1
|
Bhatia V, Chandel A, Minhas Y, Kushawaha SK. "Advances in biomarker discovery and diagnostics for alzheimer's disease". Neurol Sci 2025; 46:2419-2436. [PMID: 39893357 DOI: 10.1007/s10072-025-08023-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by intracellular neurofibrillary tangles with tau protein and extracellular β-amyloid plaques. Early and accurate diagnosis is crucial for effective treatment and management. OBJECTIVE The purpose of this review is to investigate new technologies that improve diagnostic accuracy while looking at the current diagnostic criteria for AD, such as clinical evaluations, cognitive testing, and biomarker-based techniques. METHODS A thorough review of the literature was done in order to assess both conventional and contemporary diagnostic methods. Multimodal strategies integrating clinical, imaging, and biochemical evaluations were emphasised. The promise of current developments in biomarker discovery was also examined, including mass spectrometry and artificial intelligence. RESULTS Current diagnostic approaches include cerebrospinal fluid (CSF) biomarkers, imaging tools (MRI, PET), cognitive tests, and new blood-based markers. Integrating these technologies into multimodal diagnostic procedures enhances diagnostic accuracy and distinguishes dementia from other conditions. New technologies that hold promise for improving biomarker identification and diagnostic reliability include mass spectrometry and artificial intelligence. CONCLUSION Advancements in AD diagnostics underscore the need for accessible, minimally invasive, and cost-effective techniques to facilitate early detection and intervention. The integration of novel technologies with traditional methods may significantly enhance the accuracy and feasibility of AD diagnosis.
Collapse
Affiliation(s)
- Vandana Bhatia
- Department of Pharmacology, Laureate Institute of Pharmacy Kathog, Kangra, 177101, India.
| | - Anjali Chandel
- Department of Pharmacology, Laureate Institute of Pharmacy Kathog, Kangra, 177101, India
| | - Yavnika Minhas
- Department of Pharmacology, Laureate Institute of Pharmacy Kathog, Kangra, 177101, India
| | | |
Collapse
|
2
|
Sun J, Geng W, Wang Y, Li H, Tan R, Tu Y. An innovative electrochemiluminescent immunosensor using dual amplified signals from AuNPs@CoSn(OH) 6 for the detection of the AD biomarker: amyloid beta 1-40. Analyst 2025; 150:2259-2269. [PMID: 40326625 DOI: 10.1039/d5an00048c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Alzheimer's disease (AD) is a degenerative condition of the nervous system that causes severe damage to patients' daily activities and quality of life. Amyloid beta 1-40 protein (Aβ40), which is involved in the formation of cerebral plaques, is one of the crucial biomarkers related to AD. Herein, a novel and highly sensitive immunosensor for the detection of Aβ40 is developed. Using a reinforced indium tin oxide-coated glass with a nanocomposite of gold nanoparticle-enhanced CoSn(OH)6 (AuNPs@CoSn(OH)6) to trigger the electrochemiluminescence (ECL) of luminol as the sensing signal, the immunosensor is fabricated by immobilizing the Aβ40 antibody onto it. By integrating the high immune specificity, excellent conductivity and catalytic activity of the nanocomposite, the resultant immunosensor can be successfully employed to detect the target in real samples. The formation of the immune complex leads to increased steric hindrance and electron transfer resistance, which in turn causes a declined ECL output when the target Aβ40 binds to the antibody on the sensor surface. Under optimized conditions, the developed ECL immunosensor exhibits a linear response for Aβ40 ranging from 1 to 800 pg mL-1 and a low detection limit of 0.47 pg mL-1. Experimentally, it is demonstrated to be highly sensitive, specific, reproducible and stable. This work extends the application of the perovskite CoSn(OH)6 and AuNPs in the field of ECL immunosensing and provides a novel strategy for clinical research on Alzheimer's disease.
Collapse
Affiliation(s)
- Jiaojing Sun
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, Suzhou, 215123, P. R. China.
| | - Wenqing Geng
- First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Yueju Wang
- First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Huiling Li
- First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
- Nursing School, Suzhou Medical College of Soochow University, Suzhou, 215006, P. R. China
| | - Rong Tan
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, Suzhou, 215123, P. R. China.
- School of Material Engineering, Changshu Institute of Technology, Suzhou, 215500, P. R. China.
| | - Yifeng Tu
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, Suzhou, 215123, P. R. China.
| |
Collapse
|
3
|
McFarlane O, Kozakiewicz M, Kędziora-Kornatowska K, Gałęska-Śliwka A, Wojciechowska M. Older Amyloid Beta as a Candidate Blood Biomarker of Early Cognitive Decline in the Elderly-A Preliminary Study. Curr Issues Mol Biol 2025; 47:203. [PMID: 40136456 PMCID: PMC11940961 DOI: 10.3390/cimb47030203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
(1) Background/Objectives: The pathogenic process of Alzheimer's disease (AD) is known to begin decades before its clinical onset. This period, although imperceptible to the patient, encompasses a gradual neuronal loss. The first symptoms of dementia, often classified as mild cognitive impairment (MCI), in many cases converts into incipient AD, but can also remain stable or even reverse to cognitive norm. An easy and fast blood-based method of identifying patients at risk of conversion to AD would allow for the application of disease-altering therapies. This preliminary study focuses on the identification and assessment of the relationship between plasma amyloid beta (Aβ) and cognitive performance in older Polish adults with respect to its adequacy as a biomarker of an early cognitive deterioration. (2) Methods: The preliminary research sample consisted of 230 participants, 109 females and 121 males, aged 65 plus. The association between plasma Aβ concentrations with cognitive status, gender, and age were assessed. The analyses were conducted in three categories of cognitive performance: cognitive norm, mild cognitive impairment, and mild dementia, based on results of the Mini-Mental State Examination (MMSE) and functional tests. (3) Results: No significant differences in plasma Aβ levels for different cognitive statuses were identified. No significant differences were found in Aβ levels based on age or gender. (4) Conclusions: In order to thoroughly explore the power of research on plasma Aβ with respect to early cognitive deterioration, further prospective studies are required.
Collapse
Affiliation(s)
- Oliwia McFarlane
- Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Faculty of Health Sciences, Department of Law and Health Policy, Świętojańska 20, 85-077 Bydgoszcz, Poland
| | - Mariusz Kozakiewicz
- Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Faculty of Health Sciences, Department of Geriatrics, Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland
| | - Kornelia Kędziora-Kornatowska
- Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Faculty of Health Sciences, Department of Geriatrics, Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland
| | - Anita Gałęska-Śliwka
- Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Faculty of Health Sciences, Department of Law and Health Policy, Świętojańska 20, 85-077 Bydgoszcz, Poland
| | - Milena Wojciechowska
- Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Faculty of Health Sciences, Department of Law and Health Policy, Świętojańska 20, 85-077 Bydgoszcz, Poland
| |
Collapse
|
4
|
Wang Y, Niu X, Zhen W, Zhang B, Chen L, Liu Y, Sun W, Peng D. Blood biomarkers of Alzheimer's disease: findings from proteomics. Postgrad Med J 2025:qgaf039. [PMID: 40094333 DOI: 10.1093/postmj/qgaf039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Alzheimer's disease, the most prevalent cause of dementia, is a worldwide health problem. Proteomics is the systematic study of proteins and peptides to provide comprehensive descriptions. Aiming to obtain a more accurate and convenient clinical diagnosis, researchers are working on blood biomarkers. METHOD This review synthesizes findings from previous studies investigating blood biomarkers for Alzheimer's disease using proteomic approaches. RESULTS We summarized the application of blood proteomics as diagnostic biomarkers and associations with clinical indicators such as neuropsychological performances, Aβ deposition and brain atrophy in Alzheimer's disease, and mild cognitive impairment. CONCLUSION In summary, blood proteomics is suggested to be promising in biomarkers of Alzheimer's disease.
Collapse
Affiliation(s)
- Yuye Wang
- Department of neurology, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang district, Beijing 100029, China
| | - Xiaoqian Niu
- Department of neurology, Xijing Hospital, Fourth Military Medical University, No. 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Weizhe Zhen
- Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing 100029, China
| | - Bin Zhang
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 2 Yinghua East Street, Chaoyang district, Beijing 100029, China
| | - Leian Chen
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 2 Yinghua East Street, Chaoyang district, Beijing 100029, China
| | - Yuchen Liu
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 2 Yinghua East Street, Chaoyang district, Beijing 100029, China
| | - Wei Sun
- Core Facility of Instrument, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, No. 5 Dong Dan San Tiao, Dongcheng District, Beijing 100005, China
| | - Dantao Peng
- Department of neurology, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang district, Beijing 100029, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 2 Yinghua East Street, Chaoyang district, Beijing 100029, China
| |
Collapse
|
5
|
Chen W, Li J, Guo J, Li L, Wu H. Diagnosis and therapy of Alzheimer's disease: Light-driven heterogeneous redox processes. Adv Colloid Interface Sci 2024; 332:103253. [PMID: 39067260 DOI: 10.1016/j.cis.2024.103253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 07/30/2024]
Abstract
Light-driven heterogeneous processes are promising approaches for diagnosing and treating Alzheimer's disease (AD) by regulating its relevant biomolecules. The molecular understanding of the heterogeneous interface environment and its interaction with target biomolecules is important. This review critically appraises the advances in AD early diagnosis and therapy employing heterogeneous light-driven redox processes, encompassing photoelectrochemical (PEC) biosensing, photodynamic therapy, photothermal therapy, PEC therapy, and photoacoustic therapy. The design strategies for heterogeneous interfaces based on target biomolecules and applications are also compiled. Finally, the remaining challenges and future perspectives are discussed. The present review may promote the fundamental understanding of AD diagnosis and therapy and facilitate interdisciplinary studies at the junction of nanotechnology and bioscience.
Collapse
Affiliation(s)
- Wenting Chen
- Macau Institute of Materials Science and Engineering (MIMSE), Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa 999078, Macau
| | - Jiahui Li
- Macau Institute of Materials Science and Engineering (MIMSE), Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa 999078, Macau
| | - Jiaxin Guo
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Liang Li
- Macau Institute of Materials Science and Engineering (MIMSE), Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa 999078, Macau
| | - Hao Wu
- Macau Institute of Materials Science and Engineering (MIMSE), Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa 999078, Macau.
| |
Collapse
|
6
|
Vagiakis I, Bakirtzis C, Andravizou A, Pirounides D. Unlocking the Potential of Vessel Density and the Foveal Avascular Zone in Optical Coherence Tomography Angiography as Biomarkers in Alzheimer's Disease. Healthcare (Basel) 2024; 12:1589. [PMID: 39201148 PMCID: PMC11353459 DOI: 10.3390/healthcare12161589] [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: 07/15/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Alzheimer's disease is the most prevalent form of dementia. Apart from its traditional clinical diagnostic methods, novel ocular imaging biomarkers have the potential to significantly enhance the diagnosis of Alzheimer's disease. Ophthalmologists might be able to play a crucial role in this multidisciplinary approach, aiding in the early detection and diagnosis of Alzheimer's disease through the use of advanced retinal imaging techniques. This systematic literature review the utilization of optical coherence tomography angiography biomarkers, specifically vessel density and the foveal avascular zone, for the diagnosis of Alzheimer's disease. A comprehensive search was performed across multiple academic journal databases, including 11 relevant studies. The selected studies underwent thorough analysis to assess the potential of these optical coherence tomography angiography biomarkers as diagnostic tools for Alzheimer's disease. The assessment of vessel density and the foveal avascular zone have emerged as a promising avenue for identifying and diagnosing Alzheimer's disease. However, it is imperative to acknowledge that further targeted investigations are warranted to address the inherent limitations of the existing body of literature. These limitations encompass various factors such as modest sample sizes, heterogeneity among study populations, disparities in optical coherence tomography angiography imaging protocols, and inconsistencies in the reported findings. In order to establish the clinical utility and robustness of these biomarkers in Alzheimer's disease diagnosis, future research endeavors should strive to overcome these limitations by implementing larger-scale studies characterized by standardized protocols and comprehensive assessments.
Collapse
Affiliation(s)
- Iordanis Vagiakis
- Department of Ophthalmology, AHEPA University Hospital, 54626 Thessaloniki, Greece;
| | - Christos Bakirtzis
- Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece;
| | - Athina Andravizou
- Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece;
| | - Demetrios Pirounides
- Department of Ophthalmology, AHEPA University Hospital, 54626 Thessaloniki, Greece;
| |
Collapse
|
7
|
Jiang YQ, Wei YP, Liu XP, Chen JS, Mao CJ, Jin BK. Strong cathode electroluminescence biosensor based on CeO 2 functionalized PCN-222@Ag NPs for sensitive detection of p-Tau-181 protein. J Colloid Interface Sci 2024; 665:144-151. [PMID: 38520931 DOI: 10.1016/j.jcis.2024.03.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/03/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
Electrochemiluminescence (ECL) biosensors provide a convenient and high sensitivity method for early disease diagnosis. However, creating luminophore arrays relying on powerful ECL signals remains a daunting task. Porphyrin-centered metal organic frameworks (MOFs) exhibit remarkable potential in ECL sensing applications. In this paper, based on a simple one-pot synthesis method, PCN-222@Ag NPs doped with CeO2 was synthesized to enhance the ECL performance. Due to the strong catalytic ability of CeO2, the ECL signal strength of the new material PCN-222@CeO2@Ag NPs is much higher than that of the PCN-222@Ag NPs and PCN-222. The luminous properties of PCN-222@CeO2@Ag NPs become more intense and stable due to the excellent electronic conductivity of Ag NPs. Based on the fact that CuS@PDA composite can quench the ECL signal of PCN-222@CeO2@Ag NPs, we constructed a novel sandwich ECL immune sensor for the detection of phosphorylated Tau 181 (p-Tau-181) protein. The ECL sensor has a great linear relationship with p-Tau-181 protein concentration, ranging from 1 pg/mL to 100 ng/mL. The detection limit is as low as 0.147 pg/mL. This work provides new ideas for developing sensitive ECL sensors for the p-Tau-181 protein, the marker of Alzheimer's disease.
Collapse
Affiliation(s)
- Yun-Qi Jiang
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China
| | - Yu-Ping Wei
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China
| | - Xing-Pei Liu
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China
| | - Jing-Shuai Chen
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China
| | - Chang-Jie Mao
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China.
| | - Bao-Kang Jin
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China
| |
Collapse
|
8
|
Seo H, Brand L, Wang H. Learning semi-supervised enrichment of longitudinal imaging-genetic data for improved prediction of cognitive decline. BMC Med Inform Decis Mak 2024; 24:61. [PMID: 38807132 PMCID: PMC11134626 DOI: 10.1186/s12911-024-02455-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/05/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Alzheimer's Disease (AD) is a progressive memory disorder that causes irreversible cognitive decline. Given that there is currently no cure, it is critical to detect AD in its early stage during the disease progression. Recently, many statistical learning methods have been presented to identify cognitive decline with temporal data, but few of these methods integrate heterogeneous phenotype and genetic information together to improve the accuracy of prediction. In addition, many of these models are often unable to handle incomplete temporal data; this often manifests itself in the removal of records to ensure consistency in the number of records across participants. RESULTS To address these issues, in this work we propose a novel approach to integrate the genetic data and the longitudinal phenotype data to learn a fixed-length "enriched" biomarker representation derived from the temporal heterogeneous neuroimaging records. Armed with this enriched representation, as a fixed-length vector per participant, conventional machine learning models can be used to predict clinical outcomes associated with AD. CONCLUSION The proposed method shows improved prediction performance when applied to data derived from Alzheimer's Disease Neruoimaging Initiative cohort. In addition, our approach can be easily interpreted to allow for the identification and validation of biomarkers associated with cognitive decline.
Collapse
Affiliation(s)
- Hoon Seo
- Department of Computer Science, Colorado School of Mines, Golden, Colorado, 80401, USA
| | - Lodewijk Brand
- Department of Computer Science, Colorado School of Mines, Golden, Colorado, 80401, USA
| | - Hua Wang
- Department of Computer Science, Colorado School of Mines, Golden, Colorado, 80401, USA.
| |
Collapse
|
9
|
Jomeiri A, Navin AH, Shamsi M. Longitudinal MRI analysis using a hybrid DenseNet-BiLSTM method for Alzheimer's disease prediction. Behav Brain Res 2024; 463:114900. [PMID: 38341100 DOI: 10.1016/j.bbr.2024.114900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/16/2023] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
Alzheimer's disease is a progressive neurological disorder characterized by brain atrophy and cell death, leading to cognitive decline and impaired functioning. Previous research has primarily focused on using cross-sectional data for Alzheimer's disease identification, but analyzing longitudinal sequential MR images is crucial for improved diagnostic accuracy and understanding disease progression. However, existing deep learning models face challenges in learning spatial and temporal features from such data. To address these challenges, this study presents a novel hybrid DenseNet-BiLSTM method for Alzheimer's disease prediction using longitudinal MRI analysis. The proposed framework combines Convolutional DenseNet for spatial information extraction and joined BiLSTM layers for capturing temporal characteristics and relationships between longitudinal images at different time points. This approach overcomes issues like overfitting, vanishing gradients, and incomplete patient data. We evaluated the model on 684 longitudinal MRI images from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database, including normal controls, individuals with mild cognitive impairment, and Alzheimer's disease patients. The results demonstrate high classification accuracy, with 95.28% for AD/CN, 88.19% for NC/MCI, 83.51% for sMCI/pMCI, and 92.14% for MCI/AD. These findings highlight the substantial improvement in Alzheimer's disease diagnosis achieved through the utilization of longitudinal MRI images. The contributions of this study lie in both the deep learning and medical domains. In the deep learning domain, our hybrid framework effectively learns spatial and temporal features from longitudinal data, addressing the challenges associated with multi-dimensional and sequential time series data. In the medical domain, our study emphasizes the importance of analyzing baseline and longitudinal MR images for accurate diagnosis and understanding disease progression.
Collapse
Affiliation(s)
- Alireza Jomeiri
- Department of Computer Engineering, Qom Branch, Islamic Azad University, Qom, Iran
| | - Ahmad Habibizad Navin
- Department of Computer Engineering, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| | - Mahboubeh Shamsi
- Department of Engineering, Qom University of Technology, Qom, Iran
| |
Collapse
|
10
|
Lista S, Mapstone M, Caraci F, Emanuele E, López-Ortiz S, Martín-Hernández J, Triaca V, Imbimbo C, Gabelle A, Mielke MM, Nisticò R, Santos-Lozano A, Imbimbo BP. A critical appraisal of blood-based biomarkers for Alzheimer's disease. Ageing Res Rev 2024; 96:102290. [PMID: 38580173 DOI: 10.1016/j.arr.2024.102290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/18/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024]
Abstract
Biomarkers that predict the clinical onset of Alzheimer's disease (AD) enable the identification of individuals in the early, preclinical stages of the disease. Detecting AD at this point may allow for more effective therapeutic interventions and optimized enrollment for clinical trials of novel drugs. The current biological diagnosis of AD is based on the AT(N) classification system with the measurement of brain deposition of amyloid-β (Aβ) ("A"), tau pathology ("T"), and neurodegeneration ("N"). Diagnostic cut-offs for Aβ1-42, the Aβ1-42/Aβ1-40 ratio, tau and hyperphosphorylated-tau concentrations in cerebrospinal fluid have been defined and may support AD clinical diagnosis. Blood-based biomarkers of the AT(N) categories have been described in the AD continuum. Cross-sectional and longitudinal studies have shown that the combination of blood biomarkers tracking neuroaxonal injury (neurofilament light chain) and neuroinflammatory pathways (glial fibrillary acidic protein) enhance sensitivity and specificity of AD clinical diagnosis and improve the prediction of AD onset. However, no international accepted cut-offs have been identified for these blood biomarkers. A kit for blood Aβ1-42/Aβ1-40 is commercially available in the U.S.; however, it does not provide a diagnosis, but simply estimates the risk of developing AD. Although blood-based AD biomarkers have a great potential in the diagnostic work-up of AD, they are not ready for the routine clinical use.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Mark Mapstone
- Department of Neurology, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA.
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania 95125, Italy; Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, Troina 94018, Italy.
| | | | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Juan Martín-Hernández
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Rome 00015, Italy.
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy.
| | - Audrey Gabelle
- Memory Resources and Research Center, Montpellier University of Excellence i-site, Montpellier 34295, France.
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA.
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", Rome 00133, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome 00143, Italy.
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain; Physical Activity and Health Research Group (PaHerg), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid 28041, Spain.
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma 43122, Italy.
| |
Collapse
|
11
|
Zhuang H, Cao X, Tang X, Zou Y, Yang H, Liang Z, Yan X, Chen X, Feng X, Shen L. Investigating metabolic dysregulation in serum of triple transgenic Alzheimer's disease male mice: implications for pathogenesis and potential biomarkers. Amino Acids 2024; 56:10. [PMID: 38315232 PMCID: PMC10844422 DOI: 10.1007/s00726-023-03375-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/11/2023] [Indexed: 02/07/2024]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease that lacks convenient and accessible peripheral blood diagnostic markers and effective drugs. Metabolic dysfunction is one of AD risk factors, which leaded to alterations of various metabolites in the body. Pathological changes of the brain can be reflected in blood metabolites that are expected to explain the disease mechanisms or be candidate biomarkers. The aim of this study was to investigate the changes of targeted metabolites within peripheral blood of AD mouse model, with the purpose of exploring the disease mechanism and potential biomarkers. Targeted metabolomics was used to quantify 256 metabolites in serum of triple transgenic AD (3 × Tg-AD) male mice. Compared with controls, 49 differential metabolites represented dysregulation in purine, pyrimidine, tryptophan, cysteine and methionine and glycerophospholipid metabolism. Among them, adenosine, serotonin, N-acetyl-5-hydroxytryptamine, and acetylcholine play a key role in regulating neural transmitter network. The alteration of S-adenosine-L-homocysteine, S-adenosine-L-methionine, and trimethylamine-N-oxide in AD mice serum can served as indicator of AD risk. The results revealed the changes of metabolites in serum, suggesting that metabolic dysregulation in periphery in AD mice may be related to the disturbances in neuroinhibition, the serotonergic system, sleep function, the cholinergic system, and the gut microbiota. This study provides novel insights into the dysregulation of several key metabolites and metabolic pathways in AD, presenting potential avenues for future research and the development of peripheral biomarkers.
Collapse
Affiliation(s)
- Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Yongdong Zou
- Center for Instrumental Analysis, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Hongbo Yang
- Center for Instrumental Analysis, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Zhiyuan Liang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xi Yan
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, People's Republic of China
| | - Xiaolu Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, People's Republic of China
| | - Xingui Feng
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, People's Republic of China.
| |
Collapse
|
12
|
Kim JS, Kim MG, Ryu JE, Lee YB, Liu QF, Kim KK, Cho SH, Shin SJ, Koo BS, Choi HK. Effect of woohwangchungsimwon and donepezil co-treatment on cognitive function and serum metabolic profiles in a scopolamine-induced model of Alzheimer's disease. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117359. [PMID: 37924999 DOI: 10.1016/j.jep.2023.117359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Woohwangchungsimwon (WCW) is a traditional medicine used in East Asian countries to treat central nervous system disorders. Reported pharmacological properties include antioxidant effects, enhanced learning and memory, and protection against ischemic neuronal cell death, supporting its use in treating neurodegenerative diseases like Alzheimer's disease (AD). AIM OF THE STUDY The study aims to assess the effects of co-treatment with WCW and donepezil on cognitive functions and serum metabolic profiles in a scopolamine-induced AD model. MATERIALS AND METHODS Cell viability and reactive oxygen species (ROS) levels were measured in amyloid β-peptide25-35 (Aβ25-35)-induced SH-SY5Y cells. An AD model was established in ICR mice by intraperitoneal scopolamine administration. Animals underwent the step-through passive avoidance test (PAT) and Morris water maze (MWM) test. Hippocampal tissues were collected to examine specific protein expression. Serum metabolic profiles were analyzed using nuclear magnetic resonance (NMR) spectroscopy. RESULTS Co-treatment with WCW and donepezil increased cell viability and reduced ROS production in Aβ25-35-induced SH-SY5Y cells compared to that with donepezil treatment alone. Co-treatment improved cognitive functions and was comparable to donepezil treatment alone in the PAT and MWM tests. Pathways related to tyrosine, phenylalanine, and tryptophan biosynthesis, phenylalanine metabolism, and cysteine and methionine metabolism were altered by co-treatment. Levels of tyrosine and methionine, major serum metabolites in these pathways, were significantly reduced after co-treatment. CONCLUSIONS Co-treatment with WCW and donepezil shows promise as a therapeutic strategy for AD and is comparable to donepezil alone in improving cognitive function. Reduced tyrosine and methionine levels after co-treatment may enhance cognitive function by mitigating hypertyrosinemia and hyperhomocysteinemia, known risk factors for AD. The serum metabolic profiles obtained in this study can serve as a foundation for developing other bioactive compounds using a scopolamine-induced mouse model.
Collapse
Affiliation(s)
- Jung-Seop Kim
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Man-Gi Kim
- Department of Neuropsychiatry, College of Korean Medicine, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea
| | - Ji Eun Ryu
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Ye-Been Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Quan Feng Liu
- Department of Neuropsychiatry, College of Korean Medicine, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea
| | - Kwang Ki Kim
- Department of Neurology, Dongguk University Ilsan Hospital, Goyang, Gyeonggi-do, Republic of Korea
| | - Seung-Hun Cho
- Department of Neuropsychiatry, College of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul, Republic of Korea
| | - Sung Joon Shin
- Division of Nephrology, Department of Internal Medicine, Medical Cannabis Center, Dongguk University Ilsan Hospital, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea
| | - Byung-Soo Koo
- Department of Neuropsychiatry, College of Korean Medicine, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea.
| | - Hyung-Kyoon Choi
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Bhandari M, Tiwari RK, Chanda S, Bonde GV. Targeting angiogenesis, inflammation, and oxidative stress in Alzheimer's diseases. TARGETING ANGIOGENESIS, INFLAMMATION, AND OXIDATIVE STRESS IN CHRONIC DISEASES 2024:215-249. [DOI: 10.1016/b978-0-443-13587-3.00003-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
14
|
Tufail S, Sherwani MA, Shamim Z, Abdullah, Goh KW, Alomary MN, Ansari MA, Almosa AA, Ming LC, Abdullah ADI, Khan FB, Menhali AA, Mirza S, Ayoub MA. 2D nanostructures: Potential in diagnosis and treatment of Alzheimer's disease. Biomed Pharmacother 2024; 170:116070. [PMID: 38163396 DOI: 10.1016/j.biopha.2023.116070] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/06/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024] Open
Abstract
Two-dimensional (2D) nanomaterials have garnered enormous attention seemingly due to their unusual architecture and properties. Graphene and graphene oxide based 2D nanomaterials remained the most sought after for several years but the quest to design superior 2D nanomaterials which can find wider application gave rise to development of non-graphene 2D materials as well. Consequently, in addition to graphene based 2D nanomaterials, 2D nanostructures designed using macromolecules (such as DNAs, proteins, peptides and peptoids), transition metal dichalcogenides, transition-metal carbides and/or nitrides (MXene), black phosphorous, chitosan, hexagonal boron nitrides, and graphitic carbon nitride, and covalent organic frameworks have been developed. Interestingly, these 2D nanomaterials have found applications in diagnosis and treatment of various diseases including Alzheimer's disease (AD). Although AD is one of the most debilitating neurodegenerative conditions across the globe; unfortunately, there remains a paucity of effective diagnostic and/or therapeutic intervention for it till date. In this scenario, nanomaterial-based biosensors, or therapeutics especially 2D nanostructures are emerging to be promising in this regard. This review summarizes the diagnostic and therapeutic platforms developed for AD using 2D nanostructures. Collectively, it is worth mentioning that these 2D nanomaterials would seemingly provide an alternative and intriguing platform for biomedical interventions.
Collapse
Affiliation(s)
- Saba Tufail
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA.
| | | | - Zahid Shamim
- Department of Electrical Engineering, Faculty of Engineering and Technology, Aligarh Muslim University, Aligarh, India
| | - Abdullah
- Department of Pharmacy, University of Malakand, Khyber Pakhtunkhwa, Pakistan
| | - Khang Wen Goh
- Faculty Data Science and Information Technology, INTI International University, Nilai, Malaysia
| | - Mohammad N Alomary
- Advanced Diagnostic and Therapeutic Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.
| | - Abdulaziz Abdullah Almosa
- Wellness and Preventive Medicine Institute, King AbdulAziz City of Science and Technology, Riyadh, Saudi Arabia.
| | - Long Chiau Ming
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia.
| | - Amar Daud Iskandar Abdullah
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia.
| | - Farheen Badrealam Khan
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates; Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, PO Box 127788, Abu Dhabi, United Arab Emirates.
| | - Asma Al Menhali
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates.
| | - Sameer Mirza
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates.
| | - Mohammed Akli Ayoub
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, PO Box 127788, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
15
|
Jethwa A, Stöckl L. Optimized Pre-analytical Handling Protocol for Blood-Based Biomarkers of Alzheimer's Disease. Methods Mol Biol 2024; 2785:67-73. [PMID: 38427188 DOI: 10.1007/978-1-0716-3774-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The therapeutic management of patients with Alzheimer's disease (AD) has been hindered by poor diagnostic accuracy. As such, there is an unmet clinical need for tools that can detect and diagnose the disease in its early stages. Compared with cerebrospinal fluid (CSF)-based biomarkers or positron emission tomography (PET), the use of reliable blood-based biomarkers could offer an accessible and minimally invasive method of streamlining diagnosis in the clinical setting. However, the influence of pre-analytical processing and sample handling parameters on the accurate measurement of protein biomarkers is well established, especially for AD CSF-based biomarkers. In this chapter, we provide recommendations for an optimal sample handling protocol for the analysis of blood-based biomarkers specifically for amyloid pathology in AD.
Collapse
|
16
|
Toader C, Dobrin N, Brehar FM, Popa C, Covache-Busuioc RA, Glavan LA, Costin HP, Bratu BG, Corlatescu AD, Popa AA, Ciurea AV. From Recognition to Remedy: The Significance of Biomarkers in Neurodegenerative Disease Pathology. Int J Mol Sci 2023; 24:16119. [PMID: 38003309 PMCID: PMC10671641 DOI: 10.3390/ijms242216119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
With the inexorable aging of the global populace, neurodegenerative diseases (NDs) like Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) pose escalating challenges, which are underscored by their socioeconomic repercussions. A pivotal aspect in addressing these challenges lies in the elucidation and application of biomarkers for timely diagnosis, vigilant monitoring, and effective treatment modalities. This review delineates the quintessence of biomarkers in the realm of NDs, elucidating various classifications and their indispensable roles. Particularly, the quest for novel biomarkers in AD, transcending traditional markers in PD, and the frontier of biomarker research in ALS are scrutinized. Emergent susceptibility and trait markers herald a new era of personalized medicine, promising enhanced treatment initiation especially in cases of SOD1-ALS. The discourse extends to diagnostic and state markers, revolutionizing early detection and monitoring, alongside progression markers that unveil the trajectory of NDs, propelling forward the potential for tailored interventions. The synergy between burgeoning technologies and innovative techniques like -omics, histologic assessments, and imaging is spotlighted, underscoring their pivotal roles in biomarker discovery. Reflecting on the progress hitherto, the review underscores the exigent need for multidisciplinary collaborations to surmount the challenges ahead, accelerate biomarker discovery, and herald a new epoch of understanding and managing NDs. Through a panoramic lens, this article endeavors to provide a comprehensive insight into the burgeoning field of biomarkers in NDs, spotlighting the promise they hold in transforming the diagnostic landscape, enhancing disease management, and illuminating the pathway toward efficacious therapeutic interventions.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Nicolaie Dobrin
- Department of Neurosurgery, Clinical Emergency Hospital “Prof. Dr. Nicolae Oblu”, 700309 Iasi, Romania
| | - Felix-Mircea Brehar
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
- Department of Neurosurgery, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Constantin Popa
- Department of Neurology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
- Medical Science Section, Romanian Academy, 060021 Bucharest, Romania
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Luca Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Horia Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Antonio Daniel Corlatescu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Andrei Adrian Popa
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
- Medical Science Section, Romanian Academy, 060021 Bucharest, Romania
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
17
|
Wang Y, Sun Y, Wang Y, Jia S, Qiao Y, Zhou Z, Shao W, Zhang X, Guo J, Zhang B, Niu X, Wang Y, Peng D. Identification of novel diagnostic panel for mild cognitive impairment and Alzheimer's disease: findings based on urine proteomics and machine learning. Alzheimers Res Ther 2023; 15:191. [PMID: 37925455 PMCID: PMC10625308 DOI: 10.1186/s13195-023-01324-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/04/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND Alzheimer's disease is a prevalent disease with a heavy global burden. Proteomics is the systematic study of proteins and peptides to provide comprehensive descriptions. Aiming to obtain a more accurate and convenient clinical diagnosis, researchers are working for better biomarkers. Urine is more convenient which could reflect the change of disease at an earlier stage. Thus, we conducted a cross-sectional study to investigate novel diagnostic panels. METHODS We firstly enrolled participants from China-Japan Friendship Hospital from April 2022 to November 2022, collected urine samples, and conducted an LC-MS/MS analysis. In parallel, clinical data were collected, and clinical examinations were performed. After statistical and bioinformatics analyses, significant risk factors and differential urinary proteins were determined. We attempt to investigate diagnostic panels based on machine learning including LASSO and SVM. RESULTS Fifty-seven AD patients, 43 MCI patients, and 62 CN subjects were enrolled. A total of 3366 proteins were identified, and 608 urine proteins were finally included in the analysis. There were 33 significantly differential proteins between the AD and CN groups and 15 significantly differential proteins between the MCI and CN groups. AD diagnostic panel included DDC, CTSC, EHD4, GSTA3, SLC44A4, GNS, GSTA1, ANXA4, PLD3, CTSH, HP, RPS3, CPVL, age, and APOE ε4 with an AUC of 0.9989 in the training test and 0.8824 in the test set while MCI diagnostic panel included TUBB, SUCLG2, PROCR, TCP1, ACE, FLOT2, EHD4, PROZ, C9, SERPINA3, age, and APOE ε4 with an AUC of 0.9985 in the training test and 0.8143 in the test set. Besides, diagnostic proteins were weakly correlated with cognitive functions. CONCLUSIONS In conclusion, the procedure is convenient, non-invasive, and useful for diagnosis, which could assist physicians in differentiating AD and MCI from CN.
Collapse
Affiliation(s)
- Yuye Wang
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yu Sun
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yu Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Shuhong Jia
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yanan Qiao
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Zhi Zhou
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Wen Shao
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiangfei Zhang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jing Guo
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Bin Zhang
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiaoqian Niu
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.
| | - Dantao Peng
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China.
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.
| |
Collapse
|
18
|
Al Abdullah S, Najm L, Ladouceur L, Ebrahimi F, Shakeri A, Al-Jabouri N, Didar TF, Dellinger K. Functional Nanomaterials for the Diagnosis of Alzheimer's Disease: Recent Progress and Future Perspectives. ADVANCED FUNCTIONAL MATERIALS 2023; 33:2302673. [PMID: 39309539 PMCID: PMC11415277 DOI: 10.1002/adfm.202302673] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Indexed: 09/25/2024]
Abstract
Alzheimer's disease (AD) is one of the main causes of dementia worldwide, whereby neuronal death or malfunction leads to cognitive impairment in the elderly population. AD is highly prevalent, with increased projections over the next few decades. Yet current diagnostic methods for AD occur only after the presentation of clinical symptoms. Evidence in the literature points to potential mechanisms of AD induction beginning before clinical symptoms start to present, such as the formation of amyloid beta (Aβ) extracellular plaques and neurofibrillary tangles (NFTs). Biomarkers of AD, including Aβ 40, Aβ 42, and tau protein, amongst others, show promise for early AD diagnosis. Additional progress is made in the application of biosensing modalities to measure and detect significant changes in these AD biomarkers within patient samples, such as cerebral spinal fluid (CSF) and blood, serum, or plasma. Herein, a comprehensive review of the emerging nano-biomaterial approaches to develop biosensors for AD biomarkers' detection is provided. Advances, challenges, and potential of electrochemical, optical, and colorimetric biosensors, focusing on nanoparticle-based (metallic, magnetic, quantum dots) and nanostructure-based biomaterials are discussed. Finally, the criteria for incorporating these emerging nano-biomaterials in clinical settings are presented and assessed, as they hold great potential for enhancing early-onset AD diagnostics.
Collapse
Affiliation(s)
- Saqer Al Abdullah
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, NC 27401, USA
| | - Lubna Najm
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| | - Liane Ladouceur
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada
| | - Farbod Ebrahimi
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, NC 27401, USA
| | - Amid Shakeri
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada
| | - Nadine Al-Jabouri
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Tohid F Didar
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada
- Institute for Infectious Disease Research (IIDR), 1280 Main St W, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, NC 27401, USA
| |
Collapse
|
19
|
Gao F, Li F, Wang J, Yu H, Li X, Chen H, Wang J, Qin D, Li Y, Liu S, Zhang X, Wang ZH. SERS-Based Optical Nanobiosensors for the Detection of Alzheimer's Disease. BIOSENSORS 2023; 13:880. [PMID: 37754114 PMCID: PMC10526933 DOI: 10.3390/bios13090880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/28/2023]
Abstract
Alzheimer's disease (AD) is a leading cause of dementia, impacting millions worldwide. However, its complex neuropathologic features and heterogeneous pathophysiology present significant challenges for diagnosis and treatment. To address the urgent need for early AD diagnosis, this review focuses on surface-enhanced Raman scattering (SERS)-based biosensors, leveraging the excellent optical properties of nanomaterials to enhance detection performance. These highly sensitive and noninvasive biosensors offer opportunities for biomarker-driven clinical diagnostics and precision medicine. The review highlights various types of SERS-based biosensors targeting AD biomarkers, discussing their potential applications and contributions to AD diagnosis. Specific details about nanomaterials and targeted AD biomarkers are provided. Furthermore, the future research directions and challenges for improving AD marker detection using SERS sensors are outlined.
Collapse
Affiliation(s)
- Feng Gao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fang Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jianhao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hang Yu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiang Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hongyu Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiabei Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Dongdong Qin
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yiyi Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Songyan Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xi Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhi-Hao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (F.G.); (F.L.); (J.W.); (H.Y.); (X.L.); (H.C.); (J.W.); (D.Q.); (Y.L.); (S.L.); (X.Z.)
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
20
|
Xu C, Zhao L, Dong C. The performance of plasma phosphorylated tau231 in detecting Alzheimer's disease: A systematic review with meta-analysis. Eur J Neurosci 2023; 58:3132-3149. [PMID: 37501373 DOI: 10.1111/ejn.16085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023]
Abstract
Cerebrospinal fluid (CSF) phosphorylated tau231 (P-tau231) is associated with neuropathological outcomes of Alzheimer's disease (AD). The invasive access of cerebrospinal fluid has greatly stimulated interest in the identification of blood-based P-tau231, and the recent advent of single-molecule array assay for the quantification of plasma P-tau231 may provide a turning point to evaluate the usefulness of P-tau231 as an AD-related biomarker. Yet, in the plasma P-tau231 literature, findings with regard to its diagnostic utility have been inconsistent, and thus, we aimed to statistically investigate the potential of plasma P-tau231 in the context of AD via meta-analysis. Publications on plasma P-tau231 were systematically retrieved from PubMed, EMBASE, the Cochrane library and Web of Science databases. A total of 10 studies covering 2007 participants were included, and we conducted random-effect or fixed-effect meta-analysis, sensitivity analysis and publication bias analysis using the STATA SE 14.0 software. According to our quantitative integration, plasma P-tau231 increased from cognitively unimpaired (CU) populations to mild cognitive impairment to AD and showed significant changes in pairwise comparisons of AD, mild cognitive impairment and CU. Plasma P-tau231 level was significantly higher in CU controls with positive amyloid-β (Aβ) status compared with Aβ-negative CU group. Additionally, the excellent diagnostic accuracy of plasma P-tau231 for asymptomatic Aβ pathology was verified by the pooled value of area under the receiver operating characteristic curves (standard mean difference [95% confidence interval]: .75 [.69, .81], P < 0.00001). Overall, the increased plasma P-tau231 concentrations were found in relation to the early development and progression of AD.
Collapse
Affiliation(s)
- Chang Xu
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Li Zhao
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunbo Dong
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
21
|
Whitfield JF, Rennie K, Chakravarthy B. Alzheimer's Disease and Its Possible Evolutionary Origin: Hypothesis. Cells 2023; 12:1618. [PMID: 37371088 PMCID: PMC10297544 DOI: 10.3390/cells12121618] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/29/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The enormous, 2-3-million-year evolutionary expansion of hominin neocortices to the current enormity enabled humans to take over the planet. However, there appears to have been a glitch, and it occurred without a compensatory expansion of the entorhinal cortical (EC) gateway to the hippocampal memory-encoding system needed to manage the processing of the increasing volume of neocortical data converging on it. The resulting age-dependent connectopathic glitch was unnoticed by the early short-lived populations. It has now surfaced as Alzheimer's disease (AD) in today's long-lived populations. With advancing age, processing of the converging neocortical data by the neurons of the relatively small lateral entorhinal cortex (LEC) inflicts persistent strain and high energy costs on these cells. This may result in their hyper-release of harmless Aβ1-42 monomers into the interstitial fluid, where they seed the formation of toxic amyloid-β oligomers (AβOs) that initiate AD. At the core of connectopathic AD are the postsynaptic cellular prion protein (PrPC). Electrostatic binding of the negatively charged AβOs to the positively charged N-terminus of PrPC induces hyperphosphorylation of tau that destroys synapses. The spread of these accumulating AβOs from ground zero is supported by Aβ's own production mediated by target cells' Ca2+-sensing receptors (CaSRs). These data suggest that an early administration of a strongly positively charged, AβOs-interacting peptide or protein, plus an inhibitor of CaSR, might be an effective AD-arresting therapeutic combination.
Collapse
Affiliation(s)
- James F. Whitfield
- Human Health Therapeutics, National Research Council, Ottawa, ON K1A 0R6, Canada
| | | | | |
Collapse
|
22
|
Kurz C, Stöckl L, Schrurs I, Suridjan I, Gürsel SÜ, Bittner T, Jethwa A, Perneczky R. Impact of pre-analytical sample handling factors on plasma biomarkers of Alzheimer's disease. J Neurochem 2023; 165:95-105. [PMID: 36625424 DOI: 10.1111/jnc.15757] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/07/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023]
Abstract
An unmet need exists for reliable plasma biomarkers of amyloid pathology, in the clinical laboratory setting, to streamline diagnosis of Alzheimer's disease (AD). For routine clinical use, a biomarker must provide robust and reliable results under pre-analytical sample handling conditions. We investigated the impact of different pre-analytical sample handling procedures on the levels of seven plasma biomarkers in development for potential routine use in AD. Using (1) fresh (never frozen) and (2) previously frozen plasma, we evaluated the effects of (A) storage time and temperature, (B) freeze/thaw (F/T) cycles, (C) anticoagulants, (D) tube transfer, and (E) plastic tube types. Blood samples were prospectively collected from patients with cognitive impairment undergoing investigation in a memory clinic. β-amyloid 1-40 (Aβ40), β-amyloid 1-42 (Aβ42), apolipoprotein E4, glial fibrillary acidic protein, neurofilament light chain, phosphorylated-tau (phospho-tau) 181, and phospho-tau-217 were measured using Elecsys® plasma prototype immunoassays. Recovery signals for each plasma biomarker and sample handling parameter were calculated. For all plasma biomarkers measured, pre-analytical effects were comparable between fresh (never frozen) and previously frozen samples. All plasma biomarkers tested were stable for ≤24 h at 4°C when stored as whole blood and ethylenediaminetetraacetic acid (EDTA) plasma. Recovery signals were acceptable for up to five tube transfers, or two F/T cycles, and in both polypropylene and low-density polyethylene tubes. For all plasma biomarkers except Aβ42 and Aβ40, analyte levels were largely comparable between EDTA, lithium heparin, and sodium citrate tubes. Aβ42 and Aβ40 were most sensitive to pre-analytical handling, and the effects could only be partially compensated by the Aβ42/Aβ40 ratio. We provide recommendations for an optimal sample handling protocol for analysis of plasma biomarkers for amyloid pathology AD, to improve the reproducibility of future studies on plasma biomarkers assays and for potential use in routine clinical practice.
Collapse
Affiliation(s)
- Carolin Kurz
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | | | | | | | - Selim Üstün Gürsel
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | | | | | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.,German Center for Neurodegenerative Disorders (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK.,Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| |
Collapse
|
23
|
Panikkar D, Vivek S, Crimmins E, Faul J, Langa KM, Thyagarajan B. Pre-Analytical Variables Influencing Stability of Blood-Based Biomarkers of Neuropathology. J Alzheimers Dis 2023; 95:735-748. [PMID: 37574735 PMCID: PMC11520930 DOI: 10.3233/jad-230384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
BACKGROUND Sample collection and preanalytical protocols may significantly impact the results of large-scale epidemiological studies incorporating blood-based biomarkers of neuropathology. OBJECTIVE To evaluate the stability and assay variability of several blood-based biomarkers of neuropathology for common preanalytical conditions. METHODS We collected serum and plasma samples from 41 participants and evaluated the effect of processing delay of up to 72 h when stored at 4∘C, three freeze-thaw cycles, and a combination of 48-h processing delay when stored at 4∘C and three freeze-thaw cycles on biomarker stability. Using the Simoa assay (Quanterix Inc.), we measured amyloid-β 40 (Aβ40), amyloid-β 42 (Aβ42), neurofilament light (NfL), glial fibrillary acidic protein (GFAP), and phosphorylated tau 181 (p-tau-181). RESULTS We found that Aβ40 and Aβ42 levels significantly decreased after a 24-h processing delay in both plasma and serum samples, and a single freeze-thaw cycle (p < 0.0001). Nevertheless, serum Aβ42/40 ratio remained stable with a processing delay up to 48 h while plasma Aβ42/40 ratio showed only small but significant increase with a delay up to 72 h. Both plasma and serum GFAP and NfL levels were only modestly affected by processing delay and freeze-thaw cycles. Plasma p-tau-181 levels notably increased with a 24-, 48-, and 72-h processing delay, but remained stable in serum. Intra-individual variation over two weeks was minimal for all biomarkers and their levels were substantially lower in serum when compared with plasma. CONCLUSION These results suggest that standardizing preanalytical variables will allow robust measurements of biomarkers of neuropathology in population studies.
Collapse
Affiliation(s)
- Daniel Panikkar
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Sithara Vivek
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Eileen Crimmins
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Jessica Faul
- Institute for Social Research, Survey Research Center, University of Michigan, Ann Arbor, MI, USA
| | - Kenneth M. Langa
- Institute for Social Research, Survey Research Center, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Veterans Affairs Ann Arbor Center for Clinical Management Research, Ann Arbor, MI, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
24
|
Chiara C, Gilda C, Daniela M, Antonio C, Miriana M, Marcello M, Elena S, Roberta L, Ciro C, Vincenzo BM. A two-year longitudinal study of retinal vascular impairment in patients with amnestic mild cognitive impairment. Front Aging Neurosci 2022; 14:993621. [PMID: 36420311 PMCID: PMC9678013 DOI: 10.3389/fnagi.2022.993621] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
ObjectiveTo evaluate the relation between retinal vascular impairment and cognitive decline in patients with amnestic mild cognitive impairment (aMCI) over time.MethodsSpectral domain-optical coherence tomography (SD-OCT) and OCT angiography study was performed in aMCI patients over 2 years follow-up and compared to baseline.ResultsThirty-eight eyes from 19 aMCI patients were evaluated. Structural and vascular OCT measures were reduced at follow-up except for vessel density (VD) of the choriocapillaris, unchanged, and foveal avascular zone, which was increased; no changes in any parameter were found in 18 age-matched healthy controls. Overall, these findings were confirmed when patients were evaluated separately according to progression to dementia. Only non-converters to dementia showed significant VD reduction in the deep capillary plexuses (coeff. β = −4.20; p < 0.001), may be for an initial massive VD depletion becoming less evident with progression of the disease. MMSE reduction was associated with a higher ganglion cell complex reduction (coeff. β = 0.10; p = 0.04) and a higher VD reduction in the radial peripapillary capillary (RPC) plexus (coeff. β = 0.14; p = 0.02) in the whole patient group, while it was associated with a higher VD reduction only in RPC plexus in converters (coeff. β = 0.21; p < 0.001).ConclusionOur data shows vascular impairment progression in the inner retina of aMCI patients and support the hypothesis that vascular changes may contribute to the onset and progression of Alzheimer’s disease. Other follow-up studies, with a larger number of patients, are needed to better define VD as a potential biomarker.
Collapse
|
25
|
Nardini M, Ciasca G, Lauria A, Rossi C, Di Giacinto F, Romanò S, Di Santo R, Papi M, Palmieri V, Perini G, Basile U, Alcaro FD, Di Stasio E, Bizzarro A, Masullo C, De Spirito M. Sensing red blood cell nano-mechanics: Toward a novel blood biomarker for Alzheimer's disease. Front Aging Neurosci 2022; 14:932354. [PMID: 36204549 PMCID: PMC9530048 DOI: 10.3389/fnagi.2022.932354] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Red blood cells (RBCs) are characterized by a remarkable elasticity, which allows them to undergo very large deformation when passing through small vessels and capillaries. This extreme deformability is altered in various clinical conditions, suggesting that the analysis of red blood cell (RBC) mechanics has potential applications in the search for non-invasive and cost-effective blood biomarkers. Here, we provide a comparative study of the mechanical response of RBCs in patients with Alzheimer's disease (AD) and healthy subjects. For this purpose, RBC viscoelastic response was investigated using atomic force microscopy (AFM) in the force spectroscopy mode. Two types of analyses were performed: (i) a conventional analysis of AFM force-distance (FD) curves, which allowed us to retrieve the apparent Young's modulus, E; and (ii) a more in-depth analysis of time-dependent relaxation curves in the framework of the standard linear solid (SLS) model, which allowed us to estimate cell viscosity and elasticity, independently. Our data demonstrate that, while conventional analysis of AFM FD curves fails in distinguishing the two groups, the mechanical parameters obtained with the SLS model show a very good classification ability. The diagnostic performance of mechanical parameters was assessed using receiving operator characteristic (ROC) curves, showing very large areas under the curves (AUC) for selected biomarkers (AUC > 0.9). Taken all together, the data presented here demonstrate that RBC mechanics are significantly altered in AD, also highlighting the key role played by viscous forces. These RBC abnormalities in AD, which include both a modified elasticity and viscosity, could be considered a potential source of plasmatic biomarkers in the field of liquid biopsy to be used in combination with more established indicators of the pathology.
Collapse
Affiliation(s)
- Matteo Nardini
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gabriele Ciasca
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alessandra Lauria
- Unitá Operativa Complessa Neuroriabilitazione ad Alta Intensitá, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cristina Rossi
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Flavio Di Giacinto
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Sabrina Romanò
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Riccardo Di Santo
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Massimiliano Papi
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Valentina Palmieri
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Istituto dei Sistemi Complessi (ISC), Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Giordano Perini
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Umberto Basile
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesca D. Alcaro
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Enrico Di Stasio
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alessandra Bizzarro
- Unitáă Operativa Complessa Continuità assistenziale, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Masullo
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Sezione di Neurologia, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco De Spirito
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
26
|
Mumtaz I, Ayaz MO, Khan MS, Manzoor U, Ganayee MA, Bhat AQ, Dar GH, Alghamdi BS, Hashem AM, Dar MJ, Ashraf GM, Maqbool T. Clinical relevance of biomarkers, new therapeutic approaches, and role of post-translational modifications in the pathogenesis of Alzheimer's disease. Front Aging Neurosci 2022; 14:977411. [PMID: 36158539 PMCID: PMC9490081 DOI: 10.3389/fnagi.2022.977411] [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: 06/24/2022] [Accepted: 08/18/2022] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that causes progressive loss of cognitive functions like thinking, memory, reasoning, behavioral abilities, and social skills thus affecting the ability of a person to perform normal daily functions independently. There is no definitive cure for this disease, and treatment options available for the management of the disease are not very effective as well. Based on histopathology, AD is characterized by the accumulation of insoluble deposits of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs). Although several molecular events contribute to the formation of these insoluble deposits, the aberrant post-translational modifications (PTMs) of AD-related proteins (like APP, Aβ, tau, and BACE1) are also known to be involved in the onset and progression of this disease. However, early diagnosis of the disease as well as the development of effective therapeutic approaches is impeded by lack of proper clinical biomarkers. In this review, we summarized the current status and clinical relevance of biomarkers from cerebrospinal fluid (CSF), blood and extracellular vesicles involved in onset and progression of AD. Moreover, we highlight the effects of several PTMs on the AD-related proteins, and provide an insight how these modifications impact the structure and function of proteins leading to AD pathology. Finally, for disease-modifying therapeutics, novel approaches, and targets are discussed for the successful treatment and management of AD.
Collapse
Affiliation(s)
- Ibtisam Mumtaz
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| | - Mir Owais Ayaz
- Laboratory of Cell and Molecular Biology, Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Centre for Scientific and Innovative Research, Ghaziabad, Utter Pradesh, India
| | - Mohamad Sultan Khan
- Neurobiology and Molecular Chronobiology Laboratory, Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Umar Manzoor
- Laboratory of Immune and Inflammatory Disease, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, South Korea
| | - Mohd Azhardin Ganayee
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, India
| | - Aadil Qadir Bhat
- Laboratory of Cell and Molecular Biology, Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Centre for Scientific and Innovative Research, Ghaziabad, Utter Pradesh, India
| | - Ghulam Hassan Dar
- Sri Pratap College, Cluster University Srinagar, Jammu and Kashmir, India
| | - Badrah S. Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pre-clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anwar M. Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohd Jamal Dar
- Laboratory of Cell and Molecular Biology, Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Centre for Scientific and Innovative Research, Ghaziabad, Utter Pradesh, India
| | - Gulam Md. Ashraf
- Pre-clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tariq Maqbool
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| |
Collapse
|
27
|
Proteomic Discovery and Validation of Novel Fluid Biomarkers for Improved Patient Selection and Prediction of Clinical Outcomes in Alzheimer’s Disease Patient Cohorts. Proteomes 2022; 10:proteomes10030026. [PMID: 35997438 PMCID: PMC9397030 DOI: 10.3390/proteomes10030026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/13/2022] [Accepted: 07/23/2022] [Indexed: 01/25/2023] Open
Abstract
Alzheimer’s disease (AD) is an irreversible neurodegenerative disease characterized by progressive cognitive decline. The two cardinal neuropathological hallmarks of AD include the buildup of cerebral β amyloid (Aβ) plaques and neurofibrillary tangles of hyperphosphorylated tau. The current disease-modifying treatments are still not effective enough to lower the rate of cognitive decline. There is an urgent need to identify early detection and disease progression biomarkers that can facilitate AD drug development. The current established readouts based on the expression levels of amyloid beta, tau, and phospho-tau have shown many discrepancies in patient samples when linked to disease progression. There is an urgent need to identify diagnostic and disease progression biomarkers from blood, cerebrospinal fluid (CSF), or other biofluids that can facilitate the early detection of the disease and provide pharmacodynamic readouts for new drugs being tested in clinical trials. Advances in proteomic approaches using state-of-the-art mass spectrometry are now being increasingly applied to study AD disease mechanisms and identify drug targets and novel disease biomarkers. In this report, we describe the application of quantitative proteomic approaches for understanding AD pathophysiology, summarize the current knowledge gained from proteomic investigations of AD, and discuss the development and validation of new predictive and diagnostic disease biomarkers.
Collapse
|
28
|
Chen L, Niu X, Wang Y, Lv S, Zhou X, Yang Z, Peng D. Plasma tau proteins for the diagnosis of mild cognitive impairment and Alzheimer's disease: A systematic review and meta-analysis. Front Aging Neurosci 2022; 14:942629. [PMID: 35959295 PMCID: PMC9358685 DOI: 10.3389/fnagi.2022.942629] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveDetecting plasma tau biomarkers used to be impossible due to their low concentrations in blood samples. Currently, new high-sensitivity assays made it a reality. We performed a systematic review and meta-analysis in order to test the accuracy of plasma tau protein in diagnosing Alzheimer's disease (AD) or mild cognitive impairment (MCI).MethodsWe searched PubMed, Cochrane, Embase and Web of Science databases, and conducted correlation subgroup analysis, sensitivity analysis and publication bias analysis using R Programming Language.ResultsA total of 56 studies were included. Blood t-tau and p-tau levels increased from controls to MCI to AD patients, and showed significant changes in pairwise comparisons of AD, MCI and normal cognition. P-tau217 was more sensitive than p-tau181 and p-tau231 in different cognition periods. In addition, ultrasensitive analytical platforms, immunomagnetic reduction (IMR), increased the diagnostic value of tau proteins, especially the diagnostic value of t-tau.ConclusionBoth t-tau and p-tau are suitable AD blood biomarkers, and p-tau217 is more sensitive than other tau biomarkers to differentiate MCI and AD. Detection techniques also have an impact on biomarkers' results. New ultrasensitive analytical platforms of IMR increase the diagnostic value of both t-tau and p-tau biomarkers.Systematic review registrationhttps://www.crd.york.ac.uk/PROSPERO/, registration number: CRD42021264701.
Collapse
Affiliation(s)
- Leian Chen
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaoqian Niu
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yuye Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuang Lv
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xiao Zhou
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ziyuan Yang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Dantao Peng
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
- *Correspondence: Dantao Peng
| |
Collapse
|
29
|
Tissot C, Therriault J, Kunach P, L Benedet A, Pascoal TA, Ashton NJ, Karikari TK, Servaes S, Lussier FZ, Chamoun M, Tudorascu DL, Stevenson J, Rahmouni N, Poltronetti NM, Pallen V, Bezgin G, Kang MS, Mathotaarachchi SS, Wang YT, Fernandez Arias J, Ferreira PCL, Ferrari-Souza JP, Vanmechelen E, Blennow K, Zetterberg H, Gauthier S, Rosa-Neto P. Comparing tau status determined via plasma pTau181, pTau231 and [ 18F]MK6240 tau-PET. EBioMedicine 2022; 76:103837. [PMID: 35134647 PMCID: PMC8844756 DOI: 10.1016/j.ebiom.2022.103837] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/24/2021] [Accepted: 01/11/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Tau in Alzheimer's disease (AD) is assessed via cerebrospinal fluid (CSF) and Positron emission tomography (PET). Novel methods to detect phosphorylated tau (pTau) in blood have been recently developed. We aim to investigate agreement of tau status as determined by [18F]MK6240 tau-PET, plasma pTau181 and pTau231. METHODS We assessed cognitively unimpaired young, cognitively unimpaired, mild cognitive impairment and AD individuals with [18F]MK6240, plasma pTau181, pTau 231, [18F]AZD4694 amyloid-PET and MRI. A subset underwent CSF assessment. We conducted ROC curves to obtain cut-off values for plasma pTau epitopes. Individuals were categorized as positive or negative in all biomarkers. We then compared the distribution among concordant and discordant groups in relation to diagnosis, Aβ status, APOEε4 status, [18F]AZD4694 global SUVR, hippocampal volume and CSF pTau181. FINDINGS The threshold for positivity was 15.085 pg/mL for plasma pTau181 and 17.652 pg/mL for plasma pTau231. Most individuals had concordant statuses, however, 18% of plasma181/PET, 26% of plasma231/PET and 25% of the pTau231/pTau181 were discordant. Positivity to at least one biomarker was often accompanied by diagnosis of cognitive impairment, Aβ positivity, APOEε4 carriership, higher levels of [18F]AZD4694 global SUVR, hippocampal atrophy and CSF pTau181. INTERPRETATION Plasma pTau181, pTau231 and [18F]MK6240 seem to reflect different stages of tau progression. Plasma biomarkers can be useful in the context of diagnostic information and clinical trials, to evaluate the disease stage. Moreover, they seem to confidently evaluate tau-PET positivity. FUNDING Moreover, this study was supported by Weston Brain Institute, Canadian Institute of Health Research and Fonds de Recherche du Québec.
Collapse
Affiliation(s)
- Cécile Tissot
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Joseph Therriault
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Peter Kunach
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Andréa L Benedet
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Tharick A Pascoal
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada; University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicholas J Ashton
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden; King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sweden; University of Pittsburgh, Pittsburgh, PA, USA
| | - Stijn Servaes
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Firoza Z Lussier
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Mira Chamoun
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | | | - Jenna Stevenson
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Nesrine Rahmouni
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Nina Margherita Poltronetti
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Vanessa Pallen
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Gleb Bezgin
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Min Su Kang
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Sulantha S Mathotaarachchi
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Yi-Ting Wang
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | - Jaime Fernandez Arias
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada
| | | | - João Pedro Ferrari-Souza
- University of Pittsburgh, Pittsburgh, PA, USA; Graduate program in Biological Sciences, Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, United Kingdom
| | - Henrik Zetterberg
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden; UK Dementia Research Institute at UCL, London, United Kingdom; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Serge Gauthier
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Douglas Hospital Research Centre, Verdun, QC, Canada
| | - Pedro Rosa-Neto
- McGill University, Montreal, QC, Canada; McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC room 3149, Verdun, QC H4H 1R3, Canada; Translational Neuroimaging Laboratory, Alzheimer's Disease Research Unit, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, McGill University Research Centre for Studies in Aging, Douglas Research Institute, Montreal, Canada.
| |
Collapse
|
30
|
Mill J, Patel V, Okonkwo O, Li L, Raife T. Erythrocyte sphingolipid species as biomarkers of Alzheimer's disease. J Pharm Anal 2022; 12:178-185. [PMID: 35573876 PMCID: PMC9073235 DOI: 10.1016/j.jpha.2021.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 06/08/2021] [Accepted: 07/13/2021] [Indexed: 01/25/2023] Open
Abstract
Diagnosing Alzheimer's disease (AD) in the early stage is challenging. Informative biomarkers can be of great value for population-based screening. Metabolomics studies have been used to find potential biomarkers, but commonly used tissue sources can be difficult to obtain. The objective of this study was to determine the potential utility of erythrocyte metabolite profiles in screening for AD. Unlike some commonly-used sources such as cerebrospinal fluid and brain tissue, erythrocytes are plentiful and easily accessed. Moreover, erythrocytes are metabolically active, a feature that distinguishes this sample source from other bodily fluids like plasma and urine. In this preliminary pilot study, the erythrocyte metabolomes of 10 histopathologically confirmed AD patients and 10 patients without AD (control (CTRL)) were compared. Whole blood was collected post-mortem and erythrocytes were analyzed using ultra-performance liquid chromatography tandem mass spectrometry. Over 750 metabolites were identified in AD and CTRL erythrocytes. Seven were increased in AD while 24 were decreased (P<0.05). The majority of the metabolites increased in AD were associated with amino acid metabolism and all of the decreased metabolites were associated with lipid metabolism. Prominent among the potential biomarkers were 10 sphingolipid or sphingolipid-related species that were consistently decreased in AD patients. Sphingolipids have been previously implicated in AD and other neurological conditions. Furthermore, previous studies have shown that erythrocyte sphingolipid concentrations vary widely in normal, healthy adults. Together, these observations suggest that certain erythrocyte lipid phenotypes could be markers of risk for development of AD.
Collapse
Affiliation(s)
- Jericha Mill
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Vihar Patel
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Ozioma Okonkwo
- Clinical Science Center, Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Corresponding author. School of Pharmacy and Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Thomas Raife
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
- Corresponding author.
| |
Collapse
|
31
|
Dey KK, Sun H, Wang Z, Niu M, Wang H, Jiao Y, Sun X, Li Y, Peng J. Proteomic Profiling of Cerebrospinal Fluid by 16-Plex TMT-Based Mass Spectrometry. Methods Mol Biol 2022; 2420:21-37. [PMID: 34905163 PMCID: PMC8890903 DOI: 10.1007/978-1-0716-1936-0_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mass spectrometry (MS) has become a mainstream platform for comprehensive profiling of proteome, especially with the improvement of multiplexed tandem mass tag labeling coupled with two-dimensional liquid chromatography and tandem mass spectrometry (TMT-LC/LC-MS/MS). Recently, we have established a robust method for direct profiling of undepleted cerebrospinal fluid (CSF) proteome with the 16-plex TMTpro method, in which we optimized parameters in experimental steps of sample preparation, TMT labeling, LC/LC fractionation, tandem mass spectrometry, and computational data processing. The extensive LC fractionation not only enhances proteome coverage of the CSF but also alleviates ratio distortion of TMT quantification. The crucial quality control steps and improvements specific for the TMT16 analysis are highlighted. More than 3000 proteins can be quantified in a single experiment from 16 different CSF samples. This multiplexed method offers a powerful tool for profiling a variety of complex biofluids samples such as CSF, serum/plasma, and other clinical specimens.
Collapse
|
32
|
Chen YR, Liang CS, Chu H, Voss J, Kang XL, O'Connell G, Jen HJ, Liu D, Shen Hsiao ST, Chou KR. Diagnostic accuracy of blood biomarkers for Alzheimer's disease and amnestic mild cognitive impairment: A meta-analysis. Ageing Res Rev 2021; 71:101446. [PMID: 34391944 DOI: 10.1016/j.arr.2021.101446] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 07/11/2021] [Accepted: 08/09/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To examine the diagnostic accuracy of blood-based biomarkers for detecting Alzheimer's disease (AD) and amnestic mild cognitive impairment (aMCI). METHODS Seven electronic databases were comprehensively searched for studies evaluating the diagnostic accuracy of blood-based biomarkers for detecting AD or aMCI up to July 31, 2020. The pooled sensitivity, specificity, and the diagnostic odds ratio (DOR) were calculated using a hierarchical summary receiver operating characteristic model. RESULTS A total of 17 studies (n = 2,083) were included. In differentiating patients with AD from the controls, the DOR was 32.2 for the plasma Aβ42 (sensitivity = 88 %, specificity = 81 %), 29.1 for the plasma Aβ oligomer (sensitivity = 80 %, specificity = 88 %), and 52.1 for the plasma tau (sensitivity = 90 %, specificity = 87 %). For differentiating aMCI from the controls, the DOR was 60.4 for the plasma Aβ42 (sensitivity = 86 %, specificity = 90 %) and 49.1 for the plasma tau (sensitivity = 79 %, specificity = 94 %). The use of ultra-high sensitive technology explained the heterogeneity in the diagnostic performance of blood-based biomarkers (P = .01). CONCLUSIONS We suggest that blood-based biomarkers are minimally invasive and cost-effective tools for detecting AD; however, the evidence for detecting aMCI was still limited.
Collapse
|
33
|
NAD + supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer's disease via cGAS-STING. Proc Natl Acad Sci U S A 2021; 118:2011226118. [PMID: 34497121 DOI: 10.1073/pnas.2011226118] [Citation(s) in RCA: 297] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2021] [Indexed: 11/18/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive and fatal neurodegenerative disorder. Impaired neuronal bioenergetics and neuroinflammation are thought to play key roles in the progression of AD, but their interplay is not clear. Nicotinamide adenine dinucleotide (NAD+) is an important metabolite in all human cells in which it is pivotal for multiple processes including DNA repair and mitophagy, both of which are impaired in AD neurons. Here, we report that levels of NAD+ are reduced and markers of inflammation increased in the brains of APP/PS1 mutant transgenic mice with beta-amyloid pathology. Treatment of APP/PS1 mutant mice with the NAD+ precursor nicotinamide riboside (NR) for 5 mo increased brain NAD+ levels, reduced expression of proinflammatory cytokines, and decreased activation of microglia and astrocytes. NR treatment also reduced NLRP3 inflammasome expression, DNA damage, apoptosis, and cellular senescence in the AD mouse brains. Activation of cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) are associated with DNA damage and senescence. cGAS-STING elevation was observed in the AD mice and normalized by NR treatment. Cell culture experiments using microglia suggested that the beneficial effects of NR are, in part, through a cGAS-STING-dependent pathway. Levels of ectopic (cytoplasmic) DNA were increased in APP/PS1 mutant mice and human AD fibroblasts and down-regulated by NR. NR treatment induced mitophagy and improved cognitive and synaptic functions in APP/PS1 mutant mice. Our findings suggest a role for NAD+ depletion-mediated activation of cGAS-STING in neuroinflammation and cellular senescence in AD.
Collapse
|
34
|
Wang YY, Sun YP, Luo YM, Peng DH, Li X, Yang BY, Wang QH, Kuang HX. Biomarkers for the Clinical Diagnosis of Alzheimer's Disease: Metabolomics Analysis of Brain Tissue and Blood. Front Pharmacol 2021; 12:700587. [PMID: 34366852 PMCID: PMC8333692 DOI: 10.3389/fphar.2021.700587] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/08/2021] [Indexed: 01/09/2023] Open
Abstract
With an increase in aging populations worldwide, age-related diseases such as Alzheimer's disease (AD) have become a global concern. At present, a cure for neurodegenerative disease is lacking. There is an urgent need for a biomarker that can facilitate the diagnosis, classification, prognosis, and treatment response of AD. The recent emergence of highly sensitive mass-spectrometry platforms and high-throughput technology can be employed to discover and catalog vast datasets of small metabolites, which respond to changed status in the body. Metabolomics analysis provides hope for a better understanding of AD as well as the subsequent identification and analysis of metabolites. Here, we review the state-of-the-art emerging candidate biomarkers for AD.
Collapse
Affiliation(s)
- Yang-Yang Wang
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yan-Ping Sun
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu-Meng Luo
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Dong-Hui Peng
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiao Li
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Bing-You Yang
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiu-Hong Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hai-Xue Kuang
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
35
|
Uddin MS, Kabir MT, Jakaria M, Sobarzo-Sánchez E, Barreto GE, Perveen A, Hafeez A, Bin-Jumah MN, Abdel-Daim MM, Ashraf GM. Exploring the Potential of Neuroproteomics in Alzheimer's Disease. Curr Top Med Chem 2021; 20:2263-2278. [PMID: 32493192 DOI: 10.2174/1568026620666200603112030] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 04/27/2020] [Accepted: 05/08/2020] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is progressive brain amyloidosis that damages brain regions associated with memory, thinking, behavioral and social skills. Neuropathologically, AD is characterized by intraneuronal hyperphosphorylated tau inclusions as neurofibrillary tangles (NFTs), and buildup of extracellular amyloid-beta (Aβ) peptide as senile plaques. Several biomarker tests capturing these pathologies have been developed. However, for the full clinical expression of the neurodegenerative events of AD, there exist other central molecular pathways. In terms of understanding the unidentified underlying processes for the progression and development of AD, a complete comprehension of the structure and composition of atypical aggregation of proteins is essential. Presently, to aid the prognosis, diagnosis, detection, and development of drug targets in AD, neuroproteomics is elected as one of the leading essential tools for the efficient exploratory discovery of prospective biomarker candidates estimated to play a crucial role. Therefore, the aim of this review is to present the role of neuroproteomics to analyze the complexity of AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | | | - Md Jakaria
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Chile,Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Spain
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia,Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ghulam M Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
36
|
Habiba U, Merlin S, Lim JKH, Wong VHY, Nguyen CTO, Morley JW, Bui BV, Tayebi M. Age-Specific Retinal and Cerebral Immunodetection of Amyloid-β Plaques and Oligomers in a Rodent Model of Alzheimer's Disease. J Alzheimers Dis 2021; 76:1135-1150. [PMID: 32597800 DOI: 10.3233/jad-191346] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Amyloid-β soluble oligomers (Aβo) are believed to be the cause of the pathophysiology underlying Alzheimer's disease (AD) and are normally detected some two decades before clinical onset of the disease. Retinal pathology associated with AD pathogenesis has previously been reported, including ganglion cell loss, accumulation of Aβ deposits in the retina, and reduction of nerve fiber layer thickness as well as abnormalities of the microvasculature. OBJECTIVE This study's aim is to better understand the relationship between brain and retinal Aβo deposition and in particular to quantify levels of the toxic Aβo as a function of age in the retina of a rodent model of AD. METHODS Retinas and brain tissue from 5×FAD mice were stained with Congo red, Thioflavin-T (Th-T), and Aβ plaque-specific and Aβo-specific antibodies. RESULTS We show that retinas displayed an age-dependent increase of Th-T-specific amyloid fibrils. Staining with anti-Aβ antibody confirmed the presence of the Aβ plaques in all 5×FAD retinas tested. In contrast, staining with anti-Aβo antibody showed an age-dependent decrease of retinal Aβo. Of note, Aβo was observed mainly in the retinal nuclear layers. Finally, we confirmed the localization of Aβo to neurons, typically accumulating in late endosomes, indicating possible impairment of the endocytic pathway. CONCLUSION Our results demonstrate the presence of intraneuronal Aβo in the retina and its accumulation inversely correlated with retinal Aβ plaque deposition, indicating an age-related conversion in this animal model. These results support the development of an early AD diagnostic test targeting Aβo in the eye.
Collapse
Affiliation(s)
- Umma Habiba
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Sam Merlin
- School of Science & Health, Western Sydney University, Campbelltown, NSW, Australia
| | - Jeremiah K H Lim
- Department of Optometry and Vision Sciences, University of Melbourne, Victoria, Australia
| | - Vickie H Y Wong
- Department of Optometry and Vision Sciences, University of Melbourne, Victoria, Australia
| | - Christine T O Nguyen
- Department of Optometry and Vision Sciences, University of Melbourne, Victoria, Australia
| | - John W Morley
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Victoria, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
37
|
Goldwaser EL, Acharya NK, Wu H, Godsey GA, Sarkar A, DeMarshall CA, Kosciuk MC, Nagele RG. Evidence that Brain-Reactive Autoantibodies Contribute to Chronic Neuronal Internalization of Exogenous Amyloid-β1-42 and Key Cell Surface Proteins During Alzheimer's Disease Pathogenesis. J Alzheimers Dis 2021; 74:345-361. [PMID: 32039847 PMCID: PMC7175946 DOI: 10.3233/jad-190962] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Blood-brain barrier (BBB) permeability is a recognized early feature of Alzheimer’s disease (AD). In the present study, we examined consequences of increased BBB permeability on the development of AD-related pathology by tracking selected leaked plasma components and their interactions with neurons in vivo and in vitro. Histological sections of cortical regions of postmortem AD brains were immunostained to determine the distribution of amyloid-β1-42 (Aβ42), cathepsin D, IgG, GluR2/3, and alpha7 nicotinic acetylcholine receptor (α7nAChR). Results revealed that chronic IgG binding to pyramidal neurons coincided with internalization of Aβ42, IgG, GluR2/3, and α7nAChR as well as lysosomal compartment expansion in these cells in regions of AD pathology. To test possible mechanistic interrelationships of these phenomena, we exposed differentiated SH-SY5Y neuroblastoma cells to exogenous, soluble Aβ42 peptide and serum from AD and control subjects. The rate and extent of Aβ42 internalization in these cells was enhanced by serum containing neuron-binding IgG autoantibodies. This was confirmed by treating cells with individual antibodies specific for α7nAChR, purified IgG from AD or non-AD sera, and sera devoid of IgG, in the presence of 100 nM Aβ42. Initial co-localization of IgG, α7nAChR, and Aβ42 was temporally and spatially linked to early endosomes (Rab11) and later to lysosomes (LAMP-1). Aβ42 internalization was attenuated by treatment with monovalent F(ab) antibody fragments generated from purified IgG from AD serum and then rescued by coupling F(ab) fragments with divalent human anti-Fab. Overall, results suggest that cross-linking of neuron-binding autoantibodies targeting cell surface proteins can accelerate intraneuronal Aβ42 deposition in AD.
Collapse
Affiliation(s)
- Eric L Goldwaser
- University of Maryland Medical Center and Sheppard Pratt Health System, Department of Psychiatry, Baltimore, MD, USA.,Biomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA.,Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, USA
| | - Nimish K Acharya
- Biomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA.,Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, USA.,Department of Geriatrics and Gerontology, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - Hao Wu
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, USA
| | - George A Godsey
- Biomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA.,Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, USA
| | - Abhirup Sarkar
- Biomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA.,Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, USA
| | - Cassandra A DeMarshall
- Biomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA.,Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, USA
| | - Mary C Kosciuk
- Biomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA.,Department of Geriatrics and Gerontology, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - Robert G Nagele
- Biomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA.,Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, USA.,Department of Geriatrics and Gerontology, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| |
Collapse
|
38
|
Habiba U, Descallar J, Kreilaus F, Adhikari UK, Kumar S, Morley JW, Bui BV, Hamaoui MK, Tayebi M. Detection of retinal and blood Aβ oligomers with nanobodies. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2021; 13:e12193. [PMID: 33977118 PMCID: PMC8101010 DOI: 10.1002/dad2.12193] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022]
Abstract
Introduction Abnormal retinal changes are increasingly recognized as an early pathological change in Alzheimer's disease (AD). Although amyloid beta oligomers (Aβo) have been shown to accumulate in the blood and retina of AD patients and animals, it is not known whether the early Aβo deposition precedes their accumulation in brain. Methods and results Using nanobodies targeting Aβ1‐40 and Aβ1‐42 oligomers we were able to detect Aβ oligomers in the retina and blood but not in the brain of 3‐month‐old APP/PS1 mice. Furthermore, Aβ plaques were detected in the brain but not the retina of 3‐month‐old APP/PS1 mice. Conclusion These results suggest that retinal accumulation of Aβo originates from peripheral blood and precedes cognitive decline and Aβo deposition in the brain. This provides a very strong basis to develop and implement an “eye test” for early detection of AD using nanobodies targeting retinal Aβ.
Collapse
Affiliation(s)
- Umma Habiba
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| | - Joseph Descallar
- South Western Sydney Clinical School, Faculty of Medicine UNSW, Liverpool Hospital Liverpool New South Wales Australia.,Ingham Institute of Applied Medical Research Liverpool New South Wales Australia
| | - Fabian Kreilaus
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| | - Utpal K Adhikari
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| | - Sachin Kumar
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| | - John W Morley
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences University of Melbourne Melbourne Victoria Australia
| | - Maya K Hamaoui
- Department of Neurosurgery Maxine Dunitz Neurosurgical Research Institute and Department of Biomedical Sciences Cedars-Sinai Medical Center Los Angeles California USA
| | - Mourad Tayebi
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| |
Collapse
|
39
|
Hadjidemetriou M, Rivers-Auty J, Papafilippou L, Eales J, Kellett KAB, Hooper NM, Lawrence CB, Kostarelos K. Nanoparticle-Enabled Enrichment of Longitudinal Blood Proteomic Fingerprints in Alzheimer's Disease. ACS NANO 2021; 15:7357-7369. [PMID: 33730479 PMCID: PMC8155389 DOI: 10.1021/acsnano.1c00658] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Blood-circulating biomarkers have the potential to detect Alzheimer's disease (AD) pathology before clinical symptoms emerge and to improve the outcomes of clinical trials for disease-modifying therapies. Despite recent advances in understanding concomitant systemic abnormalities, there are currently no validated or clinically used blood-based biomarkers for AD. The extremely low concentration of neurodegeneration-associated proteins in blood necessitates the development of analytical platforms to address the "signal-to-noise" issue and to allow an in-depth analysis of the plasma proteome. Here, we aimed to discover and longitudinally track alterations of the blood proteome in a transgenic mouse model of AD, using a nanoparticle-based proteomics enrichment approach. We employed blood-circulating, lipid-based nanoparticles to extract, analyze and monitor AD-specific protein signatures and to systemically uncover molecular pathways associated with AD progression. Our data revealed the existence of multiple proteomic signals in blood, indicative of the asymptomatic stages of AD. Comprehensive analysis of the nanoparticle-recovered blood proteome by label-free liquid chromatography-tandem mass spectrometry resulted in the discovery of AD-monitoring signatures that could discriminate the asymptomatic phase from amyloidopathy and cognitive deterioration. While the majority of differentially abundant plasma proteins were found to be upregulated at the initial asymptomatic stages, the abundance of these molecules was significantly reduced as a result of amyloidosis, suggesting a disease-stage-dependent fluctuation of the AD-specific blood proteome. The potential use of the proposed nano-omics approach to uncover information in the blood that is directly associated with brain neurodegeneration was further exemplified by the recovery of focal adhesion cascade proteins. We herein propose the integration of nanotechnology with already existing proteomic analytical tools in order to enrich the identification of blood-circulating signals of neurodegeneration, reinvigorating the potential clinical utility of the blood proteome at predicting the onset and kinetics of the AD progression trajectory.
Collapse
Affiliation(s)
- Marilena Hadjidemetriou
- Nanomedicine
Lab, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom
- (M.H.)
| | - Jack Rivers-Auty
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, United Kingdom
| | - Lana Papafilippou
- Nanomedicine
Lab, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom
| | - James Eales
- Division
of Cardiovascular Sciences, School of Medical Sciences, Faculty of
Biology, Medicine and Health, The University
of Manchester M13 9PT, Manchester, United Kingdom
| | - Katherine A. B. Kellett
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, United Kingdom
| | - Nigel M. Hooper
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, United Kingdom
| | - Catherine B. Lawrence
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, United Kingdom
| | - Kostas Kostarelos
- Nanomedicine
Lab, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom
- (K.K.)
| |
Collapse
|
40
|
Tzekaki EE, Tsolaki M, Pantazaki ΑA, Geromichalos G, Lazarou E, Kozori M, Sinakos Z. The pleiotropic beneficial intervention of olive oil intake on the Alzheimer's disease onset via fibrinolytic system. Exp Gerontol 2021; 150:111344. [PMID: 33836262 DOI: 10.1016/j.exger.2021.111344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 12/31/2022]
Abstract
The daily consumption of Extra Virgin Olive Oil (EVOO) in Mediterranean nutrition is tightly associated with lower frequency of many diseases' appearance, including Alzheimer's disease (AD). Fibrinolytic system is already assumed to be involved in AD pathophysiology through various factors, especially plasminogen activator inhibitor-1 (PAI-1), a2-antiplasmin (α2ΑP) and tissue plasminogen activator (tPA). We, here, present a biochemical study, as a continuation of a clinical trial of a cohort of 84 participants, focusing on the pleiotropic effect of the annual EVOO consumption on the fibrinolytic factors of Mild Cognitive Impairment (MCI) patients. The levels of all these fibrinolytic factors, measured by Enzyme-Linked Immunosorbent Assay (ELISA) method, were reduced in the serum of MCI patients annually administered with EVOO, versus not treated MCI patients, as well as AD patients. The well-established AD hallmarks (Aβ1-40 and Aβ1-42 species, tau, and p-tau) of MCI patients' group, annually administered with EVOO, were restored to levels equal to those of the cognitively-healthy group; in contrast to those patients not being administered, and their AD hallmarks levels increased at the end of the year. Moreover, one of the EVOO annual consumption multimodal effects on the MCI patients focused on the levels of an oxidative stress trademark, malondialdehyde (MDA), which displayed also a visible quenching; On the other hand, an increase exhibited in the MCI patients not consuming EVOO one year after, was attributed to the lack of the EVOO anti-oxidative properties. These outcomes are exploitable towards the establishment of natural products like EVOO, as a preventive remedy fighting this neurodegenerative disorder, AD. CLINICAL TRIAL REGISTRATION: https://clinicaltrials.gov/ct2/show/NCT03362996 MICOIL gov Identifier: NCT03362996.
Collapse
Affiliation(s)
- Elena E Tzekaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Makedonia, Greece
| | - Magda Tsolaki
- 1(st) Department of Neurology, Medical School, "AHEPA" General Hospital Medical School, Aristotle University of Thessaloniki, Faculty of Health Sciences, 54124 Thessaloniki, Makedonia, Greece; Greek Association of Alzheimer's Disease and Related Disorders - GAADRD, Greece.
| | - Αnastasia A Pantazaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Makedonia, Greece.
| | - George Geromichalos
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Makedonia, Greece
| | - Eftychia Lazarou
- Greek Association of Alzheimer's Disease and Related Disorders - GAADRD, Greece
| | - Mahi Kozori
- Greek Association of Alzheimer's Disease and Related Disorders - GAADRD, Greece
| | - Zacharias Sinakos
- Emeritus Professor of Hematology, Medical School, Aristotle University of Thessaloniki, Faculty of Health Sciences, Greece
| |
Collapse
|
41
|
San-Martin R, Johns E, Quispe Mamani G, Tavares G, Phillips NA, Fraga FJ. A method for diagnosis support of mild cognitive impairment through EEG rhythms source location during working memory tasks. Biomed Signal Process Control 2021. [DOI: 10.1016/j.bspc.2021.102499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
42
|
Liu D, Fu D, Zhang L, Sun L. Detection of amyloid-beta by Fmoc-KLVFF self-assembled fluorescent nanoparticles for Alzheimer’s disease diagnosis. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2020.09.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
43
|
Plasma osteopontin as a biomarker of Alzheimer's disease and vascular cognitive impairment. Sci Rep 2021; 11:4010. [PMID: 33597603 PMCID: PMC7889621 DOI: 10.1038/s41598-021-83601-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/21/2021] [Indexed: 12/22/2022] Open
Abstract
Cerebrovascular disease (CeVD) and neurodegenerative dementia such as Alzheimer’s disease (AD) are frequently associated comorbidities in the elderly, sharing common risk factors and pathophysiological mechanisms including neuroinflammation. Osteopontin (OPN) is an inflammatory marker found upregulated in vascular diseases as well as in AD. However, its involvement in vascular dementia (VaD) and pre-dementia stages, namely cognitive impairment no dementia (CIND), both of which fall under the spectrum of vascular cognitive impairment (VCI), has yet to be examined. Its correlations with inflammatory cytokines in cognitive impairment also await investigation. 80 subjects with no cognitive impairment (NCI), 160 with CIND and 144 with dementia were included in a cross-sectional study on a Singapore-based memory clinic cohort. All subjects underwent comprehensive clinical, neuropsychological and brain neuroimaging assessments, together with clinical diagnoses based on established criteria. Blood samples were collected and OPN as well as inflammatory cytokines interleukin (IL)-6, IL-8 and tumor necrosis factor (TNF) were measured using immunoassays. Multivariate regression analyses showed significant associations between increased OPN and VCI groups, namely CIND with CeVD, AD with CeVD and VaD. Interestingly, higher OPN was also significantly associated with AD even in the absence of CeVD. We further showed that increased OPN significantly associated with neuroimaging markers of CeVD and neurodegeneration, including cortical infarcts, lacunes, white matter hyperintensities and brain atrophy. OPN also correlated with elevated levels of IL-6, IL-8 and TNF. Our findings suggest that OPN may play a role in both VCI and neurodegenerative dementias. Further longitudinal analyses are needed to assess the prognostic utility of OPN in disease prediction and monitoring.
Collapse
|
44
|
Deniz K, Ho CCG, Malphrus KG, Reddy JS, Nguyen T, Carnwath TP, Crook JE, Lucas JA, Graff-Radford NR, Carrasquillo MM, Ertekin-Taner N. Plasma Biomarkers of Alzheimer's Disease in African Americans. J Alzheimers Dis 2021; 79:323-334. [PMID: 33252078 PMCID: PMC7902984 DOI: 10.3233/jad-200828] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background/Objective: The aim of this study was to determine if plasma concentrations of 5 surrogate markers of Alzheimer’s disease (AD) pathology and neuroinflammation are associated with disease status in African Americans. Methods: We evaluated 321 African Americans (159 AD, 162 controls) from the Florida Consortium for African-American Alzheimer’s Disease Studies (FCA3DS). Five plasma proteins reflecting AD neuropathology or inflammation (Aβ42, tau, IL6, IL10, TNFα) were tested for associations with AD, age, sex, APOE and MAPT genotypes, and for pairwise correlations. Results: Plasma tau levels were higher in AD when adjusted for biological and technical covariates. APOEɛ4 was associated with lower plasma Aβ42 and tau levels. Older age was associated with higher plasma Aβ42, tau, and TNFα. Females had lower IL10 levels. Inflammatory proteins had strong pairwise correlations amongst themselves and with Aβ42. Conclusion: We identified effects of demographic and genetic variants on five potential plasma biomarkers in African Americans. Plasma inflammatory biomarkers and Aβ42 may reflect correlated pathologies and elevated plasma tau may be a biomarker of AD in this population.
Collapse
Affiliation(s)
- Kaancan Deniz
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL, USA
| | | | | | - Joseph S Reddy
- Mayo Clinic, Department of Health Science Research, Jacksonville, FL, USA
| | - Thuy Nguyen
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL, USA
| | - Troy P Carnwath
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL, USA
| | - Julia E Crook
- Mayo Clinic, Department of Health Science Research, Jacksonville, FL, USA
| | - John A Lucas
- Mayo Clinic, Department of Psychiatry and Psychology, Jacksonville, FL, USA
| | | | | | - Nilüfer Ertekin-Taner
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL, USA.,Mayo Clinic, Department of Neurology, Jacksonville, FL, USA
| |
Collapse
|
45
|
Snyder PJ, Alber J, Alt C, Bain LJ, Bouma BE, Bouwman FH, DeBuc DC, Campbell MC, Carrillo MC, Chew EY, Cordeiro MF, Dueñas MR, Fernández BM, Koronyo-Hamaoui M, La Morgia C, Carare RO, Sadda SR, van Wijngaarden P, Snyder HM. Retinal imaging in Alzheimer's and neurodegenerative diseases. Alzheimers Dement 2021; 17:103-111. [PMID: 33090722 PMCID: PMC8062064 DOI: 10.1002/alz.12179] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 12/22/2022]
Abstract
In the last 20 years, research focused on developing retinal imaging as a source of potential biomarkers for Alzheimer's disease and other neurodegenerative diseases, has increased significantly. The Alzheimer's Association and the Alzheimer's & Dementia: Diagnosis, Assessment, Disease Monitoring editorial team (companion journal to Alzheimer's & Dementia) convened an interdisciplinary discussion in 2019 to identify a path to expedite the development of retinal biomarkers capable of identifying biological changes associated with AD, and for tracking progression of disease severity over time. As different retinal imaging modalities provide different types of structural and/or functional information, the discussion reflected on these modalities and their respective strengths and weaknesses. Discussion further focused on the importance of defining the context of use to help guide the development of retinal biomarkers. Moving from research to context of use, and ultimately to clinical evaluation, this article outlines ongoing retinal imaging research today in Alzheimer's and other brain diseases, including a discussion of future directions for this area of study.
Collapse
Affiliation(s)
- Peter J. Snyder
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| | - Jessica Alber
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| | - Clemens Alt
- Wellman Center for Photomedicine and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lisa J. Bain
- Independent Science Writer, Elverson, Pennsylvania
| | - Brett E. Bouma
- Harvard Medical School, Massachusetts General Hospital and Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Massachusetts
| | - Femke H. Bouwman
- Neurologist, Alzheimer Center Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Melanie C.W. Campbell
- Physics and Astronomy, Optometry and Vision Science and Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Maria C. Carrillo
- Medical & Scientific Relations, Alzheimer’s Association, Chicago, Illinois
| | - Emily Y. Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - M. Francesca Cordeiro
- Imperial College London, UCL Institute of Ophthalmology, ICORG Western Eye Hospital, London, UK
| | - Michael R. Dueñas
- Chief Public Health Officer (Ret.), American Optometric Association, Washington, D.C
| | | | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute and Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Chiara La Morgia
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie, University of Bologna, Italy
| | | | - Srinivas R. Sadda
- Doheny Eye Institute, Los Angeles, California
- Department of Ophthalmology, UCLA, Los Angeles, California
| | - Peter van Wijngaarden
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Parkville, Australia
| | - Heather M. Snyder
- Medical & Scientific Relations, Alzheimer’s Association, Chicago, Illinois
| |
Collapse
|
46
|
Guo Z, Peng X, Li HY, Wang Y, Qian Y, Wang Z, Ye D, Ji X, Wang Z, Wang Y, Chen D, Lei H. Evaluation of Peripheral Immune Dysregulation in Alzheimer's Disease and Vascular Dementia. J Alzheimers Dis 2020; 71:1175-1186. [PMID: 31498124 DOI: 10.3233/jad-190666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Immune dysregulation has been observed in the brain and blood of patients with Alzheimer's disease (AD). However, a convenient assay to evaluate peripheral immune dysregulation in AD has not been developed, partly due to the inconsistent observations from different studies. We hypothesized that peripheral immune dysregulation may only exist in a subpopulation of AD patients; therefore it may be valuable to identify this subpopulation with a convenient assay. Along this line, we selected 14 candidate genes based on our analysis of microarray data on peripheral blood of AD and other diseases. We used RT-qPCR to examine the expression of these 14 genes in a cohort of 288 subjects, including 74 patients with AD, 64 patients with mild cognitive impairment (MCI), 51 patients with vascular dementia (VaD), and 99 elderly controls with no cognitive dysfunction/impairment. Seven of these 14 genes displayed significant difference in group comparison. Switching from group comparison to individualized evaluation revealed more in-depth information. First, there existed a wide dynamic range for the expression of these immune genes in peripheral blood even within the control group. Second, for the vast majority of the patients (AD, VaD, and MCI patients), the expression of these genes fell within the dynamic range of the control group. Third, a small portion of outliers were observed in the patient groups, more so in the VaD group than that in the AD or MCI groups. This is our first attempt to conduct personalized evaluation of peripheral immune dysregulation in AD and VaD. These findings may be applicable to the identification of peripheral immune dysregulation in AD and VaD patients which may lead to tailored treatment toward those patients.
Collapse
Affiliation(s)
- Zongjun Guo
- Department of Geriatrics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xing Peng
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,Cunji Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Hui-Yun Li
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yunlai Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Ying Qian
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,Cunji Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Zhihong Wang
- Department of Geriatrics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dongqing Ye
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Xiaoyun Ji
- Department of Geriatrics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhixin Wang
- Qingdao Chengyang People's Hospital, Qingdao, China
| | - Yanjiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Dongwan Chen
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Hongxing Lei
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,Cunji Medical School, University of Chinese Academy of Sciences, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
47
|
Szegedi S, Dal‐Bianco P, Stögmann E, Traub‐Weidinger T, Rainer M, Masching A, Schmidl D, Werkmeister RM, Chua J, Schmetterer L, Garhöfer G. Anatomical and functional changes in the retina in patients with Alzheimer's disease and mild cognitive impairment. Acta Ophthalmol 2020; 98:e914-e921. [PMID: 32212415 PMCID: PMC7687124 DOI: 10.1111/aos.14419] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/04/2020] [Indexed: 01/14/2023]
Abstract
PURPOSE There is evidence that mild cognitive impairment (MCI) or Alzheimer's disease (AD) is accompanied by alterations in the retina. The current study was performed to investigate structural and functional changes in patients with systemic neurodegenerative disease. METHODS A total of 47 patients with either MCI or AD and 43 healthy age- and sex-matched control subjects were included. Inclusion criteria for MCI were abnormal memory function and a mini-mental state examination (MMSE) score >26 points, for patients with AD a diagnosis of probable AD of mild to moderate degree and an MMSE score in the range of 20-26. Retinal blood flow was measured using a Doppler optical coherence tomography (OCT) system. Retinal vessel diameter, oxygen saturation and flicker-induced vasodilatation were measured using a Vessel Analyzer. Retinal nerve fibre layer thickness (RNFLT) was assessed using an OCT system. RESULTS Global RNFLT was lower in patients compared to healthy controls (93.7 ± 12.8 µm versus 99.1 ± 9.0 µm, p = 0.02). The same was found in regards to retinal arterial blood flow, which was 9.3 ± 2.4 and 12.3 ± 3.2 μl/min in the patient and control groups, respectively (p < 0.001). Mean retinal arterial diameter was reduced in patients (76.0 ± 8.9 µm versus 80.6 ± 8.0 µm, p = 0.03). Arteriovenous difference in oxygen saturation was lower in patients (20.4 ± 5.1% versus 23.5 ± 4.0%, p < 0.01). No difference in the flicker response was observed. CONCLUSION In patients with MCI and AD, arteriovenous difference in oxygen saturation, retinal blood flow and arterial vessel diameter was reduced. No difference was found in flicker response between groups. This indicates alterations in retinal oxygen metabolism in patients with neurodegenerative disease.
Collapse
Affiliation(s)
- Stephan Szegedi
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
| | - Peter Dal‐Bianco
- Department of NeurologyMedical University of ViennaViennaAustria
| | | | - Tatjana Traub‐Weidinger
- Division of Nuclear MedicineDepartment of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Michael Rainer
- Department of PsychiatrySocial and Medical Centre East – Danube HospitalViennaAustria
- Karl Landsteiner Institute for Memory and Alzheimer ResearchViennaAustria
| | - Andreas Masching
- Department of PsychiatrySocial and Medical Centre East – Danube HospitalViennaAustria
| | - Doreen Schmidl
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
| | - René M. Werkmeister
- Center for Medical Physics and Biomedical EngineeringMedical University of ViennaViennaAustria
| | - Jacqueline Chua
- Singapore Eye Research InstituteSingapore National Eye CentreSingapore CitySingapore
- Ophthalmology and Visual Sciences Academic Clinical ProgramDuke‐NUS Medical SchoolSingapore CitySingapore
| | - Leopold Schmetterer
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
- Center for Medical Physics and Biomedical EngineeringMedical University of ViennaViennaAustria
- Singapore Eye Research InstituteSingapore National Eye CentreSingapore CitySingapore
- Ophthalmology and Visual Sciences Academic Clinical ProgramDuke‐NUS Medical SchoolSingapore CitySingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore CitySingapore
- Institute of Molecular and Clinical OphthalmologyBaselSwitzerland
| | - Gerhard Garhöfer
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
| |
Collapse
|
48
|
Zhu Y, Gong L, He C, Wang Q, Ren Q, Xie C. Default Mode Network Connectivity Moderates the Relationship Between the APOE Genotype and Cognition and Individualizes Identification Across the Alzheimer's Disease Spectrum. J Alzheimers Dis 2020; 70:843-860. [PMID: 31282419 DOI: 10.3233/jad-190254] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although previous studies have investigated the effects of the apolipoprotein E (APOE) ɛ4 genotype on the default mode network (DMN) in the Alzheimer's disease (AD) spectrum, it is still unclear how the APOE genotype regulates the DMN and subsequently affects cognitive decline in the AD spectrum. One hundred sixty-nine subjects with resting-state functional magnetic resonance imaging data and neuropsychological test scores were selected from the Alzheimer's Disease Neuroimaging Initiative. The main effects and interaction of the APOE genotype and disease status on the DMN were explored. A moderation analysis was performed to investigate the relationship among the APOE genotype, DMN connectivity, and cognition. Additionally, the pair-wised DMN connectivity was used to classify AD spectrum, and the classification accuracy was validated. Compared to APOEɛ4 non-carriers, APOEɛ4 carriers showed the opposite trajectory of DMN connectivity across the AD spectrum. Specifically, the strengths in the posterior cingulate cortex (PCC) connecting with the right precuneus, insular, and fusiform area (FFA) were positively correlated with Mini-Mental State Examination (MMSE) scores in APOEɛ4 non-carriers but not in APOEɛ4 carriers. Furthermore, PCC-right FFA connectivity could moderate the effects of the APOE genotype on MMSE scores across the disease groups. More importantly, using a receiver operating characteristic analysis, these altered connectivities yielded strong classification powers in a pathological stage-dependent manner in the AD spectrum. These findings first identified the intrinsic DMN connectivity moderating the effect of the APOE genotype on cognition and provided a pathological stage-dependent neuroimaging biomarker for early differentiation of the AD spectrum.
Collapse
Affiliation(s)
- Yao Zhu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Liang Gong
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Cancan He
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Qing Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Qingguo Ren
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Chunming Xie
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | | |
Collapse
|
49
|
Chua XY, Chai YL, Chew WS, Chong JR, Ang HL, Xiang P, Camara K, Howell AR, Torta F, Wenk MR, Hilal S, Venketasubramanian N, Chen CP, Herr DR, Lai MKP. Immunomodulatory sphingosine-1-phosphates as plasma biomarkers of Alzheimer's disease and vascular cognitive impairment. Alzheimers Res Ther 2020; 12:122. [PMID: 32998767 PMCID: PMC7528375 DOI: 10.1186/s13195-020-00694-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND There has been ongoing research impetus to uncover novel blood-based diagnostic and prognostic biomarkers for Alzheimer's disease (AD), vascular dementia (VaD), and related cerebrovascular disease (CEVD)-associated conditions within the spectrum of vascular cognitive impairment (VCI). Sphingosine-1-phosphates (S1Ps) are signaling lipids which act on the S1PR family of cognate G-protein-coupled receptors and have been shown to modulate neuroinflammation, a process known to be involved in both neurodegenerative and cerebrovascular diseases. However, the status of peripheral S1P in AD and VCI is at present unclear. METHODS We obtained baseline bloods from individuals recruited into an ongoing longitudinal cohort study who had normal cognition (N = 80); cognitive impairment, no dementia (N = 160); AD (N = 113); or VaD (N = 31), along with neuroimaging assessments of cerebrovascular diseases. Plasma samples were processed for the measurements of major S1P species: d16:1, d17:1, d18:0, and d18:1, along with pro-inflammatory cytokines interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF). Furthermore, in vitro effects of S1Ps on cytokine expression were also studied in an astrocytoma cell line and in rodent primary astrocytes. RESULTS Of the S1Ps species measured, only d16:1 S1P was significantly reduced in the plasma of VaD, but not AD, patients, while the d18:1 to d16:1 ratios were increased in all cognitive subgroups (CIND, AD, and VaD). Furthermore, d18:1 to d16:1 ratios correlated with levels of IL-6, IL-8, and TNF. In both primary astrocytes and an astroglial cell line, treatment with d16:1 or d18:1 S1P resulted in the upregulation of mRNA transcripts of pro-inflammatory cytokines, with d18:1 showing a stronger effect than d16:1. Interestingly, co-treatment assays showed that the addition of d16:1 reduced the extent of d18:1-mediated gene expression, indicating that d16:1 may function to "fine-tune" the pro-inflammatory effects of d18:1. CONCLUSION Taken together, our data suggest that plasma d16:1 S1P may be useful as a diagnostic marker for VCI, while the d18:1 to d16:1 S1P ratio is an index of dysregulated S1P-mediated immunomodulation leading to chronic inflammation-associated neurodegeneration and cerebrovascular damage.
Collapse
Affiliation(s)
- Xin Ying Chua
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, 117597, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, 117597, Singapore
- Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
| | - Wee Siong Chew
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, 117597, Singapore
| | - Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, 117597, Singapore
- Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, 117597, Singapore
- Cancer Science Institute, National University of Singapore, Kent Ridge, Singapore
| | - Ping Xiang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, 117597, Singapore
| | - Kaddy Camara
- Department of Chemistry, University of Connecticut, Storrs, CT, USA
| | - Amy R Howell
- Department of Chemistry, University of Connecticut, Storrs, CT, USA
| | - Federico Torta
- Singapore Lipidomics Incubator (SLING), Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Markus R Wenk
- Singapore Lipidomics Incubator (SLING), Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Saima Hilal
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, 117597, Singapore
- Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Kent Ridge, Singapore
| | | | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, 117597, Singapore
- Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, 117597, Singapore.
- Department of Biology, San Diego State University, San Diego, CA, USA.
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, 117597, Singapore.
- Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore.
| |
Collapse
|
50
|
McDade E, Bednar MM, Brashear HR, Miller DS, Maruff P, Randolph C, Ismail Z, Carrillo MC, Weber CJ, Bain LJ, Hake AM. The pathway to secondary prevention of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12069. [PMID: 32885024 PMCID: PMC7453146 DOI: 10.1002/trc2.12069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/09/2020] [Indexed: 11/11/2022]
Abstract
Alzheimer's disease (AD) is a continuum consisting of a preclinical stage that occurs decades before symptoms appear. As researchers make advances in investigating the continuum, the importance of developing drugs for secondary prevention is garnering increased discussion. For efficacious drug development for secondary prevention it is important to define what are the earliest biological stages of AD. The Alzheimer's Association Research Roundtable convened November 27 to 28, 2018 to focus on pre-clinical AD. This review will address the biological approach to defining pre-clinical AD, detection, identification of at-risk individuals, and lessons learned from trials such as A4 and TOMMORROW.
Collapse
Affiliation(s)
- Eric McDade
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Martin M. Bednar
- Takeda Pharmaceuticals International Co.Americas, Inc.CambridgeMassachusettsUSA
| | | | | | | | - Christopher Randolph
- MedAvante‐ProPhaseHamiltonNew JerseyUSA
- Department of NeurologyLoyola University Medical CenterMaywoodIllinoisUSA
| | - Zahinoor Ismail
- Cumming School of MedicineThe University of CalgaryCalgaryCanada
| | | | | | - Lisa J. Bain
- Independent Science WriterElversonPennsylvaniaUSA
| | - Ann Marie Hake
- Eli Lilly and CompanyIndianapolisIndianaUSA
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|