AβR values and negative Amy-PET (CSF AβR+/AmyPET-). FDG-PET and all CSF markers (Aβ42, AβR, p-Tau, t-Tau) were suggestive of Alzheimer's disease (AD) in 5 of these 6 cases. CONCLUSION 1) CSF AβR is the CSF amyloid marker that shows the better level of agreement with Amy-PET results; 2) The use of FDG-PET and CSF-Tau markers in CSFAβR+/Amy-PET-discordant cases can support AD diagnosis; 3) Disagreement between positive CSF AβR and negative Amy-PET in symptomatic aged AD patients could be due to the variability in plaques conformation and a negative Amy-PET scan cannot be always sufficient to rule out AD.
Collapse
Affiliation(s)
- Gemma Lombardi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,Fondazione Filippo Turati, Pistoia, Italy
| | | | | | - Cristina Polito
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", Nuclear Medicine Unit, University of Florence, Florence, Italy
| | - Sonia Padiglioni
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | | | - Valentina Berti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", Nuclear Medicine Unit, University of Florence, Florence, Italy
| | | | - Irene Piaceri
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,Fondazione IRCCS Don Carlo Gnocchi, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,Fondazione IRCCS Don Carlo Gnocchi, Florence, Italy
| |
Collapse
|
145
|
Gouilly D, Tisserand C, Nogueira L, Saint-Lary L, Rousseau V, Benaiteau M, Rafiq M, Carlier J, Milongo-Rigal E, Pagès JC, Pariente J. Taking the A Train? Limited Consistency of Aβ42 and the Aβ42/40 Ratio in the AT(N) Classification. J Alzheimers Dis 2021; 83:1033-1038. [PMID: 34397413 DOI: 10.3233/jad-210236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The consistency of cerebrospinal fluid amyloid-β (Aβ)42/40 ratio and Aβ 42 has not been assessed in the AT(N) classification system. We analyzed the classification changes of the dichotomized amyloid status (A+/A-) in 363 patients tested for Alzheimer's disease biomarkers after Aβ 42 was superseded by the Aβ 42/40 ratio. The consistency of Aβ 42 and the Aβ 42/40 ratio was very low. Notably, the proportions of "false" A+T-patients were considerable (74-91%) and corresponded mostly to patients not clinically diagnosed with Alzheimer's disease. Our results suggest that the interchangeability of Aβ 42/40 ratio and Aβ 42 is limited for classifying patients in clinical setting using the AT(N) scheme.
Collapse
Affiliation(s)
| | - Camille Tisserand
- Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - Leonor Nogueira
- Department of Cell Biology and Cytology, CHU Toulouse Purpan, France
| | - Laura Saint-Lary
- Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| | - Vanessa Rousseau
- Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| | - Marie Benaiteau
- Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - Marie Rafiq
- Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - Jasmine Carlier
- Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - Emilie Milongo-Rigal
- Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | | | - Jérémie Pariente
- Toulouse Neuroimaging Center, Toulouse, France.,Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| |
Collapse
|
146
|
Canevelli M, Rossi PD, Astrone P, Consorti E, Vanacore N, Cesari M. "Real world" eligibility for aducanumab. J Am Geriatr Soc 2021; 69:2995-2998. [PMID: 34331706 DOI: 10.1111/jgs.17390] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/10/2021] [Accepted: 07/19/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Marco Canevelli
- Department of Human Neuroscience, Sapienza University, Rome, Italy.,National Center for Disease Prevention and Health Promotion, National Institute of Health, Rome, Italy
| | - Paolo Dionigi Rossi
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Astrone
- Subacute Care Unit, Azienda Socio Sanitaria Territoriale (ASST) Lodi, Lodi, Italy
| | - Ernesto Consorti
- Fellowship in Geriatrics and Gerontology, University of Milan, Italy
| | - Nicola Vanacore
- National Center for Disease Prevention and Health Promotion, National Institute of Health, Rome, Italy
| | - Matteo Cesari
- Geriatric Unit, IRCCS Istituti Clinici Scientifici Maugeri, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
147
|
Neurotoxic Soluble Amyloid Oligomers Drive Alzheimer's Pathogenesis and Represent a Clinically Validated Target for Slowing Disease Progression. Int J Mol Sci 2021; 22:ijms22126355. [PMID: 34198582 PMCID: PMC8231952 DOI: 10.3390/ijms22126355] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
A large body of clinical and nonclinical evidence supports the role of neurotoxic soluble beta amyloid (amyloid, Aβ) oligomers as upstream pathogenic drivers of Alzheimer's disease (AD). Recent late-stage trials in AD that have evaluated agents targeting distinct species of Aβ provide compelling evidence that inhibition of Aβ oligomer toxicity represents an effective approach to slow or stop disease progression: (1) only agents that target soluble Aβ oligomers show clinical efficacy in AD patients; (2) clearance of amyloid plaque does not correlate with clinical improvements; (3) agents that predominantly target amyloid monomers or plaque failed to show clinical effects; and (4) in positive trials, efficacy is greater in carriers of the ε4 allele of apolipoprotein E (APOE4), who are known to have higher brain concentrations of Aβ oligomers. These trials also show that inhibiting Aβ neurotoxicity leads to a reduction in tau pathology, suggesting a pathogenic sequence of events where amyloid toxicity drives an increase in tau formation and deposition. The late-stage agents with positive clinical or biomarker data include four antibodies that engage Aβ oligomers (aducanumab, lecanemab, gantenerumab, and donanemab) and ALZ-801, an oral agent that fully blocks the formation of Aβ oligomers at the clinical dose.
Collapse
|
148
|
Cullen NC, Leuzy A, Janelidze S, Palmqvist S, Svenningsson AL, Stomrud E, Dage JL, Mattsson-Carlgren N, Hansson O. Plasma biomarkers of Alzheimer's disease improve prediction of cognitive decline in cognitively unimpaired elderly populations. Nat Commun 2021; 12:3555. [PMID: 34117234 PMCID: PMC8196018 DOI: 10.1038/s41467-021-23746-0] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Plasma biomarkers of amyloid, tau, and neurodegeneration (ATN) need to be characterized in cognitively unimpaired (CU) elderly individuals. We therefore tested if plasma measurements of amyloid-β (Aβ)42/40, phospho-tau217 (P-tau217), and neurofilament light (NfL) together predict clinical deterioration in 435 CU individuals followed for an average of 4.8 ± 1.7 years in the BioFINDER study. A combination of all three plasma biomarkers and basic demographics best predicted change in cognition (Pre-Alzheimer's Clinical Composite; R2 = 0.14, 95% CI [0.12-0.17]; P < 0.0001) and subsequent AD dementia (AUC = 0.82, 95% CI [0.77-0.91], P < 0.0001). In a simulated clinical trial, a screening algorithm combining all three plasma biomarkers would reduce the required sample size by 70% (95% CI [54-81]; P < 0.001) with cognition as trial endpoint, and by 63% (95% CI [53-70], P < 0.001) with subsequent AD dementia as trial endpoint. Plasma ATN biomarkers show usefulness in cognitively unimpaired populations and could make large clinical trials more feasible and cost-effective.
Collapse
Affiliation(s)
| | - Antoine Leuzy
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | | | - Sebastian Palmqvist
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Anna L Svenningsson
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | | | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
- Department of Neurology, Skåne University Hospital, Lund, Sweden.
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden.
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
149
|
Sancesario GM, Di Lazzaro G, Alwardat M, Biticchi B, Basile V, Salimei C, Colona VL, Sinibaldi Salimei P, Bernardini S, Mercuri NB, Pisani A, Schirinzi T. Amyloid-β42/Neurogranin Ratio as a Potential Index for Cognitive Impairment in Parkinson's Disease. J Alzheimers Dis 2021; 76:1171-1178. [PMID: 32597810 DOI: 10.3233/jad-200344] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Synaptopathy is critical in pathophysiology of Parkinson's disease (PD). Cerebrospinal fluid (CSF) levels of neurogranin (NG) and amyloid-β42 (Aβ42) are considered markers of synaptic dysfunction in neurodegenerative diseases. OBJECTIVE To evaluate the CSF synaptopathy-related biomarkers, especially the novel Aβ42/NG ratio, in PD, establishing possible associations with cognitive level and other clinical parameters. METHODS Levels of NG, Aβ42, amyloid-β40, total and phosphorylated tau, and Aβ42/NG ratio were measured in 30 PD patients and 30 controls and correlated with cognitive and motor parameters. The accuracy in distinguishing the cognitive status was determined. RESULTS NG and Aβ42 were significantly reduced in PD, with higher NG levels in patients with worse cognition. The Aβ42/NG ratio showed a direct correlation with Mini-Mental State Examination, independently from age and sex, and differentiated cognitively impaired patients with 92% sensitivity and 71.4% specificity, accuracy higher than NG alone. No correlations resulted with motor disturbances or therapy. CONCLUSIONS The novel Aβ42/NG ratio couples either presynaptic or postsynaptic markers of synaptic dysfunction, representing a potential global index of synaptopathy, useful to track cognitive functions in PD.
Collapse
Affiliation(s)
- Giulia Maria Sancesario
- IRCCS Fondazione Santa Lucia, Rome, Italy.,Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, Rome, Italy
| | - Giulia Di Lazzaro
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Mohammad Alwardat
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Benedetta Biticchi
- Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, Rome, Italy
| | - Valerio Basile
- Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, Rome, Italy
| | - Chiara Salimei
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Vito Luigi Colona
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | | | - Sergio Bernardini
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- IRCCS Fondazione Santa Lucia, Rome, Italy.,Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Antonio Pisani
- IRCCS Fondazione Santa Lucia, Rome, Italy.,Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Tommaso Schirinzi
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| |
Collapse
|
150
|
Reyes-Pablo AE, Campa-Córdoba BB, Luna-Viramontes NI, Ontiveros-Torres MÁ, Villanueva-Fierro I, Bravo-Muñoz M, Sáenz-Ibarra B, Barbosa O, Guadarrama-Ortíz P, Garcés-Ramírez L, de la Cruz F, Harrington CR, Martínez-Robles S, González-Ballesteros E, Perry G, Pacheco-Herrero M, Luna-Muñoz J. National Dementia BioBank: A Strategy for the Diagnosis and Study of Neurodegenerative Diseases in México. J Alzheimers Dis 2021; 76:853-862. [PMID: 32568191 DOI: 10.3233/jad-191015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We recently developed the National Dementia Biobank in México (BioBanco Nacional de Demencias, BND) as a unit for diagnosis, research, and tissue transfer for research purposes. BND is associated with the Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de Mexico (UNAM), Mexico. The donation of fluids, brain, and other organs of deceased donors is crucial for understanding the underlying mechanisms of neurodegenerative diseases and for the development of successful treatment. Our laboratory research focuses on 1) analysis of the molecular processing of the proteins involved in those neurodegenerative diseases termed tauopathies and 2) the search for biomarkers for the non-invasive and early diagnosis of Alzheimer's disease.
Collapse
Affiliation(s)
- Aldelmo Emmanuel Reyes-Pablo
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores, Cuautitlán campo 1, UNAM Estado de México, México.,Escuela Nacional de Ciencias Biológicas, Depto. Fisiología, Instituto Politécnico Nacional, CDMX, México
| | - B Berenice Campa-Córdoba
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores, Cuautitlán campo 1, UNAM Estado de México, México.,Escuela Nacional de Ciencias Biológicas, Depto. Fisiología, Instituto Politécnico Nacional, CDMX, México
| | - Nabil Itzi Luna-Viramontes
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores, Cuautitlán campo 1, UNAM Estado de México, México.,Escuela Nacional de Ciencias Biológicas, Depto. Fisiología, Instituto Politécnico Nacional, CDMX, México
| | | | | | - Marely Bravo-Muñoz
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores, Cuautitlán campo 1, UNAM Estado de México, México
| | - Bárbara Sáenz-Ibarra
- Depto. de Patología, Facultad de medicina de la Universidad Autónoma de Nuevo León, Nuevo León, México
| | - Oralia Barbosa
- Jefa del Servicio de Anatomía Patológicay Citopatología del Hospital Universitario "Dr. José E. González de la UANL, Nuevo León, México
| | | | - Linda Garcés-Ramírez
- Escuela Nacional de Ciencias Biológicas, Depto. Fisiología, Instituto Politécnico Nacional, CDMX, México
| | - Fidel de la Cruz
- Escuela Nacional de Ciencias Biológicas, Depto. Fisiología, Instituto Politécnico Nacional, CDMX, México
| | - Charles R Harrington
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Sandra Martínez-Robles
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores, Cuautitlán campo 1, UNAM Estado de México, México
| | - Erik González-Ballesteros
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores, Cuautitlán campo 1, UNAM Estado de México, México
| | - George Perry
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Mar Pacheco-Herrero
- School of Medicine, Faculty of Health Sciences, Pontificia Universidad Catolica Madre y Maestra, Dominican Republic
| | - José Luna-Muñoz
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores, Cuautitlán campo 1, UNAM Estado de México, México
| |
Collapse
|