1
|
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
|
2
|
Zhuang T, Yang Y, Ren H, Zhang H, Gao C, Chen S, Shen J, Ji M, Cui Y. Novel plasma protein biomarkers: A time-dependent predictive model for Alzheimer's disease. Arch Gerontol Geriatr 2024; 129:105650. [PMID: 39427525 DOI: 10.1016/j.archger.2024.105650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND The accurate prediction of Alzheimer's disease (AD) is crucial for the efficient management of its progression. The objective of this research was to construct a new risk predictive model utilizing novel plasma protein biomarkers for predicting AD incidence in the future and analyze their potential biological correlation with AD incidence. METHODS A cohort of 440 participants aged 60 years and older from the Alzheimer's Disease Neuroimaging Initiative (ADNI) longitudinal cohort was utilized. The baseline plasma proteomics data was employed to conduct Cox regression, LASSO regression, and cross-validation to identify plasma protein signatures predictive of AD risk. Subsequently, a multivariable Cox proportional hazards model based on these signatures was constructed. The performance of the risk prediction model was evaluated using time-dependent receiver operating characteristic (t-ROC) curves and Kaplan-Meier curves. Additionally, we analyzed the correlations between protein signature expression in plasma and predicted AD risk, the time of AD onset, the expression of protein signatures in cerebrospinal fluid (CSF), the expression of CSF and plasma biomarkers, and APOE ε4 genotypes. Colocalization and Mendelian randomization analyses was conducted to investigate the association between protein features and AD risk. GEO database was utilized to analyze the differential expression of protein features in the blood and brain of AD patients. RESULTS We identified seven protein signatures (APOE, CGA, CRP, CCL26, CCL20, NRCAM, and PYY) that independently predicted AD incidence in the future. The risk prediction model demonstrated area under the ROC curve (AUC) values of 0.77, 0.76, and 0.77 for predicting AD incidence at 4, 6, and 8 years, respectively. Furthermore, the model remained stable in the range of the 3rd to the 12th year (ROC ≥ 0.74). The low-risk group, as defined by the model, exhibited a significantly later AD onset compared to the high-risk group (P < 0.0001). Moreover, all protein signatures exhibited significant correlations with AD risk (P < 0.001) and the time of AD onset (P < 0.01). There was no strong correlation between the protein expression levels in plasma and CSF, as well as AD CSF biomarkers. APOE, CGA, and CRP exhibited significantly lower expression levels in APOE ε4 positive individuals (P < 0.05). Additionally, colocalization analysis reveals a significant association between AD and SNP loci in APOE. Mendelian randomization analysis shows a negative correlation between NRCAM and AD risk. Transcriptomic analysis indicates a significant downregulation of NRCAM and PYY in the peripheral blood of AD patients (P < 0.01), while APOE, CGA, and NRCAM are significantly downregulated in the brains of AD patients (P < 0.0001). CONCLUSION Our research has successfully identified protein signatures in plasma as potential risk biomarkers that can independently predict AD onset in the future. Notably, this risk prediction model has demonstrated commendable predictive performance and stability over time. These findings underscore the promising utility of plasma protein signatures in dynamically predicting the risk of AD, thereby facilitating early screening and intervention strategies.
Collapse
Affiliation(s)
- Tianchi Zhuang
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Yingqi Yang
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Haili Ren
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Haoxiang Zhang
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Chang Gao
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Shen Chen
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Jiemiao Shen
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China.
| | - Minghui Ji
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China.
| | - Yan Cui
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China.
| |
Collapse
|
3
|
Duan S, Cai T, Liu F, Li Y, Yuan H, Yuan W, Huang K, Hoettges K, Chen M, Lim EG, Zhao C, Song P. Automatic offline-capable smartphone paper-based microfluidic device for efficient biomarker detection of Alzheimer's disease. Anal Chim Acta 2024; 1308:342575. [PMID: 38740448 DOI: 10.1016/j.aca.2024.342575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a prevalent neurodegenerative disease with no effective treatment. Efficient and rapid detection plays a crucial role in mitigating and managing AD progression. Deep learning-assisted smartphone-based microfluidic paper analysis devices (μPADs) offer the advantages of low cost, good sensitivity, and rapid detection, providing a strategic pathway to address large-scale disease screening in resource-limited areas. However, existing smartphone-based detection platforms usually rely on large devices or cloud servers for data transfer and processing. Additionally, the implementation of automated colorimetric enzyme-linked immunoassay (c-ELISA) on μPADs can further facilitate the realization of smartphone μPADs platforms for efficient disease detection. RESULTS This paper introduces a new deep learning-assisted offline smartphone platform for early AD screening, offering rapid disease detection in low-resource areas. The proposed platform features a simple mechanical rotating structure controlled by a smartphone, enabling fully automated c-ELISA on μPADs. Our platform successfully applied sandwich c-ELISA for detecting the β-amyloid peptide 1-42 (Aβ 1-42, a crucial AD biomarker) and demonstrated its efficacy in 38 artificial plasma samples (healthy: 19, unhealthy: 19, N = 6). Moreover, we employed the YOLOv5 deep learning model and achieved an impressive 97 % accuracy on a dataset of 1824 images, which is 10.16 % higher than the traditional method of curve-fitting results. The trained YOLOv5 model was seamlessly integrated into the smartphone using the NCNN (Tencent's Neural Network Inference Framework), enabling deep learning-assisted offline detection. A user-friendly smartphone application was developed to control the entire process, realizing a streamlined "samples in, answers out" approach. SIGNIFICANCE This deep learning-assisted, low-cost, user-friendly, highly stable, and rapid-response automated offline smartphone-based detection platform represents a good advancement in point-of-care testing (POCT). Moreover, our platform provides a feasible approach for efficient AD detection by examining the level of Aβ 1-42, particularly in areas with low resources and limited communication infrastructure.
Collapse
Affiliation(s)
- Sixuan Duan
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, Suzhou, 215000, China; Department of Electrical and Electronic Engineering, University of Liverpool, Foundation Building, Brownlow Hill, Liverpool, L69 7ZX, UK; Key Laboratory of Bionic Engineering, Jilin University, 5988 Renmin Street, Changchun, 130022, China
| | - Tianyu Cai
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, Suzhou, 215000, China
| | - Fuyuan Liu
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, Suzhou, 215000, China; Department of Electrical and Electronic Engineering, University of Liverpool, Foundation Building, Brownlow Hill, Liverpool, L69 7ZX, UK
| | - Yifan Li
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, Suzhou, 215000, China; Department of Electrical and Electronic Engineering, University of Liverpool, Foundation Building, Brownlow Hill, Liverpool, L69 7ZX, UK
| | - Hang Yuan
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, Suzhou, 215000, China
| | - Wenwen Yuan
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, Suzhou, 215000, China; Department of Electrical and Electronic Engineering, University of Liverpool, Foundation Building, Brownlow Hill, Liverpool, L69 7ZX, UK; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, No.28 Xianning West Road, Xi'an, 710079, China
| | - Kaizhu Huang
- Department of Electrical and Computer Engineering, Duke Kunshan University, 8 Duke Avenue, Kunshan, 215316, China
| | - Kai Hoettges
- Department of Electrical and Electronic Engineering, University of Liverpool, Foundation Building, Brownlow Hill, Liverpool, L69 7ZX, UK
| | - Min Chen
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, Suzhou, 215000, China; Department of Electrical and Electronic Engineering, University of Liverpool, Foundation Building, Brownlow Hill, Liverpool, L69 7ZX, UK
| | - Eng Gee Lim
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, Suzhou, 215000, China; Department of Electrical and Electronic Engineering, University of Liverpool, Foundation Building, Brownlow Hill, Liverpool, L69 7ZX, UK
| | - Chun Zhao
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, Suzhou, 215000, China; Department of Electrical and Electronic Engineering, University of Liverpool, Foundation Building, Brownlow Hill, Liverpool, L69 7ZX, UK
| | - Pengfei Song
- School of Advanced Technology, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, Suzhou, 215000, China; Department of Electrical and Electronic Engineering, University of Liverpool, Foundation Building, Brownlow Hill, Liverpool, L69 7ZX, UK.
| |
Collapse
|
4
|
Ikanga J, Patrick SD, Schwinne M, Patel SS, Epenge E, Gikelekele G, Tshengele N, Kavugho I, Mampunza S, Yarasheski KE, Teunissen CE, Stringer A, Levey A, Rojas JC, Chan B, Lario Lago A, Kramer JH, Boxer AL, Jeromin A, Alonso A, Spencer RJ. Sensitivity of the African neuropsychology battery memory subtests and learning slopes in discriminating APOE 4 and amyloid pathology in adult individuals in the Democratic Republic of Congo. Front Neurol 2024; 15:1320727. [PMID: 38601333 PMCID: PMC11004441 DOI: 10.3389/fneur.2024.1320727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/14/2024] [Indexed: 04/12/2024] Open
Abstract
Background The current study examined the sensitivity of two memory subtests and their corresponding learning slope metrics derived from the African Neuropsychology Battery (ANB) to detect amyloid pathology and APOEε4 status in adults from Kinshasa, the Democratic Republic of the Congo. Methods 85 participants were classified for the presence of β-amyloid pathology and based on allelic presence of APOEε4 using Simoa. All participants were screened using CSID and AQ, underwent verbal and visuospatial memory testing from ANB, and provided blood samples for plasma Aβ42, Aβ40, and APOE proteotype. Pearson correlation, linear and logistic regression were conducted to compare amyloid pathology and APOEε4 status with derived learning scores, including initial learning, raw learning score, learning over trials, and learning ratio. Results Our sample included 35 amyloid positive and 44 amyloid negative individuals as well as 42 without and 39 with APOEε4. All ROC AUC ranges for the prediction of amyloid pathology based on learning scores were low, ranging between 0.56-0.70 (95% CI ranging from 0.44-0.82). The sensitivity of all the scores ranged between 54.3-88.6, with some learning metrics demonstrating good sensitivity. Regarding APOEε4 prediction, all AUC values ranged between 0.60-0.69, with all sensitivity measures ranging between 53.8-89.7. There were minimal differences in the AUC values across learning slope metrics, largely due to the lack of ceiling effects in this sample. Discussion This study demonstrates that some ANB memory subtests and learning slope metrics can discriminate those that are normal from those with amyloid pathology and those with and without APOEε4, consistent with findings reported in Western populations.
Collapse
Affiliation(s)
- Jean Ikanga
- Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| | - Sarah D. Patrick
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
| | - Megan Schwinne
- Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta, GA, United States
| | - Saranya Sundaram Patel
- Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Emmanuel Epenge
- Department of Neurology, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Guy Gikelekele
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| | - Nathan Tshengele
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| | | | - Samuel Mampunza
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| | | | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Neurodegeneration, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, Netherlands
| | - Anthony Stringer
- Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Allan Levey
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, United States
| | - Julio C. Rojas
- Department of Neurology, University of San Francisco, Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Brandon Chan
- Department of Neurology, University of San Francisco, Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Argentina Lario Lago
- Department of Neurology, University of San Francisco, Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Joel H. Kramer
- Department of Neurology, University of San Francisco, Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Adam L. Boxer
- Department of Neurology, University of San Francisco, Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | | | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Robert J. Spencer
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
| |
Collapse
|
5
|
Chudzik A, Śledzianowski A, Przybyszewski AW. Machine Learning and Digital Biomarkers Can Detect Early Stages of Neurodegenerative Diseases. SENSORS (BASEL, SWITZERLAND) 2024; 24:1572. [PMID: 38475108 PMCID: PMC10934426 DOI: 10.3390/s24051572] [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: 01/18/2024] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer's Disease (AD) and Parkinson's Disease (PD) are devastating conditions that can develop without noticeable symptoms, causing irreversible damage to neurons before any signs become clinically evident. NDs are a major cause of disability and mortality worldwide. Currently, there are no cures or treatments to halt their progression. Therefore, the development of early detection methods is urgently needed to delay neuronal loss as soon as possible. Despite advancements in Medtech, the early diagnosis of NDs remains a challenge at the intersection of medical, IT, and regulatory fields. Thus, this review explores "digital biomarkers" (tools designed for remote neurocognitive data collection and AI analysis) as a potential solution. The review summarizes that recent studies combining AI with digital biomarkers suggest the possibility of identifying pre-symptomatic indicators of NDs. For instance, research utilizing convolutional neural networks for eye tracking has achieved significant diagnostic accuracies. ROC-AUC scores reached up to 0.88, indicating high model performance in differentiating between PD patients and healthy controls. Similarly, advancements in facial expression analysis through tools have demonstrated significant potential in detecting emotional changes in ND patients, with some models reaching an accuracy of 0.89 and a precision of 0.85. This review follows a structured approach to article selection, starting with a comprehensive database search and culminating in a rigorous quality assessment and meaning for NDs of the different methods. The process is visualized in 10 tables with 54 parameters describing different approaches and their consequences for understanding various mechanisms in ND changes. However, these methods also face challenges related to data accuracy and privacy concerns. To address these issues, this review proposes strategies that emphasize the need for rigorous validation and rapid integration into clinical practice. Such integration could transform ND diagnostics, making early detection tools more cost-effective and globally accessible. In conclusion, this review underscores the urgent need to incorporate validated digital health tools into mainstream medical practice. This integration could indicate a new era in the early diagnosis of neurodegenerative diseases, potentially altering the trajectory of these conditions for millions worldwide. Thus, by highlighting specific and statistically significant findings, this review demonstrates the current progress in this field and the potential impact of these advancements on the global management of NDs.
Collapse
Affiliation(s)
- Artur Chudzik
- Polish-Japanese Academy of Information Technology, Faculty of Computer Science, 86 Koszykowa Street, 02-008 Warsaw, Poland; (A.C.); (A.Ś.)
| | - Albert Śledzianowski
- Polish-Japanese Academy of Information Technology, Faculty of Computer Science, 86 Koszykowa Street, 02-008 Warsaw, Poland; (A.C.); (A.Ś.)
| | - Andrzej W. Przybyszewski
- Polish-Japanese Academy of Information Technology, Faculty of Computer Science, 86 Koszykowa Street, 02-008 Warsaw, Poland; (A.C.); (A.Ś.)
- UMass Chan Medical School, Department of Neurology, 65 Lake Avenue, Worcester, MA 01655, USA
| |
Collapse
|
6
|
Rehman H, Ang TFA, Tao Q, Espenilla AL, Au R, Farrer LA, Zhang X, Qiu WQ. Comparison of Commonly Measured Plasma and Cerebrospinal Fluid Proteins and Their Significance for the Characterization of Cognitive Impairment Status. J Alzheimers Dis 2024; 97:621-633. [PMID: 38143358 DOI: 10.3233/jad-230837] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
BACKGROUND Although cerebrospinal fluid (CSF) amyloid-β42 peptide (Aβ42) and phosphorylated tau (p-tau) and blood p-tau are valuable for differential diagnosis of Alzheimer's disease (AD) from cognitively normal (CN) there is a lack of validated biomarkers for mild cognitive impairment (MCI). OBJECTIVE This study sought to determine how plasma and CSF protein markers compared in the characterization of MCI and AD status. METHODS This cohort study included Alzheimer's Disease Neuroimaging Initiative (ADNI) participants who had baseline levels of 75 proteins measured commonly in plasma and CSF (257 total, 46 CN, 143 MCI, and 68 AD). Logistic regression, least absolute shrinkage and selection operator (LASSO) and Random Forest (RF) methods were used to identify the protein candidates for the disease classification. RESULTS We observed that six plasma proteins panel (APOE, AMBP, C3, IL16, IGFBP2, APOD) outperformed the seven CSF proteins panel (VEGFA, HGF, PRL, FABP3, FGF4, CD40, RETN) as well as AD markers (CSF p-tau and Aβ42) to distinguish the MCI from AD [area under the curve (AUC) = 0.75 (plasma proteins), AUC = 0.60 (CSF proteins) and AUC = 0.56 (CSF p-tau and Aβ42)]. Also, these six plasma proteins performed better than the CSF proteins and were in line with CSF p-tau and Aβ42 in differentiating CN versus MCI subjects [AUC = 0.89 (plasma proteins), AUC = 0.85 (CSF proteins) and AUC = 0.89 (CSF p-tau and Aβ42)]. These results were adjusted for age, sex, education, and APOEϵ4 genotype. CONCLUSIONS This study suggests that the combination of 6 plasma proteins can serve as an effective marker for differentiating MCI from AD and CN.
Collapse
Affiliation(s)
- Habbiburr Rehman
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ting Fang Alvin Ang
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Qiushan Tao
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Arielle Lauren Espenilla
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Rhoda Au
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Xiaoling Zhang
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
7
|
Zhang F, Petersen M, Johnson L, Hall J, O'Bryant SE. Comorbidities Incorporated to Improve Prediction for Prevalent Mild Cognitive Impairment and Alzheimer's Disease in the HABS-HD Study. J Alzheimers Dis 2023; 96:1529-1546. [PMID: 38007662 DOI: 10.3233/jad-230755] [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: 11/27/2023]
Abstract
BACKGROUND Blood biomarkers have the potential to transform Alzheimer's disease (AD) diagnosis and monitoring, yet their integration with common medical comorbidities remains insufficiently explored. OBJECTIVE This study aims to enhance blood biomarkers' sensitivity, specificity, and predictive performance by incorporating comorbidities. We assess this integration's efficacy in diagnostic classification using machine learning, hypothesizing that it can identify a confident set of predictive features. METHODS We analyzed data from 1,705 participants in the Health and Aging Brain Study-Health Disparities, including 116 AD patients, 261 with mild cognitive impairment, and 1,328 cognitively normal controls. Blood samples were assayed using electrochemiluminescence and single molecule array technology, alongside comorbidity data gathered through clinical interviews and medical records. We visually explored blood biomarker and comorbidity characteristics, developed a Feature Importance and SVM-based Leave-One-Out Recursive Feature Elimination (FI-SVM-RFE-LOO) method to optimize feature selection, and compared four models: Biomarker Only, Comorbidity Only, Biomarker and Comorbidity, and Feature-Selected Biomarker and Comorbidity. RESULTS The combination model incorporating 17 blood biomarkers and 12 comorbidity variables outperformed single-modal models, with NPV12 at 92.78%, AUC at 67.59%, and Sensitivity at 65.70%. Feature selection led to 22 chosen features, resulting in the highest performance, with NPV12 at 93.76%, AUC at 69.22%, and Sensitivity at 70.69%. Additionally, interpretative machine learning highlighted factors contributing to improved prediction performance. CONCLUSIONS In conclusion, combining feature-selected biomarkers and comorbidities enhances prediction performance, while feature selection optimizes their integration. These findings hold promise for understanding AD pathophysiology and advancing preventive treatments.
Collapse
Affiliation(s)
- Fan Zhang
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Melissa Petersen
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Leigh Johnson
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - James Hall
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Sid E O'Bryant
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
8
|
O’Bryant SE, Petersen M, Zhang F, Johnson L, German D, Hall J. Parkinson's Disease Blood Test for Primary Care. JOURNAL OF ALZHEIMER'S DISEASE & PARKINSONISM 2022; 12:545. [PMID: 37006377 PMCID: PMC10065753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Background A blood-test that could serve as a potential first step in a multi-tiered neurodiagnostic process for ruling out Parkinson's disease (PD) in primary care settings would be of tremendous value. This study therefore sought to conduct a large-scale cross-validation of our Parkinson's disease Blood Test (PDBT) for use in primary care settings. Methods Serum samples were analyzed from 846 PD and 2291 volunteer controls. Proteomic assays were run on a multiplex biomarker assay platform using Electrochemiluminescence (ECL). Diagnostic accuracy statistics were generated using area under the receiver operating characteristic curve (AUC), Sensitivity (SN), Specificity (SP) and Negative Predictive Value (NPV). Results In the training set, the PDBT reached an AUC of 0.98 when distinguishing PD cases from controls with a SN of 0.84 and SP of 0.98. When applied to the test set, the PDBT yielded an AUC of 0.96, SN of 0.79 and SP of 0.97. The PDBT obtained a negative predictive value of 99% for a 2% base rate. Conclusion The PDBT was highly successful in discriminating PD patients from control cases and has great potential for providing primary care providers with a rapid, scalable and cost-effective tool for screening out PD.
Collapse
Affiliation(s)
- Sid E. O’Bryant
- Department of Neuroscience and Pharmacology, University of North Texas Health Science Center, Fort Worth, Texas, USA
- Department of Medicine, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Melissa Petersen
- Department of Neuroscience and Pharmacology, University of North Texas Health Science Center, Fort Worth, Texas, USA
- Department of Medicine, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Fan Zhang
- Department of Neuroscience and Pharmacology, University of North Texas Health Science Center, Fort Worth, Texas, USA
- Department of Medicine, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Leigh Johnson
- Department of Neuroscience and Pharmacology, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Dwight German
- Department of Psychiatry, UT Southwestern Medical School, Dallas, Texas, USA
| | - James Hall
- Department of Neuroscience and Pharmacology, University of North Texas Health Science Center, Fort Worth, Texas, USA
- Department of Medicine, University of North Texas Health Science Center, Fort Worth, Texas, USA
| |
Collapse
|
9
|
Jung M, Pan X, Cunningham EL, Passmore AP, McGuinness B, McAuley DF, Beverland D, O’Brien S, Mawhinney T, Schott JM, Zetterberg H, Green BD. The Influence of Orthopedic Surgery on Circulating Metabolite Levels, and their Associations with the Incidence of Postoperative Delirium. Metabolites 2022; 12:616. [PMID: 35888740 PMCID: PMC9319890 DOI: 10.3390/metabo12070616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 02/05/2023] Open
Abstract
The mechanisms underlying the occurrence of postoperative delirium development are unclear and measurement of plasma metabolites may improve understanding of its causes. Participants (n = 54) matched for age and gender were sampled from an observational cohort study investigating postoperative delirium. Participants were ≥65 years without a diagnosis of dementia and presented for primary elective hip or knee arthroplasty. Plasma samples collected pre- and postoperatively were grouped as either control (n = 26, aged: 75.8 ± 5.2) or delirium (n = 28, aged: 76.2 ± 5.7). Widespread changes in plasma metabolite levels occurred following surgery. The only metabolites significantly differing between corresponding control and delirium samples were ornithine and spermine. In delirium cases, ornithine was 17.6% higher preoperatively, and spermine was 12.0% higher postoperatively. Changes were not associated with various perioperative factors. In binary logistic regression modeling, these two metabolites did not confer a significantly increased risk of delirium. These findings support the hypothesis that disturbed polyamine metabolism is an underlying factor in delirium that warrants further investigation.
Collapse
Affiliation(s)
- Mijin Jung
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, 8 Malone Road, Belfast BT9 5BN, Northern Ireland, UK; (M.J.); (X.P.)
| | - Xiaobei Pan
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, 8 Malone Road, Belfast BT9 5BN, Northern Ireland, UK; (M.J.); (X.P.)
| | - Emma L. Cunningham
- Centre for Public Health, Institute of Clinical Sciences, Queen’s University Belfast, Block B, Royal Victoria Hospital Site, Grosvenor Road, Belfast BT12 6BA, Northern Ireland, UK; (E.L.C.); (A.P.P.); (B.M.)
| | - Anthony P. Passmore
- Centre for Public Health, Institute of Clinical Sciences, Queen’s University Belfast, Block B, Royal Victoria Hospital Site, Grosvenor Road, Belfast BT12 6BA, Northern Ireland, UK; (E.L.C.); (A.P.P.); (B.M.)
| | - Bernadette McGuinness
- Centre for Public Health, Institute of Clinical Sciences, Queen’s University Belfast, Block B, Royal Victoria Hospital Site, Grosvenor Road, Belfast BT12 6BA, Northern Ireland, UK; (E.L.C.); (A.P.P.); (B.M.)
| | - Daniel F. McAuley
- Centre for Experimental Medicine, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, Northern Ireland, UK;
| | - David Beverland
- Outcomes Assessment Unit, Musgrave Park Hospital, Belfast Trust, Stockman’s Lane, Belfast BT9 7JB, Northern Ireland, UK;
| | - Seamus O’Brien
- Cardiac Surgical Intensive Care Unit, Belfast Trust, Royal Victoria Hospital, Grosvenor Road, Belfast BT12 6BA, Northern Ireland, UK; (S.O.); (T.M.)
| | - Tim Mawhinney
- Cardiac Surgical Intensive Care Unit, Belfast Trust, Royal Victoria Hospital, Grosvenor Road, Belfast BT12 6BA, Northern Ireland, UK; (S.O.); (T.M.)
| | - Jonathan M. Schott
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1E 6BT, UK; (J.M.S.); (H.Z.)
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1E 6BT, UK; (J.M.S.); (H.Z.)
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, House V, S-431 80 Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, House V, S-431 80 Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Brian D. Green
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, 8 Malone Road, Belfast BT9 5BN, Northern Ireland, UK; (M.J.); (X.P.)
| |
Collapse
|
10
|
Guo Y, Hu Z, Wang Z. Recent Advances in the Application Peptide and Peptoid in Diagnosis Biomarkers of Alzheimer's Disease in Blood. Front Mol Neurosci 2021; 14:778955. [PMID: 35002620 PMCID: PMC8733658 DOI: 10.3389/fnmol.2021.778955] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases with irreversible damage of the brain and a continuous pathophysiological process. Early detection and accurate diagnosis are essential for the early intervention of AD. Precise detection of blood biomarkers related to AD could provide a shortcut to identifying early-stage patients before symptoms. In recent years, targeting peptides or peptoids have been chosen as recognition elements in nano-sensors or fluorescence detection to increase the targeting specificity, while peptide-based probes were also developed considering their specific advantages. Peptide-based sensors and probes have been developed according to different strategies, such as natural receptors, high-throughput screening, or artificial design for AD detection. This review will briefly summarize the recent developments and trends of AD diagnosis platforms based on peptide and peptoid as recognition elements and provide insights into the application of peptide and peptoid with different sources and characteristics in the diagnosis of AD biomarkers.
Collapse
Affiliation(s)
- Yuxin Guo
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyuan Hu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- School of Nanoscience and Technology, Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
| | - Zihua Wang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
11
|
A tacrine-tetrahydroquinoline heterodimer potently inhibits acetylcholinesterase activity and enhances neurotransmission in mice. Eur J Med Chem 2021; 226:113827. [PMID: 34530383 DOI: 10.1016/j.ejmech.2021.113827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/26/2021] [Accepted: 09/02/2021] [Indexed: 11/23/2022]
Abstract
Cholinergic neurons are ubiquitous and involved in various higher brain functions including learning and memory. Patients with Alzheimer's disease exhibit significant dysfunction and loss of cholinergic neurons. Meanwhile, such cholinergic deficits can be potentially relieved pharmacologically by increasing acetylcholine. Acetylcholinesterase (AChE) inhibitors have been used to improve cholinergic transmission in the brain for two decades and have proven effective for alleviating symptoms in the early stages of Alzheimer's disease. Therefore, the search for AChE inhibitors for drug development is ongoing. The enzymatic pocket of AChE has long been the target of several drug designs over the last two decades. The peripheral and catalytic sites of AChE are simultaneously bound by several dimeric molecules, enabling more-efficient inhibition. Here, we used 6-chlorotacrine and the tetrahydroquinolone moiety of huperzine A to design and synthesize a series of heterodimers that inhibit AChE at nanomolar potency. Specifically, compound 7b inhibits AChE with an IC50 < 1 nM and spares butyrylcholinesterase. Administration of 7b to mouse brain slices restores synaptic activity impaired by pirenzepine, a muscarinic M1-selective antagonist. Moreover, oral administration of 7b to C57BL/6 mice enhances hippocampal long-term potentiation in a dose-dependent manner and is detectable in the brain tissue. All these data supported that 7b is a potential cognitive enhancer and is worth for further exploration.
Collapse
|
12
|
Supraja P, Tripathy S, Singh R, Singh V, Chaudhury G, Singh SG. Towards point-of-care diagnosis of Alzheimer's disease: Multi-analyte based portable chemiresistive platform for simultaneous detection of β-amyloid (1-40) and (1-42) in plasma. Biosens Bioelectron 2021; 186:113294. [PMID: 33971525 DOI: 10.1016/j.bios.2021.113294] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/11/2021] [Accepted: 04/28/2021] [Indexed: 01/05/2023]
Abstract
Label-free simultaneous detection of Alzheimer's disease (AD) specific biomarkers Aβ40 and Aβ42 peptides on a single platform using polypyrrole nanoparticle-based chemiresistive biosensors is reported here. The proposed interdigitated-microelectrode based inexpensive multisensor-platform can concurrently detect Aβ40 and Aβ42 in spiked-plasma in the range of 10-14 - 10-6 g/mL (with LoDs being 5.71 and 9.09 fg/mL, respectively), enabling the estimation of diagnostically significant Aβ42/Aβ40 ratio. A detailed study has been undertaken here to record the individual sensor responses against spiked-plasma samples with varying amounts and proportions of the two target peptides, towards enabling disease-progression monitoring using the Aβ-ratio. As compared to the existing cost-ineffective brain-imaging techniques such as PET and MRI, and the high-risk CSF based invasive AD biomarkers detecting procedures, the proposed approach offers a viable alternative for affordable point-of-care AD diagnostics, with possible usage in performance evaluation of therapeutic drugs. Towards point-of-care applications, the portable readout used in this work was conjugated with an android-based mobile app for data-acquisition and analysis.
Collapse
Affiliation(s)
- Patta Supraja
- Department of Electrical Engineering, Indian Institute of Technology Hyderabad, 502285, India.
| | - Suryasnata Tripathy
- Department of Electrical Engineering, Indian Institute of Technology Hyderabad, 502285, India.
| | - Ranjana Singh
- Department of Electrical Engineering, Indian Institute of Technology Hyderabad, 502285, India.
| | - Vikrant Singh
- School of Medicine, University of California Davis, USA.
| | - Gajendranath Chaudhury
- Department of Electrical Engineering, Indian Institute of Technology Hyderabad, 502285, India.
| | - Shiv Govind Singh
- Department of Electrical Engineering, Indian Institute of Technology Hyderabad, 502285, India.
| |
Collapse
|
13
|
Grossberg AN, Bettcher BM, Gorgens KA, Ledreux A. Curiosity-Based Interventions Increase Everyday Functioning Score But Not Serum BDNF Levels in a Cohort of Healthy Older Adults. FRONTIERS IN AGING 2021; 2:700838. [PMID: 35822037 PMCID: PMC9261453 DOI: 10.3389/fragi.2021.700838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/28/2021] [Indexed: 06/15/2023]
Abstract
An enriched environment is effective in stimulating learning and memory in animal models as well as in humans. Environmental enrichment increases brain-derived neurotrophic factor (BDNF) in aged rats and reduces levels of Alzheimer-related proteins in the blood, including amyloid-β (Aβ) peptides and misfolded toxic forms of tau. To address whether stimulation of curiosity, which is a form of enrichment, may provide a buffer against Alzheimer's disease (AD), we measured levels of biomarkers associated with AD at baseline and after a 6-week intervention in older adults (>65 years of age) randomized to one of three different intervention conditions. Specifically, in this pilot study, we tested the effectiveness of a traditional, structured learning environment compared to a self-motivated learning environment designed to stimulate curiosity. There were no significant differences from baseline to post-intervention in any of the groups for Aβ42/Aβ40 ratio or t-tau (total-tau) plasma levels. Serum BDNF levels decreased significantly in the control group. Interestingly, individuals who had the lowest serum BDNF levels at baseline experienced significantly higher increases in BDNF over the course of the 6-week intervention compared to individuals with higher serum BDNF levels at baseline. As expected, older individuals had lower MoCA scores. Years of education correlated negatively with Aβ levels, suggesting a protective effect of education on levels of this toxic protein. ECog scores were negatively correlated with BDNF levels, suggesting that better performance on the ECog questionnaire was associated with higher BDNF levels. Collectively, these findings did not suggest that a 6-week cognitive training intervention focused on curiosity resulted in significant alterations in blood biomarkers but showed interesting correlations between cognitive scores and BDNF levels, further supporting the role of this trophic factor in brain health in older adults.
Collapse
Affiliation(s)
- Allison N. Grossberg
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, United States
| | - Brianne M. Bettcher
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kim A. Gorgens
- Graduate School of Professional Psychology, University of Denver, Denver, CO, United States
| | - Aurélie Ledreux
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, United States
| |
Collapse
|
14
|
Sun X, Duan S, Cao A, Villagomez B, Lin R, Chen H, Pi L, Ren B, Chen R, Chen M, Ying Z, Fang S, Cao Q. RRY Inhibits Amyloid-β 1-42 Peptide Aggregation and Neurotoxicity. J Alzheimers Dis Rep 2021; 5:479-495. [PMID: 34368633 PMCID: PMC8293670 DOI: 10.3233/adr-210012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Current understanding of amyloid-β protein (Aβ) aggregation and toxicity provides an extensive list of drugs for treating Alzheimer's disease (AD); however, one of the most promising strategies for its treatment has been tri-peptides. OBJECTIVE The aim of this study is to examine those tri-peptides, such as Arg-Arg-Try (RRY), which have the potential of Aβ1-42 aggregating inhibition and Aβ clearance. METHODS In the present study, in silico, in vitro, and in vivo studies were integrated for screening tri-peptides binding to Aβ, then evaluating its inhibition of aggregation of Aβ, and finally its rescuing cognitive deficit. RESULTS In the in silico simulations, molecular docking and molecular dynamics determined that seven top-ranking tri-peptides could bind to Aβ1-42 and form stable complexes. Circular dichroism, ThT assay, and transmission electron microscope indicated the seven tri-peptides might inhibit the aggregation of Aβ1-42 in vitro. In the in vivo studies, Morris water maze, ELISA, and Diolistic staining were used, and data showed that RRY was capable of rescuing the Aβ1-42-induced cognitive deficit, reducing the Aβ1-42 load and increasing the dendritic spines in the transgenic mouse model. CONCLUSION Such converging outcomes from three consecutive studies lead us to conclude that RRY is a preferred inhibitor of Aβ1-42 aggregation and treatment for Aβ-induced cognitive deficit.
Collapse
Affiliation(s)
- Xicui Sun
- Department of Neurology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Songwei Duan
- Department of Neurology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Anna Cao
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bryan Villagomez
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Runxuan Lin
- Department of Neurology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hongxia Chen
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Liya Pi
- Department of Pediatrics in College of Medicine, University of Florida, Gainesville, FL, USA
| | - Bin Ren
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rong Chen
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Minjie Chen
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zhekang Ying
- The Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shenyun Fang
- Department of Physiology, University of Maryland, Baltimore, Maryland, USA
| | - Qi Cao
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
15
|
Zhang F, Petersen M, Johnson L, Hall J, O'Bryant SE. Recursive Support Vector Machine Biomarker Selection for Alzheimer's Disease. J Alzheimers Dis 2021; 79:1691-1700. [PMID: 33492292 DOI: 10.3233/jad-201254] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is a need for more reliable diagnostic tools for the early detection of Alzheimer's disease (AD). This can be a challenge due to a number of factors and logistics making machine learning a viable option. OBJECTIVE In this paper, we present on a Support Vector Machine Leave-One-Out Recursive Feature Elimination and Cross Validation (SVM-RFE-LOO) algorithm for use in the early detection of AD and show how the SVM-RFE-LOO method can be used for both classification and prediction of AD. METHODS Data were analyzed on n = 300 participants (n = 150 AD; n = 150 cognitively normal controls). Serum samples were assayed via a multi-plex biomarker assay platform using electrochemiluminescence (ECL). RESULTS The SVM-RFE-LOO method reduced the number of features in the model from 21 to 16 biomarkers and achieved an area under the curve (AUC) of 0.980 with a sensitivity of 94.0% and a specificity of 93.3%. When the classification and prediction performance of SVM-RFE-LOO was compared to that of SVM and SVM-RFE, we found similar performance across the models; however, the SVM-RFE-LOO method utilized fewer markers. CONCLUSION We found that 1) the SVM-RFE-LOO is suitable for analyzing noisy high-throughput proteomic data, 2) it outperforms SVM-RFE in the robustness to noise and in the ability to recover informative features, and 3) it can improve the prediction performance. Our recursive feature elimination model can serve as a general model for biomarker discovery in other diseases.
Collapse
Affiliation(s)
- Fan Zhang
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA.,Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Melissa Petersen
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA.,Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Leigh Johnson
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - James Hall
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Sid E O'Bryant
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
16
|
Corbo C, Li AA, Poustchi H, Lee GY, Stacks S, Molinaro R, Ma P, Platt T, Behzadi S, Langer R, Farias V, Farokhzad OC. Analysis of the Human Plasma Proteome Using Multi-Nanoparticle Protein Corona for Detection of Alzheimer's Disease. Adv Healthc Mater 2021; 10:e2000948. [PMID: 33169521 DOI: 10.1002/adhm.202000948] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/16/2020] [Indexed: 12/22/2022]
Abstract
As the population affected by Alzheimer's disease (AD) grows, so does the need for a noninvasive and accurate diagnostic tool. Current research reveals that AD pathogenesis begins as early as decades before clinical symptoms. The unique properties of nanoparticles (NPs) may be exploited to develop noninvasive diagnostics for early detection of AD. After exposure of NPs to biological fluids, the NP surface is altered by an unbiased but selective and reproducible adsorption of biomolecules commonly referred to as the biomolecular corona or protein corona (PC). The discovery that the plasma proteome may be differentially altered during health and disease leads to the concept of disease-specific PCs. Herein, the disease-specific PCs formed around NPs in a multi-NPs platform are employed to successfully identify subtle changes in plasma protein patterns and detect AD (>92% specificity and ≈100% sensitivity). Similar discrimination power is achieved using banked plasma samples from a cohort of patients several years prior to their diagnosis with AD. With the nanoplatform's analytic ability to analyze pathological proteomic changes into a disease-specific identifier, this promising, noninvasive technology with implications for early detection and intervention could benefit not only patients with AD but other diseases as well.
Collapse
Affiliation(s)
- Claudia Corbo
- Center for Nanomedicine Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
- School of Medicine and Surgery Nanomedicine Center Nanomib University of Milano‐Bicocca Vedano al Lambro 20854 Italy
| | - Andrew A. Li
- Tepper School of Business Carnegie Mellon University Pittsburgh PA 15213 USA
| | - Hossein Poustchi
- Digestive Oncology Research Center Digestive Disease Research Institute Tehran University of Medical Sciences Tehran 4117‐13135 Iran
| | - Gha Young Lee
- Center for Nanomedicine Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Sabrina Stacks
- Center for Nanomedicine Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Roberto Molinaro
- Department of Medicine Brigham and Women's Hospital Harvard Medical School 77 Avenue Louis Pasteur Boston MA 02115 USA
| | - Philip Ma
- Seer, Inc. 3800 Bridge Parkway Redwood City CA 94065 USA
| | - Theo Platt
- Seer, Inc. 3800 Bridge Parkway Redwood City CA 94065 USA
| | - Shahed Behzadi
- Center for Nanomedicine Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Robert Langer
- Koch Institute for Integrative Cancer Research at MIT Cambridge MA 02139‐4307 USA
| | - Vivek Farias
- Sloan School of Management Massachusetts Institute of Technology Cambridge MA 02142 USA
| | - Omid C. Farokhzad
- Center for Nanomedicine Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| |
Collapse
|
17
|
Wharton W, Jeong L, Ni L, Bay AA, Shin RJ, McCullough LE, Silverstein H, Hart AR, Swieboda D, Hu W, Hackney ME. A Pilot randomized clinical trial of adapted tango to improve cognition and psychosocial function in African American women with family history of Alzheimer's disease (ACT trial). CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100018. [PMID: 36324714 PMCID: PMC9616328 DOI: 10.1016/j.cccb.2021.100018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/30/2021] [Accepted: 06/17/2021] [Indexed: 11/24/2022]
Abstract
the adapted tango intervention was very well received as demonstrated by minimal participant attrition, satisfaction questionnaires that indicated high satisfaction, and anecdotal reports. Adapted tango intervention may be helpful in controlling or reducing markers of inflammation in AA women with a parental history of AD. Participants in tango demonstrated improvements in whole-body spatial cognition and short-term and working memory, and reduced deterioration of executive function. Although our tango group did not show large positive effect in cumulative caregiver burden post intervention, the large positive effect in role Captivity, caregiver confidence, and deprivation of intimate exchange displays some of the positive effects of this dance intervention for the caregivers.
Alzheimer's disease (AD) is a devastating, progressive neurodegenerative disease resulting in memory loss and a severe reduction in the ability to perform activities of daily living. Ethnicity-related genetic factors promoting the development of dementias among African Americans (AA) and increased risk among women for developing AD indicates that AA women with a parental history of AD are at great risk for developing AD. This phase I study assessed the impact of a 12 week, 20-lesson adapted Argentine Tango intervention (n = 24) to a no-contact control group (n = 10) on measures of plasma inflammatory markers, cognition, and motor and psychosocial performance in middle-aged AA woman at increased risk for AD by virtue of parental history. Some woman (n = 16) were also caregivers; thus, the impact of the intervention on caregiving burden was examined in this subset. Preliminary analysis of efficacy was conducted with significance tests on biomarkers and key measures of cognition, including visuospatial and executive function, balance, and strength. After 12 weeks, Tango participants had significantly decreased inflammatory cytokine, including reductions in IL-7 (p = 0.003), IFN-γ (p = 0.011), TNFα (p = 0.011), and MCP-1 (p = 0.042) compared to controls. Large effects were noted for the Tango group on tests of executive functioning (d = 0.89), and inhibition (p = 0.031). Participants in Tango improved in dynamic and static balance (p = 0.018) and functional lower body strength (p = 0.023). Secondary assessment revealed trends favoring the intervention group were noted in spatial cognition and executive function. Moderate effects were noted in caregiving burden measures among the subset of caregivers. These data demonstrate substantial reductions in inflammatory biomarkers along with cognitive and motor improvements through a non-pharmacologic, affordable intervention among a small, well-characterized cohort of AA women with a parental history of AD.
Collapse
Affiliation(s)
| | - Leanne Jeong
- Emory University College of Arts and Sciences, 550 Asbury Circle, Atlanta, GA, 30322, USA
| | - Liang Ni
- Division of Geriatrics and Gerontology, Department of Medicine, Emory School of Medicine, 1841 Clifton Rd NE, Atlanta, GA, 30307, USA
| | - Allison A. Bay
- Division of Geriatrics and Gerontology, Department of Medicine, Emory School of Medicine, 1841 Clifton Rd NE, Atlanta, GA, 30307, USA
| | - Ryan J. Shin
- Emory University College of Arts and Sciences, 550 Asbury Circle, Atlanta, GA, 30322, USA
| | - Lauren E. McCullough
- Emory University Rollins School of Public Health, 1518 Clifton Rd., Atlanta, GA, 30329, USA
| | - Hayley Silverstein
- Division of Geriatrics and Gerontology, Department of Medicine, Emory School of Medicine, 1841 Clifton Rd NE, Atlanta, GA, 30307, USA
| | | | | | - William Hu
- Division of Cognitive Neurology, Rutgers Robert Wood Johnson Medical School, 125 Paterson Street, New Brunswick, NJ 08901, USA
| | - Madeleine E. Hackney
- Emory University School of Nursing, Atlanta, GA, 30307, USA
- Division of Geriatrics and Gerontology, Department of Medicine, Emory School of Medicine, 1841 Clifton Rd NE, Atlanta, GA, 30307, USA
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, 1670 Clairmont Rd., Decatur, GA, 30033, USA
- Department of Rehabilitation Medicine, Emory School of Medicine, 1648 Pierce Dr. NE, Atlanta, GA, 30307, USA
- Birmingham/Atlanta VA Geriatric Research Clinical and Education Center
- Corresponding author at: Emory University School of Medicine, Department of Medicine, Division of Geriatrics and Gerontology, Research Health Scientist, Atlanta VA Rehabilitation R&D Center for Visual and Neurocognitive Rehabilitation, 1841 Clifton Rd. NE, #553, Atlanta, GA 30329, USA.
| |
Collapse
|
18
|
Use of Fourier-Transform Infrared Spectroscopy (FT-IR) for Monitoring Experimental Helicobacter pylori Infection and Related Inflammatory Response in Guinea Pig Model. Int J Mol Sci 2020; 22:ijms22010281. [PMID: 33396581 PMCID: PMC7795336 DOI: 10.3390/ijms22010281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/25/2020] [Accepted: 12/26/2020] [Indexed: 12/17/2022] Open
Abstract
Infections due to Gram-negative bacteria Helicobacter pylori may result in humans having gastritis, gastric or duodenal ulcer, and even gastric cancer. Investigation of quantitative changes of soluble biomarkers, correlating with H. pylori infection, is a promising tool for monitoring the course of infection and inflammatory response. The aim of this study was to determine, using an experimental model of H. pylori infection in guinea pigs, the specific characteristics of infrared spectra (IR) of sera from H. pylori infected (40) vs. uninfected (20) guinea pigs. The H. pylori status was confirmed by histological, molecular, and serological examination. The IR spectra were measured using a Fourier-transform (FT)-IR spectrometer Spectrum 400 (PerkinElmer) within the range of wavenumbers 3000–750 cm−1 and converted to first derivative spectra. Ten wavenumbers correlated with H. pylori infection, based on the chi-square test, were selected for a K-nearest neighbors (k-NN) algorithm. The wavenumbers correlating with infection were identified in the W2 and W3 windows associated mainly with proteins and in the W4 window related to nucleic acids and hydrocarbons. The k-NN for detection of H. pylori infection has been developed based on chemometric data. Using this model, animals were classified as infected with H. pylori with 100% specificity and 97% sensitivity. To summarize, the IR spectroscopy and k-NN algorithm are useful for monitoring experimental H. pylori infection and related inflammatory response in guinea pig model and may be considered for application in humans.
Collapse
|
19
|
Dwivedi V, Yaniv K, Sharon M. Beyond cells: The extracellular circulating 20S proteasomes. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166041. [PMID: 33338594 DOI: 10.1016/j.bbadis.2020.166041] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/30/2020] [Accepted: 12/04/2020] [Indexed: 01/08/2023]
Abstract
Accumulating evidence arising from numerous clinical studies indicate that assembled and functional 20S proteasome complexes circulate freely in plasma. Elevated levels of this core proteolytic complex have been found in the plasma of patients suffering from blood, skin and solid cancers, autoimmune disorders, trauma and sepsis. Moreover, in various diseases, there is a positive correlation between circulating 20S proteasome (c20S) levels and treatment efficacy and survival rates, suggesting the involvement of this under-studied c20S complex in pathophysiology. However, many aspects of this system remain enigmatic, as we still do not know the origin, biological role or mechanisms of extracellular transport and regulation of c20S proteasomes. In this review, we provide an overview of the current understanding of the c20S proteasome system and discuss the remaining gaps in knowledge.
Collapse
Affiliation(s)
- Vandita Dwivedi
- Departments of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Karina Yaniv
- Departments of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michal Sharon
- Departments of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
20
|
Rehiman SH, Lim SM, Lim FT, Chin AV, Tan MP, Kamaruzzaman SB, Ramasamy K, Abdul Majeed AB. Fibrinogen isoforms as potential blood-based biomarkers of Alzheimer's disease using a proteomics approach. Int J Neurosci 2020; 132:1014-1025. [PMID: 33280461 DOI: 10.1080/00207454.2020.1860038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Objective: Alzheimer's disease (AD), the commonest form of dementia which is characterized by progressive decline in cognitive function, can only be definitively diagnosed after death. Although biomarkers may aid diagnosis, currently available AD biomarkers, which are predominantly based on cerebrospinal fluid and neuroimaging facilities, are either invasive or costly. Blood-based biomarkers for AD diagnosis are highly sought after due to its practicality at the clinic. This study was undertaken to determine the differential protein expression in plasma amongst Malaysian AD, mild cognitive impairment (MCI) and non-AD individuals. Methods: A proteomic approach which utilized two-dimensional differential in gel electrophoresis (2 D DIGE) was performed for blood samples from 15 AD, 14 MCI and 15 non-AD individuals. Results: Mass spectrometry (MS)-based protein identification via MALDI ToF/ToF showed that fibrinogen-β-chain (spot 64) and fibrinogen-γ-chain (spot 91) with differential expression ratio >1.5 were significantly upregulated (p < 0.05) in AD patients when compared to non-AD individuals. Further data analysis using Pearson correlation found that the upregulated fibrinogen-γ-chain was weakly but significantly (p < 0.05) and inversely correlated with cognitive decline. Conclusion: Fibrinogen isoforms may play important roles in the vascular pathology of AD as well as neuroinflammation. As such, fibrinogen appears to be a promising blood-based biomarker for AD. Further validation of the present findings in larger population is now warranted.
Collapse
Affiliation(s)
- Siti Hajar Rehiman
- Collaborative Drug Discovery Research (CDDR) and Brain Degeneration and Therapeutics Research Group, Faculty of Pharmacy, University Teknologi MARA (UiTM) Cawangan Selangor, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) and Brain Degeneration and Therapeutics Research Group, Faculty of Pharmacy, University Teknologi MARA (UiTM) Cawangan Selangor, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Fei Tieng Lim
- Collaborative Drug Discovery Research (CDDR) and Brain Degeneration and Therapeutics Research Group, Faculty of Pharmacy, University Teknologi MARA (UiTM) Cawangan Selangor, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Ai-Vyrn Chin
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Maw Pin Tan
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Shahrul Bahyah Kamaruzzaman
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) and Brain Degeneration and Therapeutics Research Group, Faculty of Pharmacy, University Teknologi MARA (UiTM) Cawangan Selangor, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Abu Bakar Abdul Majeed
- Collaborative Drug Discovery Research (CDDR) and Brain Degeneration and Therapeutics Research Group, Faculty of Pharmacy, University Teknologi MARA (UiTM) Cawangan Selangor, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
21
|
Rehiman SH, Lim SM, Neoh CF, Majeed ABA, Chin AV, Tan MP, Kamaruzzaman SB, Ramasamy K. Proteomics as a reliable approach for discovery of blood-based Alzheimer's disease biomarkers: A systematic review and meta-analysis. Ageing Res Rev 2020; 60:101066. [PMID: 32294542 DOI: 10.1016/j.arr.2020.101066] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 02/08/2023]
Abstract
In order to gauge the impact of proteomics in discovery of Alzheimer's disease (AD) blood-based biomarkers, this study had systematically reviewed articles published between 1984-2019. Articles that fulfilled the inclusion criteria were assessed for risk of bias. A meta-analysis was performed for replicable candidate biomarkers (CB). Of the 1651 articles that were identified, 17 case-control and two cohort studies, as well as three combined case-control and longitudinal designs were shortlisted. A total of 207 AD and mild cognitive impairment (MCI) CB were discovered, with 48 reported in >2 studies. This review highlights six CB, namely alpha-2-macroglobulin (α2M)ps, pancreatic polypeptide (PP)ps, apolipoprotein A-1 (ApoA-1)ps, afaminp, insulin growth factor binding protein-2 (IGFBP-2)ps and fibrinogen-γ-chainp, all of which exhibited consistent pattern of regulation in >three independent cohorts. They are involved in AD pathogenesis via amyloid-beta (Aβ), neurofibrillary tangles, diabetes and cardiovascular diseases (CVD). Meta-analysis indicated that ApoA-1ps was significantly downregulated in AD (SMD = -1.52, 95% CI: -1.89, -1.16, p < 0.00001), with low inter-study heterogeneity (I2 = 0%, p = 0.59). α2Mps was significantly upregulated in AD (SMD = 0.83, 95% CI: 0.05, 1.62, p = 0.04), with moderate inter-study heterogeneity (I2 = 41%, p = 0.19). Both CB are involved in Aβ formation. These findings provide important insights into blood-based AD biomarkers discovery via proteomics.
Collapse
|
22
|
Serafín V, Gamella M, Pedrero M, Montero-Calle A, Razzino CA, Yáñez-Sedeño P, Barderas R, Campuzano S, Pingarrón JM. Enlightening the advancements in electrochemical bioanalysis for the diagnosis of Alzheimer's disease and other neurodegenerative disorders. J Pharm Biomed Anal 2020; 189:113437. [PMID: 32629192 DOI: 10.1016/j.jpba.2020.113437] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022]
Abstract
Neurodegenerative disorders (NDD), and particularly Alzheimer's disease (AD), are one of the greatest challenges facing our current medicine and society because of its increasing incidence and the high burden imposed both on patients' families and health systems. Despite this, their accurate diagnosis, mostly conducted by cerebrospinal fluid (CSF) analysis or neuroimaging techniques, costly, time-consuming, and unaffordable for most of the population, remains a complex task. In this situation, electrochemical biosensors are flourishing as promising alternative tools for the simple, fast, and low-cost diagnosis of NDD/AD. This review article provides the relevant clinical details of NDD/AD along with the closely related genetic (genetic mutations, polymorphisms of ApoE and specific miRNAs) and proteomic (amyloid-β peptides, total and phosphorylated tau protein) biomarkers circulating mostly in CSF. In addition, the article systematically enlightens a general view of the electrochemical affinity biosensors (mostly aptasensors and immunosensors) reported in the past two years for the determination of such biomarkers. The different developed strategies, analytical performances and applications are comprehensively discussed. Recent advancements in signal amplification methodologies involving smart designs and the use of nanomaterials and rational surface chemistries, as well as the challenges that must be struggled and the prospects in electrochemical affinity biosensing to bring more accessibility to NDD/AD diagnosis, prognosis, and follow-up, are also pointed out.
Collapse
Affiliation(s)
- V Serafín
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Madrid, 28040, Spain
| | - M Gamella
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Madrid, 28040, Spain
| | - M Pedrero
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Madrid, 28040, Spain
| | - A Montero-Calle
- Chronic Disease Programme, UFIEC, Carlos III Health Institute, Majadahonda, Madrid, 28220, Spain
| | - C A Razzino
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Madrid, 28040, Spain
| | - P Yáñez-Sedeño
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Madrid, 28040, Spain
| | - R Barderas
- Chronic Disease Programme, UFIEC, Carlos III Health Institute, Majadahonda, Madrid, 28220, Spain.
| | - S Campuzano
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Madrid, 28040, Spain.
| | - J M Pingarrón
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Madrid, 28040, Spain.
| |
Collapse
|
23
|
Serafín V, Razzino CA, Gamella M, Pedrero M, Povedano E, Montero-Calle A, Barderas R, Calero M, Lobo AO, Yáñez-Sedeño P, Campuzano S, Pingarrón JM. Disposable immunoplatforms for the simultaneous determination of biomarkers for neurodegenerative disorders using poly(amidoamine) dendrimer/gold nanoparticle nanocomposite. Anal Bioanal Chem 2020; 413:799-811. [PMID: 32474723 DOI: 10.1007/s00216-020-02724-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/27/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022]
Abstract
Early diagnosis in primary care settings can increase access to therapies and their efficiency as well as reduce health care costs. In this context, we report in this paper the development of a disposable immunoplatform for the rapid and simultaneous determination of two protein biomarkers recently reported to be involved in the pathological process of neurodegenerative disorders (NDD), tau protein (tau), and TAR DNA-binding protein 43 (TDP-43). The methodology involves implementation of a sandwich-type immunoassay on the surface of dual screen-printed carbon electrodes (dSPCEs) electrochemically grafted with p-aminobenzoic acid (p-ABA), which allows the covalent immobilization of a gold nanoparticle-poly(amidoamine) (PAMAM) dendrimer nanocomposite (3D-Au-PAMAM). This scaffold was employed for the immobilization of the capture antibodies (CAbs). Detector antibodies labeled with horseradish peroxidase (HRP) and amperometric detection at - 0.20 V (vs. Ag pseudo-reference electrode) using the H2O2/hydroquinone (HQ) system were used. The developed methodology exhibits high sensitivity and selectivity for determining the target proteins, with detection limits of 2.3 and 12.8 pg mL-1 for tau and TDP-43, respectively. The simultaneous determination of tau and TDP-43 was accomplished in raw plasma samples and brain tissue extracts from healthy individuals and NDD-diagnosed patients. The analysis can be performed in just 1 h using a simple one-step assay protocol and small sample amounts (5 μL plasma and 2.5 μg brain tissue extracts). Graphical abstract.
Collapse
Affiliation(s)
- Verónica Serafín
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - Claudia A Razzino
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.,Institute of Research and Development, University of Vale do Paraiba, Sao Jose dos Campos, SP, 12244-000, Brazil
| | - Maria Gamella
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - María Pedrero
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Eloy Povedano
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - Ana Montero-Calle
- Chronic Disease Programme, UFIEC, Carlos III Health Institute, Majadahonda, 28220, Madrid, Spain
| | - Rodrigo Barderas
- Chronic Disease Programme, UFIEC, Carlos III Health Institute, Majadahonda, 28220, Madrid, Spain
| | - Miguel Calero
- Chronic Disease Programme, UFIEC, Carlos III Health Institute, Majadahonda, 28220, Madrid, Spain.,Alzheimer's Center Reina Sofía Foundation - CIEN Foundation and CIBERNED, Carlos III Institute of Health, Majadahonda, 28220, Madrid, Spain
| | - Anderson O Lobo
- LIMAV - Interdisciplinary Laboratory for Advanced Materials, BioMatLab, Department of Materials Engineering, Federal University of Piaui, Teresina, PI, 64049-550, Brazil
| | - Paloma Yáñez-Sedeño
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - Susana Campuzano
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
| | - José M Pingarrón
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| |
Collapse
|
24
|
De Plano LM, Carnazza S, Franco D, Rizzo MG, Conoci S, Petralia S, Nicoletti A, Zappia M, Campolo M, Esposito E, Cuzzocrea S, Guglielmino SPP. Innovative IgG Biomarkers Based on Phage Display Microbial Amyloid Mimotope for State and Stage Diagnosis in Alzheimer's Disease. ACS Chem Neurosci 2020; 11:1013-1026. [PMID: 32176482 PMCID: PMC7997372 DOI: 10.1021/acschemneuro.9b00549] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
![]()
An
innovative approach to identify new conformational antigens
of Aβ1–42 recognized by IgG autoantibodies
as biomarkers of state and stage in Alzheimer’s disease (AD)
patients is described. In particular, through the use of bioinformatics
modeling, conformational similarities between several Aβ1–42 forms and other amyloid-like proteins with F1 capsular
antigen (Caf1) of Yersinia pestis were first found.
pVIII M13 phage display libraries were then screened against YPF19,
anti-Caf1 monoclonal antibody, and IgGs of AD patients, in alternate
biopanning cycles of a so-called “double binding” selection.
From the selected phage clones, one, termed 12III1, was found to be
able to prevent in vitro Aβ1–42-induced cytotoxicity in SH-SY5Y cells, as well as to promote disaggregation
of preformed fibrils, to a greater extent with respect to wild-type
phage (pC89). IgG levels detected by 12III1 provided a significant
level of discrimination between diseased and nondemented subjects,
as well as a good correlation with the state progression of the disease.
These results give significant impact in AD state and stage diagnosis,
paving the way for the development not only for an innovative blood
diagnostic assay for AD precise diagnosis, progressive clinical assessment,
and screening but also for new effective treatments.
Collapse
Affiliation(s)
- Laura M. De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Santina Carnazza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Domenico Franco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Maria Giovanna Rizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Sabrina Conoci
- STmicroelectronics, Stradale Primosole, 50, 95121 Catania, Italy
- Distretto Tecnologico Micro e Nano Sistemi Sicilia, Strada VII-Zona Industriale, 95121 Catania, Italy
| | | | - Alessandra Nicoletti
- Neurology Clinic, Department “G.F. Ingrassia”, Section of Neurosciences, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Mario Zappia
- Neurology Clinic, Department “G.F. Ingrassia”, Section of Neurosciences, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Salvatore P. P. Guglielmino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
25
|
Medical Histories of Control Subjects Influence the Biomarker Potential of Plasma Aβ in Alzheimer's Disease: a Meta-analysis. J Mol Neurosci 2020; 70:861-870. [PMID: 32125624 DOI: 10.1007/s12031-020-01510-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/19/2020] [Indexed: 12/19/2022]
Abstract
Whether blood amyloid-β (Aβ) could be a peripheral biomarker of Alzheimer's disease (AD) remains in dispute. In the present study, we conducted a meta-analysis with 19 citations searched from Embase, PubMed, and the Cochrane Library database. Weighted mean difference (WMD) with 95% confidence intervals (CIs) was used to estimate the effect size. We firstly analyzed the plasma Aβ40, Aβ42, and Aβ42/Aβ40 ratio in AD and control group subjects. However, only a lower level of plasma Aβ42 was figured out in AD group subjects with weak statistical significance (WMD 1.82; 95% CI 0.59, 3.06; P = 0.004; I2 = 84%). We considered that the medical histories of control subjects could influence the biomarker ability of plasma Aβ. Therefore, subgroup analyses were then carried out based on a new recruiting criterion for control subjects, defining as no afflictions of any Aβ-related diseases. Surprisingly, AD group subjects showed a significant decrease in plasma Aβ42/Aβ40 ratio with low heterogeneity among studies (WMD 0.02; 95% CI 0.02, 0.02; P < 0.00001; I2 = 0%). Moreover, not only the Aβ42/Aβ40 ratio but also Aβ42 and Aβ40 were indifferent between AD and pseudo-control subjects which might be afflicted with Aβ-related diseases. This meta-analysis demonstrated that medical histories of control subjects were interference factors impeding plasma Aβ to be a biomarker of AD.
Collapse
|
26
|
O'Bryant SE, Ferman TJ, Zhang F, Hall J, Pedraza O, Wszolek ZK, Como T, Julovich D, Mattevada S, Johnson LA, Edwards M, Hall J, Graff-Radford NR. A proteomic signature for dementia with Lewy bodies. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2019; 11:270-276. [PMID: 30923734 PMCID: PMC6424013 DOI: 10.1016/j.dadm.2019.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION We sought to determine if a proteomic profile approach developed to detect Alzheimer's disease would distinguish patients with Lewy body disease from normal controls, and if it would distinguish dementia with Lewy bodies (DLB) from Parkinson's disease (PD). METHODS Stored plasma samples were obtained from 145 patients (DLB n = 57, PD without dementia n = 32, normal controls n = 56) enrolled from patients seen in the Behavioral Neurology or Movement Disorders clinics at the Mayo Clinic, Florida. Proteomic assays were conducted and analyzed as per our previously published protocols. RESULTS In the first step, the proteomic profile distinguished the DLB-PD group from controls with a diagnostic accuracy of 0.97, sensitivity of 0.91, and specificity of 0.86. In the second step, the proteomic profile distinguished the DLB from PD groups with a diagnostic accuracy of 0.92, sensitivity of 0.94, and specificity of 0.88. DISCUSSION These data provide evidence of the potential utility of a multitiered blood-based proteomic screening method for detecting DLB and distinguishing DLB from PD.
Collapse
Affiliation(s)
- Sid E. O'Bryant
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Tanis J. Ferman
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, USA
| | - Fan Zhang
- Vermont Genetics Network, University of Vermont, Burlington, VT, USA
| | - James Hall
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Otto Pedraza
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Tori Como
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - David Julovich
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Sravan Mattevada
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Leigh A. Johnson
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Melissa Edwards
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - James Hall
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | | |
Collapse
|
27
|
Brazaca LC, Moreto JR, Martín A, Tehrani F, Wang J, Zucolotto V. Colorimetric Paper-Based Immunosensor for Simultaneous Determination of Fetuin B and Clusterin toward Early Alzheimer's Diagnosis. ACS NANO 2019; 13:13325-13332. [PMID: 31661258 DOI: 10.1021/acsnano.9b06571] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Alzheimer's disease is a devastating condition characterized by a progressive and slow brain decay in elders. Here, we developed a paper-based lateral flow immunoassay for simultaneous and fast determination of Alzheimer's blood biomarkers, fetuin B and clusterin. Selective antibodies to targeted biomarkers were immobilized on gold nanoparticles (AuNPs) and deposited on paper pads. After adding the sample on the paper-based device, the biofluid laterally flows toward the selective antibody, permitting AuNP-Ab accumulation on the test zone, which causes a color change from white to pink. Image analysis was performed using a customized algorithm for the automatic recognition of the area of analysis and color clustering. Colorimetric detection was compared to electrochemical methods for the precise quantification of biomarkers. The best performance was found for the color parameter "L*". Good linearity (R2 = 0.988 and 0.998) and reproducibility (%RSD = 2.79% and 1.82%, N = 3) were demonstrated for the quantification of fetuin B and clusterin, respectively. Furthermore, the specificity of the immunosensor was tested on mixtures of proteins, showing negligible cross-reactivity and good performance in complex environments. We believe that our biosensor has a potential for early-stage diagnosis of Alzheimer's disease and toward a better understanding of Alzheimer's developing mechanisms.
Collapse
Affiliation(s)
- Laís C Brazaca
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics , University of São Paulo , 13560-970 São Carlos , SP , Brazil
- Department of NanoEngineering , University of California, San Diego , La Jolla , California 92093 , United States
| | - José R Moreto
- Department of Aerospace Engineering , San Diego State University , San Diego , California 92182-1308 , United States
| | - Aída Martín
- Department of NanoEngineering , University of California, San Diego , La Jolla , California 92093 , United States
| | - Farshad Tehrani
- Department of NanoEngineering , University of California, San Diego , La Jolla , California 92093 , United States
| | - Joseph Wang
- Department of NanoEngineering , University of California, San Diego , La Jolla , California 92093 , United States
| | - Valtencir Zucolotto
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics , University of São Paulo , 13560-970 São Carlos , SP , Brazil
| |
Collapse
|
28
|
Monocytes exposed to plasma from patients with Alzheimer’s disease undergo metabolic reprogramming. Neurosci Res 2019; 148:54-60. [PMID: 30641113 DOI: 10.1016/j.neures.2019.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/07/2018] [Accepted: 01/09/2019] [Indexed: 12/16/2022]
|
29
|
Eke CS, Jammeh E, Li X, Carroll C, Pearson S, Ifeachor E. Identification of Optimum Panel of Blood-based Biomarkers for Alzheimer's Disease Diagnosis Using Machine Learning. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2019; 2018:3991-3994. [PMID: 30441233 DOI: 10.1109/embc.2018.8513293] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
With the increasing number of people living with Alzheimer's disease (AD), there is a need for low-cost and easy to use methods to detect AD early to facilitate access to appropriate care pathways. Neuroimaging biomarkers (such as those based on PET and MRI) and biochemical biomarkers (such as those based on CSF) are recommended by international guidelines to facilitate diagnosis. However, neuroimaging is expensive and may not be widely available and CSF testing is invasive. Bloodbased biomarkers offer the potential for the development of a low-cost and more time efficient tool to detect AD to complement CSF and neuroimaging as blood is much easier to obtain. Although no single blood biomarker is yet able to detect AD, combinations of biomarkers (also called panels) have shown good results. However, a large number of biomarkers are often needed to achieve a satisfactory detection performance. In addition, it is difficult to reproduce reported results within and across different study cohorts because of data overfitting and lack of access to the datasets used in the studies. In this study, our focus is to identify an optimum panel (in terms of the least number of blood biomarkers to meet the specified diagnostic performance of 80% sensitivity and specificity) based on a widely accessible data set, and to demonstrate a testing methodology that reinforces reproducibility of results. Realizing a panel with reduced number of markers will have significant impact on the complexity and cost of diagnosis and potential development of cost-effective point of care devices.
Collapse
|
30
|
Rahman MR, Islam T, Zaman T, Shahjaman M, Karim MR, Huq F, Quinn JMW, Holsinger RMD, Gov E, Moni MA. Identification of molecular signatures and pathways to identify novel therapeutic targets in Alzheimer's disease: Insights from a systems biomedicine perspective. Genomics 2019; 112:1290-1299. [PMID: 31377428 DOI: 10.1016/j.ygeno.2019.07.018] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 07/01/2019] [Accepted: 07/30/2019] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. However, there are no peripheral biomarkers available that can detect AD onset. This study aimed to identify the molecular signatures in AD through an integrative analysis of blood gene expression data. We used two microarray datasets (GSE4226 and GSE4229) comparing peripheral blood transcriptomes of AD patients and controls to identify differentially expressed genes (DEGs). Gene set and protein overrepresentation analysis, protein-protein interaction (PPI), DEGs-Transcription Factors (TFs) interactions, DEGs-microRNAs (miRNAs) interactions, protein-drug interactions, and protein subcellular localizations analyses were performed on DEGs common to the datasets. We identified 25 common DEGs between the two datasets. Integration of genome scale transcriptome datasets with biomolecular networks revealed hub genes (NOL6, ATF3, TUBB, UQCRC1, CASP2, SND1, VCAM1, BTF3, VPS37B), common transcription factors (FOXC1, GATA2, NFIC, PPARG, USF2, YY1) and miRNAs (mir-20a-5p, mir-93-5p, mir-16-5p, let-7b-5p, mir-708-5p, mir-24-3p, mir-26b-5p, mir-17-5p, mir-193-3p, mir-186-5p). Evaluation of histone modifications revealed that hub genes possess several histone modification sites associated with AD. Protein-drug interactions revealed 10 compounds that affect the identified AD candidate biomolecules, including anti-neoplastic agents (Vinorelbine, Vincristine, Vinblastine, Epothilone D, Epothilone B, CYT997, and ZEN-012), a dermatological (Podofilox) and an immunosuppressive agent (Colchicine). The subcellular localization of molecular signatures varied, including nuclear, plasma membrane and cytosolic proteins. In the present study, it was identified blood-cell derived molecular signatures that might be useful as candidate peripheral biomarkers in AD. It was also identified potential drugs and epigenetic data associated with these molecules that may be useful in designing therapeutic approaches to ameliorate AD.
Collapse
Affiliation(s)
- Md Rezanur Rahman
- Department of Biochemistry and Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Enayetpur, Sirajgonj, Bangladesh.
| | - Tania Islam
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
| | - Toyfiquz Zaman
- Department of Biochemistry and Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Enayetpur, Sirajgonj, Bangladesh
| | - Md Shahjaman
- Department of Statistics, Begum Rokeya University, Rangpur, Bangladesh
| | - Md Rezaul Karim
- Department of Biochemistry and Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Enayetpur, Sirajgonj, Bangladesh
| | - Fazlul Huq
- Discipline of Pathology, School of Medical Sciences, The University of Sydney, NSW 2006, Australia
| | - Julian M W Quinn
- Bone Biology Division, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - R M Damian Holsinger
- Discipline of Pathology, School of Medical Sciences, The University of Sydney, NSW 2006, Australia; Laboratory of Molecular Neuroscience and Dementia, Brain and Mind Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Esra Gov
- Department of Bioengineering, Adana Alparslan Turkes Science and Technology University, Adana, Turkey
| | - Mohammad Ali Moni
- Discipline of Pathology, School of Medical Sciences, The University of Sydney, NSW 2006, Australia; Bone Biology Division, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia.
| |
Collapse
|
31
|
Shin HY, Kim JH, Jahng GH, Jung WS, Park SU, Ko CN, Park JM. The effectiveness and safety of Kami Guibi-tang for mild cognitive impairment: study protocol of a pilot, randomized, placebo-controlled, double-blind trial. Trials 2019; 20:448. [PMID: 31331367 PMCID: PMC6647292 DOI: 10.1186/s13063-019-3567-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 07/10/2019] [Indexed: 12/03/2022] Open
Abstract
Background Mild cognitive impairment (MCI) is an intermediate phase between normal aging and dementia. Since a majority of amnestic MCI (aMCI) cases progress to Alzheimer’s disease (AD), it is considered the prodromal stage of AD and, therefore, a treatment target for the prevention of further cognitive decline. However, there is no approved treatment for MCI at present. Kami Guibi-tang (KGT) is a herbal drug used in Korean medicine to treat amnesia, insomnia, loss of appetite, and depression. We will explore the effectiveness and safety of KGT in amnestic MCI in this trial. Methods/design The study will be a single-center, randomized, placebo-controlled, double-blind trial. Eligible participants diagnosed with amnestic MCI will be randomly allocated to a treatment or control group. Participants will take KGT or placebo granules, three times a day, for 24 weeks. The primary outcomes will be changes in Seoul Neuropsychological Screening Battery (SNSB) scores, and magnetic resonance imaging (MRI) measurements including those of brain metabolites, neurotransmitters, and cerebral blood flow. The secondary outcomes will include the safety assessment, measured by changes in blood chemistry, changes in blood protein and cholesterol levels related to AD pathology, and a comparison of MRI changes between the two groups, using age and genotype as covariates. Discussion This study will be the first clinical trial to identify the therapeutic potential of Kami Guibi-tang for amnestic MCI. The findings will provide insight into the feasibility of large-scale trials to gather evidence for KGT as a treatment for MCI. Trial registration Korean Clinical Trial Registry, ID: KCT0002407. Registered on 30 March 2017. Electronic supplementary material The online version of this article (10.1186/s13063-019-3567-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hee-Yeon Shin
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,Stroke and Neurological Disorders Center, Kyung Hee University Hospital at Gangdong, 892, Dongnam-ro, Gangdong-gu, Seoul, 05278, Republic of Korea
| | - Jeong-Hwa Kim
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,Stroke and Neurological Disorders Center, Kyung Hee University Hospital at Gangdong, 892, Dongnam-ro, Gangdong-gu, Seoul, 05278, Republic of Korea
| | - Geon-Ho Jahng
- Department of Radiology, College of Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University, #892, Dongnam-ro, Gangdong-gu, Seoul, 05278, Republic of Korea
| | - Woo-Sang Jung
- Department of Cardiology and Neurology, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Seong-Uk Park
- Department of Cardiology and Neurology, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,Stroke and Neurological Disorders Center, Kyung Hee University Hospital at Gangdong, 892, Dongnam-ro, Gangdong-gu, Seoul, 05278, Republic of Korea
| | - Chang-Nam Ko
- Department of Cardiology and Neurology, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,Stroke and Neurological Disorders Center, Kyung Hee University Hospital at Gangdong, 892, Dongnam-ro, Gangdong-gu, Seoul, 05278, Republic of Korea
| | - Jung-Mi Park
- Department of Cardiology and Neurology, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea. .,Stroke and Neurological Disorders Center, Kyung Hee University Hospital at Gangdong, 892, Dongnam-ro, Gangdong-gu, Seoul, 05278, Republic of Korea.
| |
Collapse
|
32
|
O'Bryant SE, Edwards M, Zhang F, Johnson LA, Hall J, Kuras Y, Scherzer CR. Potential two-step proteomic signature for Parkinson's disease: Pilot analysis in the Harvard Biomarkers Study. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2019; 11:374-382. [PMID: 31080873 PMCID: PMC6502745 DOI: 10.1016/j.dadm.2019.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction We sought to determine if our previously validated proteomic profile for detecting Alzheimer's disease would detect Parkinson's disease (PD) and distinguish PD from other neurodegenerative diseases. Methods Plasma samples were assayed from 150 patients of the Harvard Biomarkers Study (PD, n = 50; other neurodegenerative diseases, n = 50; healthy controls, n = 50) using electrochemiluminescence and Simoa platforms. Results The first step proteomic profile distinguished neurodegenerative diseases from controls with a diagnostic accuracy of 0.94. The second step profile distinguished PD cases from other neurodegenerative diseases with a diagnostic accuracy of 0.98. The proteomic profile differed in step 1 versus step 2, suggesting that a multistep proteomic profile algorithm to detecting and distinguishing between neurodegenerative diseases may be optimal. Discussion These data provide evidence of the potential use of a multitiered blood-based proteomic screening method for detecting individuals with neurodegenerative disease and then distinguishing PD from other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sid E O'Bryant
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Melissa Edwards
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Fan Zhang
- Vermont Genetics Network, University of Vermont, Burlington, VT, USA
| | - Leigh A Johnson
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - James Hall
- Institute for Translational Research, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Yuliya Kuras
- Advanced Center for Parkinson's Disease Research of Brigham & Women's Hospital, and Harvard Medical School, Boston, MA, USA.,Precision Neurology Program, Harvard Medical School, Brigham & Women's Hospital, Boston, MA, USA
| | - Clemens R Scherzer
- Advanced Center for Parkinson's Disease Research of Brigham & Women's Hospital, and Harvard Medical School, Boston, MA, USA.,Precision Neurology Program, Harvard Medical School, Brigham & Women's Hospital, Boston, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
33
|
Kim H, Lee JU, Kim S, Song S, Sim SJ. A Nanoplasmonic Biosensor for Ultrasensitive Detection of Alzheimer's Disease Biomarker Using a Chaotropic Agent. ACS Sens 2019; 4:595-602. [PMID: 30747516 DOI: 10.1021/acssensors.8b01242] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Blood-based diagnosis (hemodiagnosis) of Alzheimer's disease (AD) is emerging as a promising alternative to cerebrospinal-fluid-based methods because blood contains various kinds of AD biomarkers, including amyloid beta 1-40, 1-42, and τ (tau) protein. However, with current technology, the accuracy of the blood-plasma-based methods is relatively low compared to the traditional methods because the concentration of AD biomarkers in blood plasma is incredibly low, and diverse interference is present in blood plasma, which hinders precise detection. Here, we suggest a nanoplasmonic biosensor using gold nanorods with a chaotropic agent for precise ultrasensitive detecting of Alzheimer's disease biomarkers in human plasma. This nanoplasmonic biosensor is based on the localized surface plasmon resonance (LSPR), which is extremely sensitive to the point where it can respond to an insignificant change of the refractive index around the gold nanoparticles. Also, using guanidine hydrochloride as a chaotropic agent, we can overcome the obstacles of blood-based AD diagnostics. In more detail, this agent interrupts the network between water molecules and weakens the hydrophobic interactions between proteins, remarkably improving detection capabilities to target τ protein. By reducing the overlapping ranges between protein levels in an age-matched control and AD patients' plasma, this system can accurately diagnose AD patients. This platform also can analyze disease from mild cognitive impairment using standardized blood biomarker tau protein, which is related to Alzheimer's disease. As a result, our platform can be applied to clinical trials, and thus it has excellent potential in the medical field.
Collapse
Affiliation(s)
- Hanbi Kim
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, South Korea
| | - Jong Uk Lee
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, South Korea
| | - Soohyun Kim
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, South Korea
| | - Sojin Song
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, South Korea
| | - Sang Jun Sim
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, South Korea
| |
Collapse
|
34
|
Martí-Juan G, Sanroma G, Piella G. Revealing heterogeneity of brain imaging phenotypes in Alzheimer's disease based on unsupervised clustering of blood marker profiles. PLoS One 2019; 14:e0211121. [PMID: 30830917 PMCID: PMC6398858 DOI: 10.1371/journal.pone.0211121] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 01/08/2019] [Indexed: 01/02/2023] Open
Abstract
Alzheimer's disease (AD) affects millions of people and is a major rising problem in health care worldwide. Recent research suggests that AD could have different subtypes, presenting differences in how the disease develops. Characterizing those subtypes could be key to deepen the understanding of this complex disease. In this paper, we used a multivariate, non-supervised clustering method over blood-based markers to find subgroups of patients defined by distinctive blood marker profiles. Our analysis on ADNI database identified 4 possible subgroups, each with a different blood profile. More importantly, we show that subgroups with different profiles have a different relationship between brain phenotypes detected in magnetic resonance imaging and disease condition.
Collapse
Affiliation(s)
- Gerard Martí-Juan
- BCN Medtech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Gerard Sanroma
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Gemma Piella
- BCN Medtech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | | |
Collapse
|
35
|
Hampel H, O'Bryant SE, Molinuevo JL, Zetterberg H, Masters CL, Lista S, Kiddle SJ, Batrla R, Blennow K. Blood-based biomarkers for Alzheimer disease: mapping the road to the clinic. Nat Rev Neurol 2018; 14:639-652. [PMID: 30297701 PMCID: PMC6211654 DOI: 10.1038/s41582-018-0079-7] [Citation(s) in RCA: 445] [Impact Index Per Article: 63.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Biomarker discovery and development for clinical research, diagnostics and therapy monitoring in clinical trials have advanced rapidly in key areas of medicine - most notably, oncology and cardiovascular diseases - allowing rapid early detection and supporting the evolution of biomarker-guided, precision-medicine-based targeted therapies. In Alzheimer disease (AD), breakthroughs in biomarker identification and validation include cerebrospinal fluid and PET markers of amyloid-β and tau proteins, which are highly accurate in detecting the presence of AD-associated pathophysiological and neuropathological changes. However, the high cost, insufficient accessibility and/or invasiveness of these assays limit their use as viable first-line tools for detecting patterns of pathophysiology. Therefore, a multistage, tiered approach is needed, prioritizing development of an initial screen to exclude from these tests the high numbers of people with cognitive deficits who do not demonstrate evidence of underlying AD pathophysiology. This Review summarizes the efforts of an international working group that aimed to survey the current landscape of blood-based AD biomarkers and outlines operational steps for an effective academic-industry co-development pathway from identification and assay development to validation for clinical use.
Collapse
Affiliation(s)
- Harald Hampel
- AXA Research Fund and Sorbonne University Chair, Paris, France.
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France.
- Brain & Spine Institute (ICM), INSERM U 1127, Paris, France.
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, AP-HP, Pitié-Salpêtrière Hospital, Paris, France.
| | - Sid E O'Bryant
- University of North Texas Health Science Center, Fort Worth, TX, USA
| | - José L Molinuevo
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Melbourne, Australia
| | - Simone Lista
- AXA Research Fund and Sorbonne University Chair, Paris, France
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, Paris, France
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Steven J Kiddle
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | | | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| |
Collapse
|
36
|
Abstract
The past decade has seen tremendous efforts in biomarker discovery and validation for neurodegenerative diseases. The source and type of biomarkers has continued to grow for central nervous system diseases, from biofluid-based biomarkers (blood or cerebrospinal fluid (CSF)), to nucleic acids, tissue, and imaging. While DNA remains a predominant biomarker used to identify familial forms of neurodegenerative diseases, various types of RNA have more recently been linked to familial and sporadic forms of neurodegenerative diseases during the past few years. Imaging approaches continue to evolve and are making major contributions to target engagement and early diagnostic biomarkers. Incorporation of biomarkers into drug development and clinical trials for neurodegenerative diseases promises to aid in the development and demonstration of target engagement and drug efficacy for neurologic disorders. This review will focus on recent advancements in developing biomarkers for clinical utility in Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS).
Collapse
Affiliation(s)
| | - Robert Bowser
- Iron Horse Diagnostics, Inc., Scottsdale, AZ, 85255, USA.
- Divisions of Neurology and Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ, 85013, USA.
| |
Collapse
|
37
|
Maeba R, Araki A, Fujiwara Y. Serum Ethanolamine Plasmalogen and Urine Myo-Inositol as Cognitive Decline Markers. Adv Clin Chem 2018; 87:69-111. [PMID: 30342713 DOI: 10.1016/bs.acc.2018.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent studies have suggested that metabolic disorders, particularly type 2 diabetes mellitus (T2DM), and dementia, including Alzheimer's disease (AD), were linked at the clinical and molecular levels. Brain insulin deficiency and resistance may be key events in AD pathology mechanistically linking AD to T2DM. Ethanolamine plasmalogens (PlsEtns) are abundant in the brain and play essential roles in neuronal function and myelin formation. As such, PlsEtn deficiency may be pathologically relevant in some neurodegenerative disorders such as AD. Decreased brain PlsEtn associated with dementia may reflect serum PlsEtn deficiency. We hypothesized that myo-inositol plays a role in myelin formation through its facilitation of PlsEtn biosynthesis. Excessive urinary myo-inositol (UMI) loss would likely result in PlsEtn deficiency potentially leading to demyelinating diseases such as dementia. Accordingly, measurement of both serum PlsEtn and baseline UMI excretion could improve the detection of cognitive impairment (CI) in a more specific and reliable manner. To verify our hypothesis, we conducted a clinical observational study of memory clinic outpatients (MCO) and cognitively normal elderly (NE) for nearly 4.5years. We demonstrated that serum PlsEtn concentration associated with UMI excretion was useful for predicting advancing dementia in patients with mild CI. Because hyperglycemia and associated insulin resistance might be a leading cause of increased baseline UMI excretion, serum PlsEtn quantitation would be useful in detecting CI among the elderly with hyperglycemia. Our findings suggest that myo-inositol is a novel candidate molecule linking T2DM to AD.
Collapse
Affiliation(s)
- Ryouta Maeba
- Department of Biochemistry, Teikyo University School of Medicine, Tokyo, Japan
| | - Atsushi Araki
- Department of Diabetes, Metabolism and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Yoshinori Fujiwara
- Research Team for Social Participation and Community Health, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
38
|
Lista S, Zetterberg H, O'Bryant SE, Blennow K, Hampel H. Evolving Relevance of Neuroproteomics in Alzheimer's Disease. Methods Mol Biol 2018; 1598:101-115. [PMID: 28508359 DOI: 10.1007/978-1-4939-6952-4_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Substantial progress in the understanding of the biology of Alzheimer's disease (AD) has been achieved over the past decades. The early detection and diagnosis of AD and other age-related neurodegenerative diseases, however, remain a challenging scientific frontier. Therefore, the comprehensive discovery (relating to all individual, converging or diverging biochemical disease mechanisms), development, validation, and qualification of standardized biological markers with diagnostic and prognostic functions with a precise performance profile regarding specificity, sensitivity, and positive and negative predictive value are warranted.Methodological innovations in the area of exploratory high-throughput technologies, such as sequencing, microarrays, and mass spectrometry-based analyses of proteins/peptides, have led to the generation of large global molecular datasets from a multiplicity of biological systems, such as biological fluids, cells, tissues, and organs. Such methodological progress has shifted the attention to the execution of hypothesis-independent comprehensive exploratory analyses (opposed to the classical hypothesis-driven candidate approach), with the aim of fully understanding the biological systems in physiology and disease as a whole. The systems biology paradigm integrates experimental biology with accurate and rigorous computational modelling to describe and foresee the dynamic features of biological systems. The use of dynamically evolving technological platforms, including mass spectrometry, in the area of proteomics has enabled to rush the process of biomarker discovery and validation for refining significantly the diagnosis of AD. Currently, proteomics-which is part of the systems biology paradigm-is designated as one of the dominant matured sciences needed for the effective exploratory discovery of prospective biomarker candidates expected to play an effective role in aiding the early detection, diagnosis, prognosis, and therapy development in AD.
Collapse
Affiliation(s)
- Simone Lista
- AXA Research Fund & UPMC Chair, Paris, France. .,Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et dela moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), HôpitalPitié-Salpêtrière, Boulevard de l'hôpital, F-75013, Paris, France.
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Sid E O'Bryant
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences, Stockholm, Sweden
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A) & Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France; Département de Neurologie, Hôpital de la Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013, Paris, France
| |
Collapse
|
39
|
A shape-code nanoplasmonic biosensor for multiplex detection of Alzheimer's disease biomarkers. Biosens Bioelectron 2018; 101:96-102. [DOI: 10.1016/j.bios.2017.10.018] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/09/2017] [Indexed: 12/19/2022]
|
40
|
Islam K, Damiati S, Sethi J, Suhail A, Pan G. Development of a Label-Free Immunosensor for Clusterin Detection as an Alzheimer's Biomarker. SENSORS (BASEL, SWITZERLAND) 2018; 18:E308. [PMID: 29361679 PMCID: PMC5795331 DOI: 10.3390/s18010308] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/10/2018] [Accepted: 01/18/2018] [Indexed: 01/01/2023]
Abstract
Clusterin (CLU) has been associated with the clinical progression of Alzheimer's disease (AD) and described as a potential AD biomarker in blood plasma. Due to the enormous attention given to cerebrospinal fluid (CSF) biomarkers for the past couple of decades, recently found blood-based AD biomarkers like CLU have not yet been reported for biosensors. Herein, we report the electrochemical detection of CLU for the first time using a screen-printed carbon electrode (SPCE) modified with 1-pyrenebutyric acid N-hydroxysuccinimide ester (Pyr-NHS) and decorated with specific anti-CLU antibody fragments. This bifunctional linker molecule contains succinylimide ester to bind protein at one end while its pyrene moiety attaches to the carbon surface by means of π-π stacking. Cyclic voltammetric and square wave voltammetric studies showed the limit of detection down to 1 pg/mL and a linear concentration range of 1-100 pg/mL with good sensitivity. Detection of CLU in spiked human plasma was demonstrated with satisfactory recovery percentages to that of the calibration data. The proposed method facilitates the cost-effective and viable production of label-free point-of-care devices for the clinical diagnosis of AD.
Collapse
Affiliation(s)
- Kamrul Islam
- Wolfson Nanomaterials & Devices Laboratory, School of Computing, Electronics and Mathematics, Faculty of Science and Engineering, University of Plymouth, Devon PL4 8AA, UK.
| | - Samar Damiati
- Department of Biochemistry, Faculty of Science, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia.
| | - Jagriti Sethi
- Wolfson Nanomaterials & Devices Laboratory, School of Computing, Electronics and Mathematics, Faculty of Science and Engineering, University of Plymouth, Devon PL4 8AA, UK.
| | - Ahmed Suhail
- Wolfson Nanomaterials & Devices Laboratory, School of Computing, Electronics and Mathematics, Faculty of Science and Engineering, University of Plymouth, Devon PL4 8AA, UK.
| | - Genhua Pan
- Wolfson Nanomaterials & Devices Laboratory, School of Computing, Electronics and Mathematics, Faculty of Science and Engineering, University of Plymouth, Devon PL4 8AA, UK.
| |
Collapse
|
41
|
Henriques AD, Benedet AL, Camargos EF, Rosa-Neto P, Nóbrega OT. Fluid and imaging biomarkers for Alzheimer's disease: Where we stand and where to head to. Exp Gerontol 2018; 107:169-177. [PMID: 29307736 DOI: 10.1016/j.exger.2018.01.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 12/29/2017] [Accepted: 01/02/2018] [Indexed: 10/18/2022]
Abstract
There is increasing evidence that a number of potentially informative biomarkers for Alzheimer disease (AD) can improve the accuracy of diagnosing this form of dementia, especially when used as a panel of diagnostic assays and interpreted in the context of neuroimaging and clinical data. Moreover, by combining the power of CSF biomarkers with neuroimaging techniques to visualize Aβ deposits (or neurodegenerative lesions), it might be possible to better identify individuals at greatest risk for developing MCI and converting to AD. The objective of this article was to review recent progress in selected imaging and chemical biomarkers for prediction, early diagnosis and progression of AD. We present our view point of a scenario that places CSF and imaging markers on the verge of general utility based on accuracy levels that already match (or even surpass) current clinical precision.
Collapse
Affiliation(s)
- Adriane Dallanora Henriques
- Medical Centre for the Elderly, University Hospital, University of Brasília (UnB), 70910-900 Brasília, DF, Brazil
| | - Andrea Lessa Benedet
- Translational Neuroimaging Laboratory, Research Centre for Studies in Aging, Douglas Hospital, McGill University, H4H 1R3 Montreal, QC, Canada
| | - Einstein Francisco Camargos
- Medical Centre for the Elderly, University Hospital, University of Brasília (UnB), 70910-900 Brasília, DF, Brazil
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, Research Centre for Studies in Aging, Douglas Hospital, McGill University, H4H 1R3 Montreal, QC, Canada; Montreal Neurological Institute, H3A 2B4 Montreal, QC, Canada
| | - Otávio Toledo Nóbrega
- Medical Centre for the Elderly, University Hospital, University of Brasília (UnB), 70910-900 Brasília, DF, Brazil.
| |
Collapse
|
42
|
Shi L, Baird AL, Westwood S, Hye A, Dobson R, Thambisetty M, Lovestone S. A Decade of Blood Biomarkers for Alzheimer's Disease Research: An Evolving Field, Improving Study Designs, and the Challenge of Replication. J Alzheimers Dis 2018; 62:1181-1198. [PMID: 29562526 PMCID: PMC5870012 DOI: 10.3233/jad-170531] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2017] [Indexed: 12/22/2022]
Abstract
Blood-based biomarkers represent a less invasive and potentially cheaper approach for aiding Alzheimer's disease (AD) detection compared with cerebrospinal fluid and some neuroimaging biomarkers. Acknowledging that many in the field have made great progress, here we review some of the work that our group has pursued to identify and validate blood-based proteomic biomarkers through both case control and AD pathology endophenotype-based approaches. Our focus is primarily to identify a minimally invasive and hopefully cost-effective blood-based biomarker to reduce screen failure in clinical trials where participants have prodromal or even pre-clinical disease. We summarize some of the key findings and approaches taken in these biomarker studies, while addressing the main challenges, including that of limited replication in the field, and discuss opportunities for biomarker development.
Collapse
Affiliation(s)
- Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, UK
| | | | - Sarah Westwood
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Abdul Hye
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, and NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Richard Dobson
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, and NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Madhav Thambisetty
- Unit of Clinical and Translational Neuroscience, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | | |
Collapse
|
43
|
Metallomics Applied to the Study of Neurodegenerative and Mental Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1055:21-37. [PMID: 29884960 DOI: 10.1007/978-3-319-90143-5_2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Biochemical imbalances, provoked by aging or a secondary illness, might directly affect the brain, causing severe problems, such as loss of memory or alteration of behavior patterns. Brain disorders are usually classified as injuries (such as stroke, hematomas, and concussions), tumors, and neurodegenerative (such as Parkinson's and Alzheimer's diseases) and mental (such as depression, bipolar disorder, schizophrenia) diseases. As the pathophysiology of these illnesses is not completely established and multiple factors are involved, metallomics, a bioanalytical strategy that allows the detection of metal ions and metalloproteins in diverse biological matrices, is of extreme relevance in identifying which elements are affected by a disease and/or treatment. Thus, determining which element ions suffer disturbances in their homeostasis during the disease progress is relevant to understand the biochemical changes and propose new drug targets. In addition, it is well known that oxidative stress plays an important role in the development of pathological neurodegenerative and mental diseases, which may be caused by metal ion dyshomeostasis, so it is also important to understand endogenous antioxidant metalloprotein and metalloenzyme mechanisms in this regard. In this context, recent applications of metallomics in the study of neurodegenerative and mental disorders are discussed in this chapter, as well as future trends in this research area.
Collapse
|
44
|
Guo R, Fan G, Zhang J, Wu C, Du Y, Ye H, Li Z, Wang L, Zhang Z, Zhang L, Zhao Y, Lu Z. A 9-microRNA Signature in Serum Serves as a Noninvasive Biomarker in Early Diagnosis of Alzheimer’s Disease. J Alzheimers Dis 2017; 60:1365-1377. [PMID: 29036818 DOI: 10.3233/jad-170343] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Rui Guo
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P. R. China
| | - Gang Fan
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P. R. China
| | - Jian Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P. R. China
| | - Chunxiao Wu
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P. R. China
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P. R. China
| | - Hui Ye
- Department of Quality Management, Blood Center of Shandong Province, Jinan, P. R. China
| | - Zhang Li
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P. R. China
| | - Lili Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P. R. China
| | - Zhihui Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P. R. China
| | - Lu Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P. R. China
| | - Yueran Zhao
- Department of Central Laboratory, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P. R. China
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, Jinan, P. R. China
| |
Collapse
|
45
|
Platelet phosphorylated TDP-43: an exploratory study for a peripheral surrogate biomarker development for Alzheimer's disease. Future Sci OA 2017; 3:FSO238. [PMID: 29134122 PMCID: PMC5674277 DOI: 10.4155/fsoa-2017-0090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022] Open
Abstract
Aim: Alzheimer's disease (AD) and other forms of dementia create a noncurable disease population in world's societies. To develop a blood-based biomarker is important so that the remedial or disease-altering therapeutic intervention for AD patients would be available at the early stage. Materials & methods: TDP-43 levels were analyzed in postmortem brain tissue and platelets of AD and control subjects. Results: We observed an increased TDP-43 (<60%) in postmortem AD brain regions and similar trends were also observed in patient's platelets. Conclusion: Platelet TDP-43 could be used as a surrogate biomarker that is measurable, reproducible and sensitive for screening the patients with some early clinical signs of AD and can be used to monitor disease prognosis. In this study, we explore to identify an Alzheimer's disease (AD)-selective phospho-specific antibody that recognizes the diseased form of TDP-43 protein in patient's blood-derived platelets. Our results suggest that selective antiphosphorylated TDP-43 antibody discriminates AD from non-demented controls and patients with amyotrophic lateral sclerosis. Therefore, platelet screening with a selective antibody could potentially be a useful tool for diagnostic purposes for AD.
Collapse
|
46
|
Lopes J, Correia M, Martins I, Henriques AG, Delgadillo I, da Cruz E Silva O, Nunes A. FTIR and Raman Spectroscopy Applied to Dementia Diagnosis Through Analysis of Biological Fluids. J Alzheimers Dis 2017; 52:801-12. [PMID: 27079713 DOI: 10.3233/jad-151163] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
To date, it is still difficult to perform an early and accurate diagnosis of dementia, therefore significant research has focused on finding new dementia biomarkers that can aid in this respect. There is an urgent need for non-invasive, rapid, and relatively inexpensive procedures for early diagnostics. Studies have demonstrated that of spectroscopic techniques, such as Fourier Transform Infrared Spectroscopy (FTIR) and Raman Spectroscopy could be a useful and accurate procedure to diagnose dementia. Given that several biochemical mechanisms related to neurodegeneration and dementia can lead to changes in plasma components and others peripheral body fluids; blood-based samples coupled to spectroscopic analyses can be used as a simple and less invasive approach.
Collapse
Affiliation(s)
- Jéssica Lopes
- iBiMED, Departamento de Ciências Médicas, Universidade de Aveiro, Aveiro, Portugal
| | - Marta Correia
- iBiMED, Departamento de Ciências Médicas, Universidade de Aveiro, Aveiro, Portugal
| | - Ilka Martins
- Grupo de Neurociências e Sinalização, iBiMED, Departamento de Ciências Médicas, Universidade de Aveiro, Aveiro, Portugal
| | - Ana Gabriela Henriques
- Grupo de Neurociências e Sinalização, iBiMED, Departamento de Ciências Médicas, Universidade de Aveiro, Aveiro, Portugal
| | - Ivonne Delgadillo
- QOPNA, Departamento de Química, Universidade de Aveiro, Aveiro, Portugal
| | - Odete da Cruz E Silva
- Grupo de Neurociências e Sinalização, iBiMED, Departamento de Ciências Médicas, Universidade de Aveiro, Aveiro, Portugal
| | - Alexandra Nunes
- iBiMED, Departamento de Ciências Médicas, Universidade de Aveiro, Aveiro, Portugal
| |
Collapse
|
47
|
Huynh RA, Mohan C. Alzheimer's Disease: Biomarkers in the Genome, Blood, and Cerebrospinal Fluid. Front Neurol 2017; 8:102. [PMID: 28373857 PMCID: PMC5357660 DOI: 10.3389/fneur.2017.00102] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 03/01/2017] [Indexed: 01/20/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that slowly destroys memory and thinking skills, resulting in behavioral changes. It is estimated that nearly 36 million are affected globally with numbers reaching 115 million by 2050. AD can only be definitively diagnosed at autopsy since its manifestations of senile plaques and neurofibrillary tangles throughout the brain cannot yet be fully captured with current imaging technologies. Current AD therapeutics have also been suboptimal. Besides identifying markers that distinguish AD from controls, there has been a recent drive to identify better biomarkers that can predict the rates of cognitive decline and neocortical amyloid burden in those who exhibit preclinical, prodromal, or clinical AD. This review covers biomarkers of three main types: genes, cerebrospinal fluid-derived, and blood-derived biomarkers. Looking ahead, cutting-edge OMICs technologies, including proteomics and metabolomics, ought to be fully tapped in order to mine even better biomarkers for AD that are more predictive.
Collapse
Affiliation(s)
- Rose Ann Huynh
- Department of Biomedical Engineering, University of Houston , Houston, TX , USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston , Houston, TX , USA
| |
Collapse
|
48
|
Abe Y, Kimura N, Takahashi R, Gotou M, Mizukami K, Uchida H, Matsubara E. Relationship between cytokine levels in the cerebrospinal fluid and 11C-Pittsburgh compound B retention in patients with mild cognitive impairment. Geriatr Gerontol Int 2017; 17:1907-1913. [PMID: 28261965 DOI: 10.1111/ggi.12991] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 11/01/2016] [Accepted: 11/22/2016] [Indexed: 01/05/2023]
Abstract
AIM In the present study, we examined the relationship between cytokine levels in the cerebrospinal fluid (CSF) and 11 C-Pittsburgh compound B (PiB) retention in patients with mild cognitive impairment. METHODS A total of 33 participants (12 men and 21 women; mean age 76.5 years) with mild cognitive impairment underwent neuropsychological assessments, PiB positron emission tomography and analysis of cytokine levels in the CSF. The CSF levels of 48 cytokines and growth factors were measured using multiplex immunoassays. PiB retention was assessed based on a standardized uptake value ratio. Mild cognitive impairment participants were classified as PiB-positive and PiB-negative, with a cut-off level of 1.4. We compared the CSF cytokine levels and Alzheimer's disease biomarkers, including β-amyloid 1-42, total tau and tau phosphorylated at threonine 181, between the two subgroups, and evaluated the correlation between PiB retention or CSF Alzheimer's disease biomarkers and CSF cytokine levels. RESULTS Cytokine levels in the CSF did not differ between the two subgroups. Macrophage inflammatory protein-1β levels in the CSF significantly correlated with PiB retention only in the PiB-positive subgroup, whereas stem cell growth factor-β levels significantly correlated with PiB retention in the PiB-negative subgroup. Furthermore, stem cell growth factor-β levels significantly correlated with total tau and tau phosphorylated at threonine 181 levels in only the PiB-negative subgroup. CONCLUSION The present findings suggest that macrophage inflammatory protein-1β and stem cell growth factor-β are associated with chronic inflammatory processes accompanied by amyloid deposition or AD pathophysiology. Geriatr Gerontol Int 2017; 17: 1907-1913.
Collapse
Affiliation(s)
- Yoshitake Abe
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| | - Noriyuki Kimura
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| | - Ryuichi Takahashi
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan.,Department of Neurology, Hyogo Prefectural Rehabilitation Hospital, Hyougo, Japan
| | - Megumi Gotou
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| | - Ken Mizukami
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| | - Hirotatsu Uchida
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| | - Etsuro Matsubara
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| |
Collapse
|
49
|
Counts SE, Ikonomovic MD, Mercado N, Vega IE, Mufson EJ. Biomarkers for the Early Detection and Progression of Alzheimer's Disease. Neurotherapeutics 2017; 14:35-53. [PMID: 27738903 PMCID: PMC5233625 DOI: 10.1007/s13311-016-0481-z] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The recent failures of potential disease-modifying drugs for Alzheimer's disease (AD) may reflect the fact that the enrolled participants in clinical trials are already too advanced to derive a clinical benefit. Thus, well-validated biomarkers for the early detection and accurate diagnosis of the preclinical stages of AD will be crucial for therapeutic advancement. The combinatorial use of biomarkers derived from biological fluids, such as cerebrospinal fluid (CSF), with advanced molecular imaging and neuropsychological testing may eventually achieve the diagnostic sensitivity and specificity necessary to identify people in the earliest stages of the disease when drug modification is most likely possible. In this regard, positive amyloid or tau tracer retention on positron emission tomography imaging, low CSF concentrations of the amyloid-β 1-42 peptide, high CSF concentrations in total tau and phospho-tau, mesial temporal lobe atrophy on magnetic resonance imaging, and temporoparietal/precuneus hypometabolism or hypoperfusion on 18F-fluorodeoxyglucose positron emission tomography have all emerged as biomarkers for the progression to AD. However, the ultimate AD biomarker panel will likely involve the inclusion of novel CSF and blood biomarkers more precisely associated with confirmed pathophysiologic mechanisms to improve its reliability for detecting preclinical AD. This review highlights advancements in biological fluid and imaging biomarkers that are moving the field towards achieving the goal of a preclinical detection of AD.
Collapse
Affiliation(s)
- Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Family Medicine, Michigan State University, Grand Rapids, MI, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, USA
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Natosha Mercado
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Irving E Vega
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Elliott J Mufson
- Department of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA.
| |
Collapse
|
50
|
O'Bryant SE, Mielke MM, Rissman RA, Lista S, Vanderstichele H, Zetterberg H, Lewczuk P, Posner H, Hall J, Johnson L, Fong YL, Luthman J, Jeromin A, Batrla-Utermann R, Villarreal A, Britton G, Snyder PJ, Henriksen K, Grammas P, Gupta V, Martins R, Hampel H. Blood-based biomarkers in Alzheimer disease: Current state of the science and a novel collaborative paradigm for advancing from discovery to clinic. Alzheimers Dement 2017; 13:45-58. [PMID: 27870940 PMCID: PMC5218961 DOI: 10.1016/j.jalz.2016.09.014] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 09/27/2016] [Indexed: 11/25/2022]
Abstract
The last decade has seen a substantial increase in research focused on the identification of blood-based biomarkers that have utility in Alzheimer's disease (AD). Blood-based biomarkers have significant advantages of being time- and cost-efficient as well as reduced invasiveness and increased patient acceptance. Despite these advantages and increased research efforts, the field has been hampered by lack of reproducibility and an unclear path for moving basic discovery toward clinical utilization. Here we reviewed the recent literature on blood-based biomarkers in AD to provide a current state of the art. In addition, a collaborative model is proposed that leverages academic and industry strengths to facilitate the field in moving past discovery only work and toward clinical use. Key resources are provided. This new public-private partnership model is intended to circumvent the traditional handoff model and provide a clear and useful paradigm for the advancement of biomarker science in AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sid E O'Bryant
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA.
| | - Michelle M Mielke
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Robert A Rissman
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, UCSD School of Medicine, La Jolla, CA, USA
| | - Simone Lista
- AXA Research Fund and UPMC Chair, Paris, France; Department de Neurologie, Institut de la Memorie et de la Maladie d'Alzheimer (IM2A) et Institut du Cerveau et du la Moelle epiniere (ICM), Hospital de la Pitie-Salpetriere, Sorbonne Universites, Universite Pierre et Marie Curie, Paris, France
| | | | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gotenburg, Molndal, Sweden; UCL Institute of Neurology, London, UK
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany; Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | | | - James Hall
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Leigh Johnson
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Yiu-Lian Fong
- Johnson & Johnson, London Innovation Center, London, UK
| | - Johan Luthman
- Neuroscience Clinical Development, Clinical Neuroscience Eisai, Woodcliff Lake, NJ, USA
| | | | | | - Alcibiades Villarreal
- Centro de Neurociencias y Unidad de Investigacion Clinica, Instituto de Investigaciones Cientificas y Servicios de Alta Tecnologia (INDICASAT AIP), Ciudad del Saber, Panama, Panama
| | - Gabrielle Britton
- Centro de Neurociencias y Unidad de Investigacion Clinica, Instituto de Investigaciones Cientificas y Servicios de Alta Tecnologia (INDICASAT AIP), Ciudad del Saber, Panama, Panama
| | - Peter J Snyder
- Department of Neurology, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Kim Henriksen
- Neurodegenerative Diseases, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Paula Grammas
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, RI, USA
| | - Veer Gupta
- Faculty of Health, Engineering and Sciences, Center of Excellence for Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Ralph Martins
- Faculty of Health, Engineering and Sciences, Center of Excellence for Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Harald Hampel
- AXA Research Fund and UPMC Chair, Paris, France; Department de Neurologie, Institut de la Memorie et de la Maladie d'Alzheimer (IM2A) et Institut du Cerveau et du la Moelle epiniere (ICM), Hospital de la Pitie-Salpetriere, Sorbonne Universites, Universite Pierre et Marie Curie, Paris, France
| |
Collapse
|