1
|
Jiao B, Ouyang Z, Liu Y, Zhang C, Xu T, Yang Q, Zhang S, Zhu Y, Wan M, Xiao X, Liu X, Zhou Y, Liao X, Zhang W, Luo S, Tang B, Shen L. Evaluating the diagnostic performance of six plasma biomarkers for Alzheimer's disease and other neurodegenerative dementias in a large Chinese cohort. Alzheimers Res Ther 2025; 17:71. [PMID: 40181481 PMCID: PMC11969745 DOI: 10.1186/s13195-025-01712-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Ethnic variations and detection methods may lead to differences in diagnostic biomarkers of dementia, and few comparative studies have evaluated the six plasma biomarkers of Alzheimer's disease (AD) and other neurodegenerative dementias in the Chinese population. METHODS A cross-sectional cohort of 668 participants were enrolled, including 245 amnesic mild cognitive impairment (aMCI) or AD patients with Aβ positive pathology, 67 with frontotemporal dementia (FTD), 100 with progressive supranuclear palsy (PSP), 72 with dementia with Lewy bodies (DLB) and 184 healthy controls. Additionally, a longitudinal subset of 19 aMCI and 30 AD patients was followed for an average period of 1 year. Plasma biomarkers, including p-tau181, p-tau217, p-tau231, NfL, GFAP, and α-synuclein, were simultaneously measured using a novel single molecular array method. Aβ42 and p-tau181 levels in CSF, amyloid PET and structural MRI were measured. RESULTS Plasma p-tau217 and p-tau231 were most effective in diagnosing aMCI/AD (AUC = 0.95 and 0.93, respectively), while p-tau217, p-tau231 and p-tau181 presented the best differential diagnosis for AD from PSP, FTD and DLB respectively (AUC = 0.84, 0.81 and 0.83). α-synuclein was presented as the best biomarker for PSP variant and behavior variant FTD subtypes (AUC = 0.81 and 0.74, respectively). Among them, p-tau217, p-tau231, GFAP and a-synuclein were negatively correlated with CSF Aβ42/40, while p-tau217 and GFAP were positively correlated with CSF p-tau181. Besides, p-tau181, p-tau217, and GFAP were associated with temporal lobe volume, while p-tau231 and GFAP were associated with frontal lobe volume. Longitudinal analysis showed the higher p-tau181 could predict the cognitive decline progression. CONCLUSIONS This study validate the practicality of blood biomarkers in the Chinese Han population using a novel single molecule immune detection method. Through the clinical performance study for several biomarkers, we found the plasma p-tau217 was the most effective biomarker in AD diagnosis, and p-tau showed high accuracy for differential diagnosis of AD from other dementia, GFAP is associated with multiple aspects of AD pathology, and frontal and temporal lobe volume, and p-tau181 can reflect the dynamic cognitive decline of AD.
Collapse
Affiliation(s)
- Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
| | - Ziyu Ouyang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yiliang Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Cong Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Tianyan Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qijie Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Sizhe Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yuan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Meidan Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xuewen Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
| | - Xixi Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
| | - Yafang Zhou
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xinxin Liao
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Weiwei Zhang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shilin Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China.
- Brain Research Center, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
2
|
Li CY, Fan LY, Lin CH, Hu CJ, Chiu MJ. Ultrasensitive Assays Detect Different Conformations of Plasma β Amyloids. ACS OMEGA 2025; 10:7256-7263. [PMID: 40028141 PMCID: PMC11865983 DOI: 10.1021/acsomega.4c10879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/18/2025] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
With the developments of ultrasensitive technologies such as immunomagnetic reduction (IMR) assay, single molecule array (SIMOA) assay, electrochemiluminescence immunoassay (ECLIA), the assay of blood-based amyloid 1-42 (Aβ1-42) becomes possible. However, the changes in measured plasma Aβ1-42 concentrations in Alzheimer's disease (AD) compared to cognitively unimpaired subjects (CU) are inconsistent. A possible reason for the inconsistency regarding various conformations of Aβ1-42 in plasma is explored in this study. Three samples with equal amounts of Aβ1-42 but different proportions of monomers and oligomers of Aβ1-42 were prepared. The Aβ1-42 composition of monomers and oligomers in samples was analyzed with Western blot. Identically diluted versions of these three samples were assayed with IMR and SIMOA for Aβ1-42 concentrations. The three diluted samples showed similar levels of Aβ1-42 assayed with IMR, whereas much lower levels for samples with more oligomers assayed with SIOMA. The results imply that IMR detects both monomers and oligomers of Aβ1-42. The measured levels of Aβ1-42 are independent of the proportions of monomer or oligomer Aβ1-42 but depend on the total amounts of Aβ1-42. In the case of SIMOA, monomers of Aβ1-42 are the primary target measured. By comparing Aβ1-42 concentrations of the plasma using IMR and SIMOA, the significant difference in plasma Aβ1-42 levels using IMR in AD compared to CU is mainly due to the formations of oligomeric Aβ1-42. Therefore, if the target molecules are monomers of Aβ1-42, SIMOA is the method of choice. Still, if the target molecules should include monomers, small and large oligomers, IMR would be an optimal consideration. In the future, the clinical implications of the proportion of oligomeric Aβ1-42 need to be elucidated.
Collapse
Affiliation(s)
- Chia-Yu Li
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ling-Yun Fan
- Departments
of Neurology, National Taiwan University
Hospital Bei-Hu Branch, Taipei 108, Taiwan
| | - Chin-Hsien Lin
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Institute
of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department
of Biomedical Engineering, National Taiwan
University, Taipei 106, Taiwan
| | - Chaur-Jong Hu
- Taipei
Neuroscience Institute, Taipei Medical University, New Taipei City, 235 Taiwan
- Department
of Neurology and Dementia Center, Taipei
Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
- Department
of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Ming-Jang Chiu
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|
3
|
Tsai CY, Su CL, Huang HT, Lin HW, Lin JW, Hei NC, Cheng WH, Chen YL, Majumdar A, Kang JH, Lee KY, Chen Z, Lin YC, Wu CJ, Kuan YC, Lin YT, Hsu CR, Lee HC, Liu WT. Mediating role of obstructive sleep apnea in altering slow-wave activity and elevating Alzheimer's disease risk: Pilot study from a northern Taiwan cohort. Sleep Health 2025; 11:80-90. [PMID: 39419711 DOI: 10.1016/j.sleh.2024.08.012] [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: 05/24/2024] [Revised: 07/31/2024] [Accepted: 08/31/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVES Obstructive sleep apnea is associated with alterations in slow-wave activity during sleep, potentially increasing the risk of Alzheimer's disease. This study investigated the associations between obstructive sleep apnea manifestations such as respiratory events, hypoxia, arousal, slow-wave patterns, and neurochemical biomarker levels. METHODS Individuals with suspected obstructive sleep apnea underwent polysomnography. Sleep disorder indices, oxygen metrics, and slow-wave activity data were obtained from the polysomnography, and blood samples were taken the following morning to determine the plasma levels of total tau (T-Tau) and amyloid beta-peptide 42 (Aβ42) by using an ultrasensitive immunomagnetic reduction assay. Subsequently, the participants were categorized into groups with low and high Alzheimer's disease risk on the basis of their computed product Aβ42 × T-Tau. Intergroup differences and the associations and mediation effects between sleep-related parameters and neurochemical biomarkers were analyzed. RESULTS Forty-two participants were enrolled, with 21 assigned to each of the low- and high-risk groups. High-risk individuals had a higher apnea-hypopnea index, oxygen desaturation index (≥3%, ODI-3%), fraction of total sleep time with oxygen desaturation (SpO2-90% TST), and arousal index and greater peak-to-peak amplitude and slope in slow-wave activity, with a correspondingly shorter duration, than did low-risk individuals. Furthermore, indices such as the apnea-hypopnea index, ODI-3% and SpO2-90% TST were found to indirectly affect slow-wave activity, thereby raising the Aβ42 × T-Tau level. CONCLUSIONS Obstructive sleep apnea manifestations, such as respiratory events and hypoxia, may influence slow-wave sleep activity (functioning as intermediaries) and may be linked to elevated neurochemical biomarker levels. However, a longitudinal study is necessary to determine causal relationships among these factors. STATEMENT OF SIGNIFICANCE This research aims to bridge gaps in understanding how obstructive sleep apnea is associated with an elevated risk of Alzheimer's disease, providing valuable knowledge for sleep and cognitive health.
Collapse
Affiliation(s)
- Cheng-Yu Tsai
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan; Sleep Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Chien-Ling Su
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Research Center of Biomedical Devices, Taipei Medical University, Taipei, Taiwan
| | - Huei-Tyng Huang
- Department of Medical Physics and Bioengineering, University College London, United Kingdom
| | - Hsin-Wei Lin
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jia-Wei Lin
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ng Cheuk Hei
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wun-Hao Cheng
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Ling Chen
- Institute of Biomedical Informatics of National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Arnab Majumdar
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Jiunn-Horng Kang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Zhihe Chen
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Yi-Chih Lin
- Sleep Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Otolaryngology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Cheng-Jung Wu
- Department of Otolaryngology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Yi-Chun Kuan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Neurology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Yin-Tzu Lin
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chia-Rung Hsu
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Hsin-Chien Lee
- Graduate Institute of Humanities in Medicine, College of Humanities & Social Sciences, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, Taipei Medical University Hospital, Taipei, Taiwan.
| | - Wen-Te Liu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan; Sleep Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.
| |
Collapse
|
4
|
Cheng YW, Lin YJ, Lin YS, Hong WP, Kuan YC, Wu KY, Hsu JL, Wang PN, Pai MC, Chen CS, Fuh JL, Hu CJ, Chiu MJ. Application of blood-based biomarkers of Alzheimer's disease in clinical practice: Recommendations from Taiwan Dementia Society. J Formos Med Assoc 2024; 123:1210-1217. [PMID: 38296698 DOI: 10.1016/j.jfma.2024.01.018] [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: 05/11/2023] [Revised: 11/29/2023] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
Blood-based biomarkers (BBM) are potentially powerful tools that assist in the biological diagnosis of Alzheimer's disease (AD) in vivo with minimal invasiveness, relatively low cost, and good accessibility. This review summarizes current evidence for using BBMs in AD, focusing on amyloid, tau, and biomarkers for neurodegeneration. Blood-based phosphorylated tau and the Aβ42/Aβ40 ratio showed consistent concordance with brain pathology measured by CSF or PET in the research setting. In addition, glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL) are neurodegenerative biomarkers that show the potential to assist in the differential diagnosis of AD. Other pathology-specific biomarkers, such as α-synuclein and TAR DNA-binding protein 43 (TDP-43), can potentially detect AD concurrent pathology. Based on current evidence, the working group from the Taiwan Dementia Society (TDS) achieved consensus recommendations on the appropriate use of BBMs for AD in clinical practice. BBMs may assist clinical diagnosis and prognosis in AD subjects with cognitive symptoms; however, the results should be interpreted by dementia specialists and combining biochemical, neuropsychological, and neuroimaging information. Further studies are needed to evaluate BBMs' real-world performance and potential impact on clinical decision-making.
Collapse
Affiliation(s)
- Yu-Wen Cheng
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Ju Lin
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yung-Shuan Lin
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Pin Hong
- Department of Neurology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Yi-Chun Kuan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan; Department of Neurology and Dementia Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yi Wu
- Department of Psychiatry, Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Jung-Lung Hsu
- Department of Neurology, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital and Chang Gung University, New Taipei City, Taiwan; Graduate Institute of Mind, Brain, & Consciousness, Taipei Medical University, Taipei, Taiwan; Brain & Consciousness Research Center, Shuang Ho Hospital, New Taipei City, Taiwan
| | - Pei-Ning Wang
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Chyi Pai
- Division of Behavioral Neurology, Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Alzheimer's Disease Research Center, National Cheng Kung University Hospital, Tainan, Taiwan; Institute of Gerontology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Sheng Chen
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Psychiatry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jong-Ling Fuh
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chaur-Jong Hu
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan; Department of Neurology and Dementia Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
5
|
Cyr B, Curiel Cid R, Loewenstein D, Vontell RT, Dietrich WD, Keane RW, de Rivero Vaccari JP. The Inflammasome Adaptor Protein ASC in Plasma as a Biomarker of Early Cognitive Changes. Int J Mol Sci 2024; 25:7758. [PMID: 39063000 PMCID: PMC11276719 DOI: 10.3390/ijms25147758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/13/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
Dementia is a group of symptoms including memory loss, language difficulties, and other types of cognitive and functional impairments that affects 57 million people worldwide, with the incidence expected to double by 2040. Therefore, there is an unmet need to develop reliable biomarkers to diagnose early brain impairments so that emerging interventions can be applied before brain degeneration. Here, we performed biomarker analyses for apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), and amyloid-β 42/40 (Aβ42/40) ratio in the plasma of older adults. Participants had blood drawn at baseline and underwent two annual clinical and cognitive evaluations. The groups tested either cognitively normal on both evaluations (NN), cognitively normal year 1 but cognitively impaired year 2 (NI), or cognitively impaired on both evaluations (II). ASC was elevated in the plasma of the NI group compared to the NN and II groups. Additionally, Aβ42 was increased in the plasma in the NI and II groups compared to the NN group. Importantly, the area under the curve (AUC) for ASC in participants older than 70 years old in NN vs. NI groups was 0.81, indicating that ASC is a promising plasma biomarker for early detection of cognitive decline.
Collapse
Affiliation(s)
- Brianna Cyr
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
| | - Rosie Curiel Cid
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
| | - David Loewenstein
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
| | | | - W. Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
| | - Robert W. Keane
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
- Department of Physiology and Biophysics, University of Miami, Miami, FL 33136, USA
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
- Department of Physiology and Biophysics, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
6
|
Hsieh CH, Ko CA, Liang CS, Yeh PK, Tsai CK, Tsai CL, Lin GY, Lin YK, Tsai MC, Yang FC. Longitudinal assessment of plasma biomarkers for early detection of cognitive changes in subjective cognitive decline. Front Aging Neurosci 2024; 16:1389595. [PMID: 38828389 PMCID: PMC11140011 DOI: 10.3389/fnagi.2024.1389595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/29/2024] [Indexed: 06/05/2024] Open
Abstract
Background Individuals experiencing subjective cognitive decline (SCD) are at an increased risk of developing mild cognitive impairment and dementia. Early identification of SCD and neurodegenerative diseases using biomarkers may help clinical decision-making and improve prognosis. However, few cross-sectional and longitudinal studies have explored plasma biomarkers in individuals with SCD using immunomagnetic reduction. Objective To identify plasma biomarkers for SCD. Methods Fifty-two participants [38 with SCD, 14 healthy controls (HCs)] underwent baseline assessments, including measurements of plasma Aβ42, Aβ40, t-tau, p-tau, and α-synuclein using immunomagnetic reduction (IMR) assays, cognitive tests and the Mini-Mental State Examination (MMSE). Following initial cross-sectional analysis, 39 individuals (29 with SCD, 10 HCs) entered a longitudinal phase for reassessment of these biomarkers and the MMSE. Biomarker outcomes across different individual categories were primarily assessed using the area under the receiver operating characteristic (ROC) curve. The SCD subgroup with an MMSE decline over one point was compared to those without such a decline. Results Higher baseline plasma Aβ1-42 levels significantly discriminated participants with SCD from HCs, with an acceptable area under the ROC curve (AUC) of 67.5% [95% confidence interval (CI), 52.7-80.0%]. However, follow-up and changes in MMSE and IMR data did not significantly differ between the SCD and HC groups (p > 0.05). Furthermore, lower baseline plasma Aβ1-42 levels were able to discriminate SCD subgroups with and without cognitive decline with a satisfied performance (AUC, 75.0%; 95% CI, 55.6-89.1%). At last, the changes in t-tau and Aβ42 × t-tau could differentiate between the two SCD subgroups (p < 0.05). Conclusion Baseline plasma Aβ42 may help identify people with SCD and predict SCD progression. The role of plasma Aβ42 levels as well as their upward trends from baseline in cases of SCD that progress to mild cognitive impairment and Alzheimer's disease require further investigation.
Collapse
Affiliation(s)
- Cheng-Hao Hsieh
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-An Ko
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Sung Liang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Kuan Yeh
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Kuang Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Lin Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Guan-Yu Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Neurology, Songshan Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Kai Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Chen Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Fu-Chi Yang
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
7
|
Schraen-Maschke S, Duhamel A, Vidal JS, Ramdane N, Vaudran L, Dussart C, Buée L, Sablonnière B, Delaby C, Allinquant B, Gabelle A, Bombois S, Lehmann S, Hanon O. The free plasma amyloid Aβ 1-42/Aβ 1-40 ratio predicts conversion to dementia for subjects with mild cognitive impairment with performance equivalent to that of the total plasma Aβ 1-42/Aβ 1-40 ratio. The BALTAZAR study. Neurobiol Dis 2024; 193:106459. [PMID: 38423192 DOI: 10.1016/j.nbd.2024.106459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND AND PURPOSE Blood-based biomarkers are a non-invasive solution to predict the risk of conversion of mild cognitive impairment (MCI) to dementia. The utility of free plasma amyloid peptides (not bound to plasma proteins and/or cells) as an early indicator of conversion to dementia is still debated, as the results of studies have been contradictory. In this context, we investigated whether plasma levels of the free amyloid peptides Aβ1-42 and Aβ1-40 and the free plasma Aβ1-42/Aβ1-40 ratio are associated with the conversion of MCI to dementia, in particular AD, over three years of follow-up in a subgroup of the BALTAZAR cohort. We also compared their predictive value to that of total plasma Aβ1-42 and Aβ1-40 levels and the total plasma Aβ1-42/Aβ1-40 ratio. METHODS The plasma Aβ1-42 and Aβ1-40 peptide assay was performed using the INNO-BIA kit (Fujirebio Europe). Free amyloid levels (defined by the amyloid fraction directly accessible to antibodies of the assay) were obtained with the undiluted plasma, whereas total amyloid levels were obtained after the dilution of plasma (1/3) with a denaturing buffer. Free and total Aβ1-42 and Aβ1-40 levels were measured at inclusion for a subgroup of participants (N = 106) with mild cognitive impairment (MCI) from the BALTAZAR study (a large-scale longitudinal multicenter cohort with a three-year follow-up). Associations between conversion and the free/total plasma Aβ1-42 and Aβ1-40 levels and Aβ1-42/Aβ1-40 ratio were analyzed using logistic and Cox Proportional Hazards models. Demographic, clinical, cognitive (MMSE, ADL and IADL), APOE, and MRI characteristics (relative hippocampal volume) were compared using non-parametric (Mann-Whitney) or parametric (Student) tests for quantitative variables and Chi-square or Fisher exact tests for qualitative variables. RESULTS The risk of conversion to dementia was lower for patients in the highest quartile of free plasma Aβ1-42/Aβ1-40 (≥ 25.8%) than those in the three lower quartiles: hazard ratio = 0.36 (95% confidence interval [0.15-0.87]), after adjustment for age, sex, education, and APOE ε4 (p-value = 0.022). This was comparable to the risk of conversion in the highest quartile of total plasma Aβ1-42/Aβ1-40: hazard ratio = 0.37 (95% confidence interval [0.16-0.89], p-value = 0.027). However, while patients in the highest quartile of total plasma Aβ1-42/Aβ1-40 showed higher MMSE scores and a higher hippocampal volume than patients in the three lowest quartiles of total plasma Aβ1-42/Aβ1-40, as well as normal CSF biomarker levels, the patients in the highest quartile of free plasma Aβ1-42/Aβ1-40 did not show any significant differences in MMSE scores, hippocampal volume, or CSF biomarker levels relative to the three lowest quartiles of free plasma Aβ1-42/Aβ1-40. CONCLUSION The free plasma Aβ1-42/Aβ1-40 ratio is associated with a risk of conversion from MCI to dementia within three years, with performance comparable to that of the total plasma Aβ1-42/Aβ1-40 ratio. Threshold levels of the free and total plasma Aβ1-42/Aβ1-40 ratio could be determined, with a 60% lower risk of conversion for patients above the threshold than those below.
Collapse
Affiliation(s)
- S Schraen-Maschke
- Univ. Lille, Inserm, CHU Lille, UMR-S1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, France.
| | - A Duhamel
- Univ. Lille, CHU Lille, ULR 2694-METRICS: Évaluation des Technologies de Santé et des Pratiques Médicales, Lille, France
| | - J S Vidal
- Université de Paris, EA 4468 and APHP, Hôpital Broca, Memory Resource and Research Centre of de Paris-Broca-Ile de France, Paris, France
| | - N Ramdane
- Univ. Lille, CHU Lille, ULR 2694-METRICS: Évaluation des Technologies de Santé et des Pratiques Médicales, Lille, France
| | - L Vaudran
- Univ. Lille, Inserm, CHU Lille, UMR-S1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, France
| | - C Dussart
- Univ. Lille, Inserm, CHU Lille, UMR-S1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, France
| | - L Buée
- Univ. Lille, Inserm, CHU Lille, UMR-S1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, France
| | - B Sablonnière
- Univ. Lille, Inserm, CHU Lille, UMR-S1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, France
| | - C Delaby
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, Montpellier, France
| | - B Allinquant
- UMR-S1266, Université Paris Cité, Institute of Psychiatry and Neurosciences, Inserm, Paris, France
| | - A Gabelle
- CMRR, Université de Montpellier, INM INSERM, CHU de Montpellier, Montpellier, France
| | - S Bombois
- Univ. Lille, Inserm, CHU Lille, UMR-S1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, Lille, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neurologie, Centre des Maladies Cognitives et Comportementales, GH Pitié-Salpêtrière, Paris, France
| | - S Lehmann
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, Montpellier, France
| | - O Hanon
- Université de Paris, EA 4468 and APHP, Hôpital Broca, Memory Resource and Research Centre of de Paris-Broca-Ile de France, Paris, France.
| |
Collapse
|
8
|
Er Zeybekler S. Polydopamine-coated hexagonal boron nitride-based electrochemical immunosensing of T-Tau as a marker of Alzheimer's disease. Bioelectrochemistry 2023; 154:108552. [PMID: 37651881 DOI: 10.1016/j.bioelechem.2023.108552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/02/2023] [Accepted: 08/26/2023] [Indexed: 09/02/2023]
Abstract
Alzheimer's disease (AD) is a complex pathological process that is one of the leading causes of dementia globally. The demand for diagnostic tools that are minimally invasive, timely, and accurate is on the rise. Total tau (T-Tau) protein in blood serum is a promising biomarker for predicting early-stage AD diagnosis. In this study, the hexagonal boron nitride (HBN) based immunosensor platform was developed to detect T-Tau in artificial blood serum. After the exfoliation of HBN, its surface was coated with polydopamine (PDA) in alkaline conditions. The Anti-T-Tau was immobilized on a hydrophilic nanocomposite surface using PDA's reactive catechol and quinone groups, eliminating the need for extra crosslinkers. The working electrode surface of the screen-printed carbon electrode (SPCE) was coated with HBN-PDA nanocomposite using the drop-casting method. The biofunctional surface was created by directly immobilizing Anti-T-Tau on the HBN-PDA nanocomposite-modified SPCE. The analytical performance of the HBN-PDA/Anti-T-Tau/T-Tau immunosensor in the presence of T-Tau isoforms was determined through electrochemical measurements. The linear detection range was 1-30 pg/mL with a detection limit of 0.42 pg/mL for T-Tau, which is suitable for detecting T-Tau in the blood serum.
Collapse
Affiliation(s)
- Simge Er Zeybekler
- Ege University, Faculty of Science Biochemistry Department, 35100 Bornova-Izmir, Turkey
| |
Collapse
|
9
|
Arar S, Haque MA, Kayed R. Protein aggregation and neurodegenerative disease: Structural outlook for the novel therapeutics. Proteins 2023:10.1002/prot.26561. [PMID: 37530227 PMCID: PMC10834863 DOI: 10.1002/prot.26561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 08/03/2023]
Abstract
Before the controversial approval of humanized monoclonal antibody lecanemab, which binds to the soluble amyloid-β protofibrils, all the treatments available earlier, for Alzheimer's disease (AD) were symptomatic. The researchers are still struggling to find a breakthrough in AD therapeutic medicine, which is partially attributable to lack in understanding of the structural information associated with the intrinsically disordered proteins and amyloids. One of the major challenges in this area of research is to understand the structural diversity of intrinsically disordered proteins under in vitro conditions. Therefore, in this review, we have summarized the in vitro applications of biophysical methods, which are aimed to shed some light on the heterogeneity, pathogenicity, structures and mechanisms of the intrinsically disordered protein aggregates associated with proteinopathies including AD. This review will also rationalize some of the strategies in modulating disease-relevant pathogenic protein entities by small molecules using structural biology approaches and biophysical characterization. We have also highlighted tools and techniques to simulate the in vivo conditions for native and cytotoxic tau/amyloids assemblies, urge new chemical approaches to replicate tau/amyloids assemblies similar to those in vivo conditions, in addition to designing novel potential drugs.
Collapse
Affiliation(s)
- Sharif Arar
- Mitchell Center for Neurodegenerative Diseases
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- Department of Chemistry, School of Science, The University of Jordan, Amman 11942, Jordan
| | - Md Anzarul Haque
- Mitchell Center for Neurodegenerative Diseases
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| |
Collapse
|
10
|
Faraz Ahmed T, Bilal Azmi M, Imtiaz F, Zaman U, Ahmed A, Shahbaz N. Plasma levels of phosphorylated tau and neurofilament light chain as potential biomarkers for Alzheimer's disease: A biochemical analysis in Pakistani population. Saudi Pharm J 2023; 31:1202-1209. [PMID: 37273267 PMCID: PMC10236364 DOI: 10.1016/j.jsps.2023.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023] Open
Abstract
The National Institute on Aging-Alzheimer's Association's research framework in 2018 proposed a molecular construct for the diagnosis of Alzheimer's disease (AD). Nonetheless, the clinical exclusionary strategy is still the mainstay of AD diagnosis in Pakistan. We looked at the plasma levels of amyloid beta-42 (Aβ-42), phosphorylated tau (P-tau), and neurofilament light (NFL) in patients with Alzheimer's clinical syndrome (ACS) and healthy controls (HC) from the Pakistani population to keep pace with the global efforts towards establishing accessible and affordable biochemical diagnostic markers for AD in Pakistan. Consultant neurologists screened patients who presented with cognitive impairment to three large tertiary care hospitals in Karachi, and after receiving informed consent, recruited participants with ACS and HC from the same facilities. We collected 5cc of blood in EDTA tubes along with demographic and lifestyle information of the subjects. Plasma aliquots were stored at -80°C after centrifugation. For analysis it was thawed at 4℃ and levels of the three proteins were measured through ELISA. Data from 28 ACS patients and 28 age matched healthy controls were evaluated. Among demographic factors, education and depression were related with health status (p = 0.03 and 0.003, respectively). NFL and P-tau mean values demonstrated a significant difference between the ACS and control groups (p = 0.003 and 0.006), however Aβ42 did not (p = 0.114). ROC analysis showed that plasma P-tau and NFL, with AUCs of 0.717 and 0.735, respectively, could substantially distinguish ACS from the HC group (p = 0.007 and 0.003, respectively). Both plasma P-tau (r = -0.389; p = 0.004) and NFL (r = -0.424; p = 0.001) levels were significantly and negatively correlated with individuals' MMSE scores. NFL and plasma P-tau show promise in differentiating AD patients from healthy individuals. However, similar larger studies are needed to validate our findings.
Collapse
Affiliation(s)
- Tehniat Faraz Ahmed
- Department of Biochemistry, Dow International Dental College, Dow University of Health Sciences (DUHS), 74200 Karachi, Pakistan
| | - Muhammad Bilal Azmi
- Department of Biochemistry, Dow Medical College, Dow University of Health Sciences (DUHS), 74200 Karachi, Pakistan
| | - Fauzia Imtiaz
- Department of Biochemistry, Dow Medical College, Dow University of Health Sciences (DUHS), 74200 Karachi, Pakistan
| | - Uzma Zaman
- Department of Biochemistry, Dow International Medical College, Dow University of Health Sciences (DUHS), 74200 Karachi, Pakistan
| | - Affan Ahmed
- Dow Medical College, Dow University of Health Sciences (DUHS), 74200 Karachi, Pakistan
| | - Naila Shahbaz
- Department of Neurology, Dr Ruth Pfau Civil Hospital Karachi, 74400 Karachi, Pakistan
| |
Collapse
|
11
|
Farvadi F, Hashemi F, Amini A, Alsadat Vakilinezhad M, Raee MJ. Early Diagnosis of Alzheimer's Disease with Blood Test; Tempting but Challenging. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2023; 12:172-210. [PMID: 38313372 PMCID: PMC10837916 DOI: 10.22088/ijmcm.bums.12.2.172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 11/25/2023] [Accepted: 12/13/2023] [Indexed: 02/06/2024]
Abstract
The increasing prevalence of Alzheimer's disease (AD) has led to a health crisis. According to official statistics, more than 55 million people globally have AD or other types of dementia, making it the sixth leading cause of death. It is still difficult to diagnose AD and there is no definitive diagnosis yet; post-mortem autopsy is still the only definite method. Moreover, clinical manifestations occur very late in the course of disease progression; therefore, profound irreversible changes have already occurred when the disease manifests. Studies have shown that in the preclinical stage of AD, changes in some biomarkers are measurable prior to any neurological damage or other symptoms. Hence, creating a reliable, fast, and affordable method capable of detecting AD in early stage has attracted the most attention. Seeking clinically applicable, inexpensive, less invasive, and much more easily accessible biomarkers for early diagnosis of AD, blood-based biomarkers (BBBs) seem to be an ideal option. This review is an inclusive report of BBBs that have been shown to be altered in the course of AD progression. The aim of this report is to provide comprehensive insight into the research status of early detection of AD based on BBBs.
Collapse
Affiliation(s)
- Fakhrossadat Farvadi
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Hashemi
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, the University of Newcastle, Newcastle, Australia
| | - Azadeh Amini
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical sciences, Tehran, Iran
| | | | - Mohammad Javad Raee
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Chiu SI, Fan LY, Lin CH, Chen TF, Lim WS, Jang JSR, Chiu MJ. Machine Learning-Based Classification of Subjective Cognitive Decline, Mild Cognitive Impairment, and Alzheimer's Dementia Using Neuroimage and Plasma Biomarkers. ACS Chem Neurosci 2022; 13:3263-3270. [PMID: 36378559 DOI: 10.1021/acschemneuro.2c00255] [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: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) progresses relentlessly from the preclinical to the dementia stage. The process begins decades before the diagnosis of dementia. Therefore, it is crucial to detect early manifestations to prevent cognitive decline. Recent advances in artificial intelligence help tackle the complex high-dimensional data encountered in clinical decision-making. In total, we recruited 206 subjects, including 69 cognitively unimpaired, 40 subjective cognitive decline (SCD), 34 mild cognitive impairment (MCI), and 63 AD dementia (ADD). We included 3 demographic, 1 clinical, 18 brain-image, and 3 plasma biomarker (Aß1-42, Aß1-40, and tau protein) features. We employed the linear discriminant analysis method for feature extraction to make data more distinguishable after dimension reduction. The sequential forward selection method was used for feature selection to identify the 12 best features for machine learning classifiers. We used both random forest and support vector machine as classifiers. The area under the receiver operative curve (AUROC) was close to 0.9 between diseased (combining ADD and MCI) and the controls. AUROC was higher than 0.85 between SCD and controls, 0.90 between MCI and SCD, and above 0.85 between ADD and MCI. We can differentiate between adjacent phases of the AD spectrum with blood biomarkers and brain MR images with the help of machine learning algorithms.
Collapse
Affiliation(s)
- Shu-I Chiu
- Department of Computer Science, National Chengchi University, Taipei 116302, Taiwan
| | - Ling-Yun Fan
- Queensland Brain Institute, University of Queensland, St Lucia, QLD 4067, Australia.,Departments of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei 108206, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei 100225, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei 100225, Taiwan
| | - Wee Shin Lim
- Department of Computer Science and Information Engineering, National Taiwan University, Taipei 106319, Taiwan
| | - Jyh-Shing Roger Jang
- Department of Computer Science and Information Engineering, National Taiwan University, Taipei 106319, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei 100225, Taiwan.,Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 106319, Taiwan.,Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei 100233, Taiwan.,Graduate Institute of Psychology, National Taiwan University, Taipei 106319, Taiwan
| |
Collapse
|
13
|
Hanon O, Vidal JS, Lehmann S, Bombois S, Allinquant B, Baret-Rose C, Tréluyer JM, Abdoul H, Gelé P, Delmaire C, Blanc F, Mangin JF, Buée L, Touchon J, Hugon J, Vellas B, Galbrun E, Benetos A, Berrut G, Paillaud E, Wallon D, Castelnovo G, Volpe-Gillot L, Paccalin M, Robert P, Godefroy O, Camus V, Belmin J, Vandel P, Novella JL, Duron E, Rigaud AS, Schraen-Maschke S, Gabelle A. Plasma amyloid beta predicts conversion to dementia in subjects with mild cognitive impairment: The BALTAZAR study. Alzheimers Dement 2022; 18:2537-2550. [PMID: 35187794 DOI: 10.1002/alz.12613] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/20/2021] [Accepted: 12/10/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Blood-based biomarkers are the next challenge for Alzheimer's disease (AD) diagnosis and prognosis. METHODS Mild cognitive impairment (MCI) participants (N = 485) of the BALTAZAR study, a large-scale longitudinal multicenter cohort, were followed-up for 3 years. A total of 165 of them converted to dementia (95% AD). Associations of conversion and plasma amyloid beta (Aβ)1-42 , Aβ1-40 , Aβ1-42 /Aβ1-40 ratio were analyzed with logistic and Cox models. RESULTS Converters to dementia had lower level of plasma Aβ1-42 (37.1 pg/mL [12.5] vs. 39.2 [11.1] , P value = .03) and lower Aβ1-42 /Aβ1-40 ratio than non-converters (0.148 [0.125] vs. 0.154 [0.076], P value = .02). MCI participants in the highest quartile of Aβ1-42 /Aβ1-40 ratio (>0.169) had a significant lower risk of conversion (hazard ratio adjusted for age, sex, education, apolipoprotein E ε4, hippocampus atrophy = 0.52 (95% confidence interval [0.31-0.86], P value = .01). DISCUSSION In this large cohort of MCI subjects we identified a threshold for plasma Aβ1-42 /Aβ1-40 ratio that may detect patients with a low risk of conversion to dementia within 3 years.
Collapse
Affiliation(s)
- Olivier Hanon
- Memory Resource and Research Centre of de Paris-Broca-Ile de France, Université de Paris, EA 4468, APHP, Hopital Broca, Paris, France
| | - Jean-Sébastien Vidal
- Memory Resource and Research Centre of de Paris-Broca-Ile de France, Université de Paris, EA 4468, APHP, Hopital Broca, Paris, France
| | - Sylvain Lehmann
- CHU Montpellier, LBPC, Inserm, Université de Montpellier, Montpellier, France
| | - Stéphanie Bombois
- CHU Lille, U1172-LilNCog, LiCEND, LabEx DISTALZ, Université de Lille, Inserm, Lille, France
| | - Bernadette Allinquant
- UMR-S 1266, Université de Paris, Institute of Psychiatric and Neurosciences, Inserm, Paris, France
| | - Christiane Baret-Rose
- UMR-S 1266, Université de Paris, Institute of Psychiatric and Neurosciences, Inserm, Paris, France
| | - Jean-Marc Tréluyer
- Clinical Research Unit, Université de Paris, APHP, Hôpital Necker, Paris, France
| | - Hendy Abdoul
- Clinical Research Unit, Université de Paris, APHP, Hôpital Necker, Paris, France
| | - Patrick Gelé
- CHU Lille, CRB/CIC1403, Université de Lille, Inserm, Lille, France
| | - Christine Delmaire
- CHU Lille, U1172-LilNCog, LiCEND, LabEx DISTALZ, Université de Lille, Inserm, Lille, France
| | - Fredéric Blanc
- CM2R, pôle de Gériatrie, Laboratoire ICube, FMTS, CNRS, équipe IMIS, Université de Strasbourg, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jean-François Mangin
- Neurospin, CEA, CNRS, cati-neuroimaging.com, CATI Multicenter Neuroimaging Platform, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Luc Buée
- CHU Lille, U1172-LilNCog, LiCEND, LabEx DISTALZ, Université de Lille, Inserm, Lille, France
| | - Jacques Touchon
- Department of Neurology, Memory Research and Resources Center of Montpellier, Inserm INM NeuroPEPs Team, Excellence Center of Neurodegenerative Disorders, Université de Montpellier, CHU Montpellier, Montpellier, France
| | - Jacques Hugon
- APHP, Groupe Hospitalier Saint Louis-Lariboisière Fernand Widal, Center of Cognitive Neurology, Université de Paris, Paris, France
| | - Bruno Vellas
- Memory Resource and Research Centre of Midi-Pyrénées, Université de Toulouse III, CHU La Grave-Casselardit, Toulouse, France
| | - Evelyne Galbrun
- Department of Gérontology 2, Sorbonne Université, APHP, Centre Hospitalier Dupuytren, Draveil, France
| | - Athanase Benetos
- Memory Resource and Research Centre of Lorraine, Université de Lorraine, CHRU de Nancy, Vandoeuvre-lès-Nancy, France
| | - Gilles Berrut
- Department of Clinical Gerontology, Memory Research Resource Center of Nantes, Université de Nantes, EA 4334 Movement-Interactions-Performance, CHU Nantes, Nantes, France
| | - Elena Paillaud
- Service de Gériatrie, Université de Paris, APHP, Hôpital Europeen Georges Pompidou, Paris, France
| | - David Wallon
- CHU de Rouen, Department of Neurology and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, CIC-CRB1404, Normandie Univ, UNIROUEN, Inserm U1245, Rouen, France
| | | | - Lisette Volpe-Gillot
- Service de Neuro-Psycho-Gériatrie, Memory Clinic, Hôpital Léopold Bellan, Paris, France
| | - Marc Paccalin
- Memory Resource and Research Centre of Poitiers, CHU de Poitiers, Poitiers, France
| | - Philippe Robert
- Memory Research Resource Center of Nice, CoBTek lab, Université Côte d'Azur, CHU de Nice, Nice, France
| | - Olivier Godefroy
- Memory Resource and Research Centre of Amiens Picardie, CHU d'Amiens-Picardie, Amiens, France
| | - Vincent Camus
- CHRU de Tours, UMR Inserm U1253, Université François-Rabelais de Tours, Tours, France
| | - Joël Belmin
- Service de Gériatrie Ambulatoire, Sorbonne Université, APHP, Hôpitaux Universitaires Pitie-Salpêtrière-Charles Foix, Paris, France
| | - Pierre Vandel
- Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, CHU de Besançon, Memory Resource and Research Centre of Besançon Franche-Comté, Université Bourgogne Franche-Comté, Besançon, France
| | - Jean-Luc Novella
- Memory Resource and Research Centre of Champagne-Ardenne, Université de Reims Champagne-Ardenne, EA 3797, CHU de Reims, Reims, France
| | - Emmanuelle Duron
- Département de gériatrie, Équipe MOODS, Inserm 1178, Université Paris-Saclay, APHP, Hôpital Paul Brousse, Villejuif, France
| | - Anne-Sophie Rigaud
- Memory Resource and Research Centre of de Paris-Broca-Ile de France, Université de Paris, EA 4468, APHP, Hopital Broca, Paris, France
| | | | - Audrey Gabelle
- Department of Neurology, Memory Research and Resources Center of Montpellier, Inserm INM NeuroPEPs Team, Excellence Center of Neurodegenerative Disorders, Université de Montpellier, CHU Montpellier, Montpellier, France
| | | |
Collapse
|
14
|
Tsai CY, Wu SM, Kuan YC, Lin YT, Hsu CR, Hsu WH, Liu YS, Majumdar A, Stettler M, Yang CM, Lee KY, Wu D, Lee HC, Wu CJ, Kang JH, Liu WT. Associations between risk of Alzheimer's disease and obstructive sleep apnea, intermittent hypoxia, and arousal responses: A pilot study. Front Neurol 2022; 13:1038735. [PMID: 36530623 PMCID: PMC9747943 DOI: 10.3389/fneur.2022.1038735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/04/2022] [Indexed: 01/30/2024] Open
Abstract
OBJECTIVES Obstructive sleep apnea (OSA) may increase the risk of Alzheimer's disease (AD). However, potential associations among sleep-disordered breathing, hypoxia, and OSA-induced arousal responses should be investigated. This study determined differences in sleep parameters and investigated the relationship between such parameters and the risk of AD. METHODS Patients with suspected OSA were recruited and underwent in-lab polysomnography (PSG). Subsequently, blood samples were collected from participants. Patients' plasma levels of total tau (T-Tau) and amyloid beta-peptide 42 (Aβ42) were measured using an ultrasensitive immunomagnetic reduction assay. Next, the participants were categorized into low- and high-risk groups on the basis of the computed product (Aβ42 × T-Tau, the cutoff for AD risk). PSG parameters were analyzed and compared. RESULTS We included 36 patients in this study, of whom 18 and 18 were assigned to the low- and high-risk groups, respectively. The average apnea-hypopnea index (AHI), apnea, hypopnea index [during rapid eye movement (REM) and non-REM (NREM) sleep], and oxygen desaturation index (≥3%, ODI-3%) values of the high-risk group were significantly higher than those of the low-risk group. Similarly, the mean arousal index and respiratory arousal index (R-ArI) of the high-risk group were significantly higher than those of the low-risk group. Sleep-disordered breathing indices, oxygen desaturation, and arousal responses were significantly associated with an increased risk of AD. Positive associations were observed among the AHI, ODI-3%, R-ArI, and computed product. CONCLUSIONS Recurrent sleep-disordered breathing, intermittent hypoxia, and arousal responses, including those occurring during the NREM stage, were associated with AD risk. However, a longitudinal study should be conducted to investigate the causal relationships among these factors.
Collapse
Affiliation(s)
- Cheng-Yu Tsai
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yi-Chun Kuan
- Sleep Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
- Dementia Center, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yin-Tzu Lin
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chia-Rung Hsu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Wen-Hua Hsu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Shin Liu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Arnab Majumdar
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Marc Stettler
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Chien-Ming Yang
- Department of Psychology, National Chengchi University, Taipei, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Dean Wu
- Sleep Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
- Dementia Center, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hsin-Chien Lee
- Department of Psychiatry, Taipei Medical University Hospital, Taipei, Taiwan
| | - Cheng-Jung Wu
- Department of Otolaryngology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Jiunn-Horng Kang
- Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, Taipei, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Wen-Te Liu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Sleep Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
15
|
Abuelezz NZ, Nasr FE, Abdel Aal WM, Molokhia T, Zaky A. Sera miR-34a, miR-29b and miR-181c as potential novel diagnostic biomarker panel for Alzheimers in the Egyptian population. Exp Gerontol 2022; 169:111961. [PMID: 36155067 DOI: 10.1016/j.exger.2022.111961] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/26/2022] [Accepted: 09/20/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Till date, there is an obvious obscurity of specific and early diagnostic biomarkers for Alzheimer's disease (AD). The promising diagnostic potential of serum miRNAs is increasingly emerging; however, rare miRNAs data originates from middle and low-income countries to provide proper validation in these highly affected populations. This study evaluated the diagnostic value of serum miR-34a, miR-29b and miR-181c in Egyptian AD patients. METHODS Expression levels of serum miR-34a, miR-29b and miR-181c were determined using quantitative real time PCR in AD patients versus healthy controls. Amyloid Beta 42 (Aβ42), Phosphorylated Tau (p-Tau) and TNF-α levels were also detected as distinctive AD markers. We further explored the correlation between miRNAs levels and Mini mental state examination (MMSE) scores. Finally, we conducted logistic regression and ROC curve analyses to evaluate the diagnostic values of the measured parameters. RESULTS Sera miR-34a, miR-29b and miR-181c were significantly downregulated in AD patients and this decrease was associated with cognitive decline. AD patients manifested significant elevation of Aβ42, pTau and TNF-α levels. The measured miRNAs showed good AD diagnostic value solely and when used together (AUC = 0.77, 95 % C·I. 0.62-0.93 at p < 0.01). Interestingly, combining miRNAs panel with Aβ42, TNF-α and pTau levels remarkably increased the diagnostic power (AUC = 0.97, 95 % C·I. 0.94-1.00 at p < 0.001) achieving sensitivity 88.2 % and specificity 91.4 %. CONCLUSION This study spots for the first time the diagnostic potential of serum miR-34a, miR-29b and miR-181c as minimally invasive AD biomarker panel in Egyptian patients and highlights their contribution in AD pathogenesis.
Collapse
Affiliation(s)
- Nermeen Z Abuelezz
- Biochemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, Egypt.
| | - Fayza Eid Nasr
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | | | - Tarek Molokhia
- Department of Psychiatry, School of Medicine, Alexandria University, Alexandria, Egypt
| | - Amira Zaky
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
16
|
Chiu PY, Yang FC, Chiu MJ, Lin WC, Lu CH, Yang SY. Relevance of plasma biomarkers to pathologies in Alzheimer's disease, Parkinson's disease and frontotemporal dementia. Sci Rep 2022; 12:17919. [PMID: 36289355 PMCID: PMC9605966 DOI: 10.1038/s41598-022-22647-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/18/2022] [Indexed: 01/20/2023] Open
Abstract
Amyloid plaques and tau tangles are pathological hallmarks of Alzheimer's disease (AD). Parkinson's disease (PD) results from the accumulation of α-synuclein. TAR DNA-binding protein (TDP-43) and total tau protein (T-Tau) play roles in FTD pathology. All of the pathological evidence was found in the biopsy. However, it is impossible to perform stein examinations in clinical practice. Assays of biomarkers in plasma would be convenient. It would be better to investigate the combinations of various biomarkers in AD, PD and FTD. Ninety-one subjects without neurodegenerative diseases, 76 patients with amnesic mild cognitive impairment (aMCI) or AD dementia, combined as AD family, were enrolled. One hundred and nine PD patients with normal cognition (PD-NC) or dementia (PDD), combined as PD family, were enrolled. Twenty-five FTD patients were enrolled for assays of plasma amyloid β 1-40 (Aβ1-40), Aβ1-42, T-Tau, α-synuclein and TDP-43 using immunomagnetic reduction (IMR). The results show that Aβs and T-Tau are major domains in AD family. α-synuclein is highly dominant in PD family. FTD is closely associated with TDP-43 and T-Tau. The dominant plasma biomarkers in AD family, PD family and FTD are consistent with pathology. This implies that plasma biomarkers are promising for precise and differential assessments of AD, PD and FTD in clinical practice.
Collapse
Affiliation(s)
- Pai-Yi Chiu
- grid.452796.b0000 0004 0634 3637Department of Neurology, Show Chwan Memorial Hospital, Chunghwa, 500 Taiwan ,MR-Guided Focus Ultrasound Center, Chang Bin Shaw Chwan Memorial Hospital, Changhwa, 505 Taiwan
| | - Fu-Chi Yang
- grid.278244.f0000 0004 0638 9360Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114 Taiwan
| | - Ming-Jang Chiu
- grid.19188.390000 0004 0546 0241Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100 Taiwan ,grid.19188.390000 0004 0546 0241Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 100 Taiwan ,grid.19188.390000 0004 0546 0241Department of Psychology, National Taiwan University, Taipei, 106 Taiwan ,grid.19188.390000 0004 0546 0241Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, 106 Taiwan
| | - Wei-Che Lin
- grid.145695.a0000 0004 1798 0922Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung, 833 Taiwan
| | - Cheng-Hsien Lu
- grid.145695.a0000 0004 1798 0922Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung, 833 Taiwan
| | | |
Collapse
|
17
|
Chen PH, Lin SI, Liao YY, Hsu WL, Cheng FY. Associations between blood-based biomarkers of Alzheimer's disease with cognition in motoric cognitive risk syndrome: A pilot study using plasma Aβ42 and total tau. Front Aging Neurosci 2022; 14:981632. [PMID: 36268195 PMCID: PMC9577229 DOI: 10.3389/fnagi.2022.981632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/20/2022] [Indexed: 01/28/2023] Open
Abstract
Background Motoric cognitive risk (MCR) syndrome is a conceptual construct that combines slow gait speed with subjective cognitive complaints and has been shown to be associated with an increased risk of developing dementia. However, the relationships between the pathology of Alzheimer's disease (AD) and MCR syndrome remain uncertain. Therefore, the purpose of this study was to determine the levels of plasma AD biomarkers (Aβ42 and total tau) and their relationships with cognition in individuals with MCR. Materials and methods This was a cross-sectional pilot study that enrolled 25 individuals with normal cognition (NC), 27 with MCR, and 16 with AD. Plasma Aβ42 and total tau (t-tau) levels were measured using immunomagnetic reduction (IMR) assays. A comprehensive neuropsychological assessment was also performed. Results The levels of plasma t-tau proteins did not differ significantly between the MCR and AD groups, but that of plasma t-tau was significantly increased in the MCR and AD groups, compared to the NC group. Visuospatial performance was significantly lower in the MCR group than in the NC group. The levels of plasma t-tau correlated significantly with the Montreal Cognitive Assessment (MoCA) and Boston naming test scores in the MCR group. Conclusion In this pilot study, we found significantly increased plasma t-tau proteins in the MCR and AD groups, compared with the NC group. The plasma t-tau levels were also significantly correlated with the cognitive function of older adults with MCR. These results implied that MCR and AD may share similar pathology. However, these findings need further confirmation in longitudinal studies.
Collapse
Affiliation(s)
- Pei-Hao Chen
- Department of Neurology, MacKay Memorial Hospital, Taipei, Taiwan,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan,Graduate Institute of Mechanical and Electrical Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Sang-I Lin
- Institute of Long-Term Care, MacKay Medical College, New Taipei City, Taiwan
| | - Ying-Yi Liao
- Department of Gerontological Health Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Wei-Ling Hsu
- Institute of Long-Term Care, MacKay Medical College, New Taipei City, Taiwan,Center of Dementia Care, MacKay Memorial Hospital, New Taipei City, Taiwan
| | - Fang-Yu Cheng
- Institute of Long-Term Care, MacKay Medical College, New Taipei City, Taiwan,*Correspondence: Fang-Yu Cheng,
| |
Collapse
|
18
|
Xu C, Zhao L, Dong C. A Review of Application of Aβ42/40 Ratio in Diagnosis and Prognosis of Alzheimer’s Disease. J Alzheimers Dis 2022; 90:495-512. [DOI: 10.3233/jad-220673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The number of patients with Alzheimer’s disease (AD) and non-Alzheimer’s disease (non-AD) has drastically increased over recent decades. The amyloid cascade hypothesis attributes a vital role to amyloid-β protein (Aβ) in the pathogenesis of AD. As the main pathological hallmark of AD, amyloid plaques consist of merely the 42 and 40 amino acid variants of Aβ (Aβ 42 and Aβ 40). The cerebrospinal fluid (CSF) biomarker Aβ 42/40 has been extensively investigated and eventually integrated into important diagnostic tools to support the clinical diagnosis of AD. With the development of highly sensitive assays and technologies, blood-based Aβ 42/40, which was obtained using a minimally invasive and cost-effective method, has been proven to be abnormal in synchrony with CSF biomarker values. This paper presents the recent progress of the CSF Aβ 42/40 ratio and plasma Aβ 42/40 for AD as well as their potential clinical application as diagnostic markers or screening tools for dementia.
Collapse
Affiliation(s)
- Chang Xu
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Li Zhao
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunbo Dong
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
19
|
Abstract
OBJECTIVE Fibromyalgia (FM) is a chronic widespread pain syndrome. Although its mechanism remains relatively unknown, accelerated neurodegeneration in the brain has been reported in patients with FM. Sleep disturbance can increase the risk of neurocognitive disorders, which are associated with tau and beta-amyloid (Aβ) protein accumulation. We hypothesize neurodegeneration in patients with FM may be associated with sleep disturbance. METHODS In this case-control study, we analyzed serum tau and Aβ levels and their association with symptom profiles for patients with FM, by recruiting 22 patients with FM and 22 age-matched healthy participants. The visual analog scale, Fibromyalgia Impact Questionnaire, pressure pain threshold test, Pittsburgh Sleep Quality Index (PSQI), Beck Depression Inventory-II, Beck Anxiety Inventory, and serum tau and beta-amyloid-42 (Aβ-42) levels were recorded. The Mann-Whitney test was conducted to compare questionnaire and protein level results between the groups. Pearson correlation test was conducted to investigate the correlation of questionnaire scores with tau and Aβ-42 levels in patients with FM. The significance level was set at P < .05. RESULTS Serum tau and Aβ-42 levels were significantly higher in patients with FM than in controls. A positive correlation between serum tau levels and PSQI scores was observed in patients with FM (r = 0.476, P = .025). We found that only sleep disturbance in patients with FM was significantly associated with higher serum tau levels among all symptom scores. CONCLUSIONS We suggest sleep disturbance may play a vital role in the pathomechanism of accelerated neurodegeneration in FM.
Collapse
|
20
|
Li K, Qu H, Ma M, Xia C, Cai M, Han F, Zhang Q, Gu X, Ma Q. Correlation Between Brain Structure Atrophy and Plasma Amyloid-β and Phosphorylated Tau in Patients With Alzheimer’s Disease and Amnestic Mild Cognitive Impairment Explored by Surface-Based Morphometry. Front Aging Neurosci 2022; 14:816043. [PMID: 35547625 PMCID: PMC9083065 DOI: 10.3389/fnagi.2022.816043] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/28/2022] [Indexed: 12/27/2022] Open
Abstract
ObjectiveTo investigate the changes in the cortical thickness of the region of interest (ROI) and plasma Aβ40, Aβ42, and phosphorylated Tau (P-Tau) concentrations in patients with Alzheimer’s disease (AD) and amnestic mild cognitive impairment (aMCI) as the disease progressed with surface-based morphometry (SBM), to analyze the correlation between ROI cortical thickness and measured plasma indexes and neuropsychological scales, and to explore the clinical value of ROI cortical thickness combined with plasma Aβ40, Aβ42, and P-Tau in the early recognition and diagnosis of AD.MethodsThis study enrolled 33 patients with AD, 48 patients with aMCI, and 33 healthy controls (normal control, NC). Concentration changes in plasma Aβ42, Aβ40, and P-Tau collected in each group were analyzed. Meanwhile, the whole brain T1 structure images (T1WI-3D-MPRAGE) of each group of patients were collected, and T1 image in AD-aMCI, AD-NC, and aMCI-NC group were analyzed and processed by SBM technology to obtain brain regions with statistical differences as clusters, and the cortical thickness of each cluster was extracted. Multivariate ordered logistic regression analysis was used to screen out the measured plasma indexes and the indexes with independent risk factors in the cortical thickness of each cluster. Three comparative receiver operating characteristic (ROC) curves of AD-aMCI, AD-NC, and aMCI-NC groups were plotted, respectively, to explore the diagnostic value of multi-factor combined prediction for cognitive impairment. The relationship between cortical thickness and plasma indexes, and between cortical thickness and Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) scores were clarified by Pearson correlation analysis.ResultsPlasma Aβ40, Aβ42, and P-Tau proteins in the NC, aMCI, and AD groups increased with the progression of AD (P < 0.01); cortical thickness reductions in the AD-aMCI groups and AD-NC groups mainly involved the bilateral superior temporal gyrus, transverse temporal gyrus, superior marginal gyrus, insula, right entorhinal cortex, right fusiform gyrus, and cingulate gyrus. However, there were no statistical significances in cortical thickness reductions in the aMCI and NC groups. The cortical thickness of the ROI was negatively correlated with plasma Aβ40, Aβ42, and P-Tau concentrations (P < 0.05), and the cortical thickness of the ROI was positively correlated with MMSE and MoCA scores. Independent risk factors such as Aβ40, Aβ42, P-Tau, and AD-NC cluster 1R (right superior temporal gyrus, temporal pole, entorhinal cortex, transverse temporal gyrus, fusiform gyrus, superior marginal gyrus, middle temporal gyrus, and inferior temporal gyrus) were combined to plot ROC curves. The diagnostic efficiency of plasma indexes was higher than that of cortical thickness indexes, the diagnostic efficiency of ROC curves after the combination of cortical thickness and plasma indexes was higher than that of cortical thickness or plasma indexes alone.ConclusionPlasma Aβ40, Aβ42, and P-Tau may be potential biomarkers for early prediction of AD. As the disease progressed, AD patients developed cortical atrophy characterized by atrophy of the medial temporal lobe. The combined prediction of these region and plasma Aβ40, Aβ42, and P-Tau had a higher diagnostic value than single-factor prediction for cognitive decline.
Collapse
Affiliation(s)
- Kaidi Li
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hang Qu
- Department of Imaging, Yangzhou First People’s Hospital, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Mingyi Ma
- Department of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Chenyu Xia
- Department of Neurology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ming Cai
- Department of Neurology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Fang Han
- Department of Imaging, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Qing Zhang
- Department of Imaging, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Xinyi Gu
- Department of Neurology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Qiang Ma
- Department of Neurology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- *Correspondence: Qiang Ma,
| |
Collapse
|
21
|
Varesi A, Carrara A, Pires VG, Floris V, Pierella E, Savioli G, Prasad S, Esposito C, Ricevuti G, Chirumbolo S, Pascale A. Blood-Based Biomarkers for Alzheimer's Disease Diagnosis and Progression: An Overview. Cells 2022; 11:1367. [PMID: 35456047 PMCID: PMC9044750 DOI: 10.3390/cells11081367] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 01/10/2023] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disease characterized by amyloid-β (Aβ) plaque deposition and neurofibrillary tangle accumulation in the brain. Although several studies have been conducted to unravel the complex and interconnected pathophysiology of AD, clinical trial failure rates have been high, and no disease-modifying therapies are presently available. Fluid biomarker discovery for AD is a rapidly expanding field of research aimed at anticipating disease diagnosis and following disease progression over time. Currently, Aβ1-42, phosphorylated tau, and total tau levels in the cerebrospinal fluid are the best-studied fluid biomarkers for AD, but the need for novel, cheap, less-invasive, easily detectable, and more-accessible markers has recently led to the search for new blood-based molecules. However, despite considerable research activity, a comprehensive and up-to-date overview of the main blood-based biomarker candidates is still lacking. In this narrative review, we discuss the role of proteins, lipids, metabolites, oxidative-stress-related molecules, and cytokines as possible disease biomarkers. Furthermore, we highlight the potential of the emerging miRNAs and long non-coding RNAs (lncRNAs) as diagnostic tools, and we briefly present the role of vitamins and gut-microbiome-related molecules as novel candidates for AD detection and monitoring, thus offering new insights into the diagnosis and progression of this devastating disease.
Collapse
Affiliation(s)
- Angelica Varesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- Almo Collegio Borromeo, 27100 Pavia, Italy
| | - Adelaide Carrara
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (A.C.); (V.F.)
| | - Vitor Gomes Pires
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA;
| | - Valentina Floris
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (A.C.); (V.F.)
| | - Elisa Pierella
- School of Medicine, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Gabriele Savioli
- Emergency Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Sakshi Prasad
- Faculty of Medicine, National Pirogov Memorial Medical University, 21018 Vinnytsya, Ukraine;
| | - Ciro Esposito
- Unit of Nephrology and Dialysis, ICS Maugeri, University of Pavia, 27100 Pavia, Italy;
| | - Giovanni Ricevuti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37129 Verona, Italy;
| | - Alessia Pascale
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
22
|
Association between cyclic variation in the heart rate index and biomarkers of neurodegenerative diseases in obstructive sleep apnea syndrome: A pilot study. J Clin Neurosci 2022; 98:37-44. [DOI: 10.1016/j.jocn.2022.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 11/14/2021] [Accepted: 01/20/2022] [Indexed: 10/19/2022]
|
23
|
Álvarez-Sánchez L, Peña-Bautista C, Baquero M, Cháfer-Pericás C. Novel Ultrasensitive Detection Technologies for the Identification of Early and Minimally Invasive Alzheimer's Disease Blood Biomarkers. J Alzheimers Dis 2022; 86:1337-1369. [PMID: 35213367 DOI: 10.3233/jad-215093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Single molecule array (SIMOA) and other ultrasensitive detection technologies have allowed the determination of blood-based biomarkers of Alzheimer's disease (AD) for diagnosis and monitoring, thereby opening up a promising field of research. OBJECTIVE To review the published bibliography on plasma biomarkers in AD using new ultrasensitive techniques. METHODS A systematic review of the PubMed database was carried out to identify reports on the use of blood-based ultrasensitive technology to identify biomarkers for AD. RESULTS Based on this search, 86 works were included and classified according to the biomarker determined. First, plasma amyloid-β showed satisfactory accuracy as an AD biomarker in patients with a high risk of developing dementia. Second, plasma t-Tau displayed good sensitivity in detecting different neurodegenerative diseases. Third, plasma p-Tau was highly specific for AD. Fourth, plasma NfL was highly sensitive for distinguishing between patients with neurodegenerative diseases and healthy controls. In general, the simultaneous determination of several biomarkers facilitated greater accuracy in diagnosing AD (Aβ42/Aβ40, p-Tau181/217). CONCLUSION The recent development of ultrasensitive technology allows the determination of blood-based biomarkers with high sensitivity, thus facilitating the early detection of AD through the analysis of easily obtained biological samples. In short, as a result of this knowledge, pre-symptomatic and early AD diagnosis may be possible, and the recruitment process for future clinical trials could be more precise. However, further studies are necessary to standardize levels of blood-based biomarkers in the general population and thus achieve reproducible results among different laboratories.
Collapse
Affiliation(s)
| | - Carmen Peña-Bautista
- Alzheimer Disease Research Group, Health Research Institute La Fe, Valencia, Spain
| | - Miguel Baquero
- Division of Neurology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | | |
Collapse
|
24
|
Mahaman YAR, Embaye KS, Huang F, Li L, Zhu F, Wang JZ, Liu R, Feng J, Wang X. Biomarkers used in Alzheimer's disease diagnosis, treatment, and prevention. Ageing Res Rev 2022; 74:101544. [PMID: 34933129 DOI: 10.1016/j.arr.2021.101544] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD), being the number one in terms of dementia burden, is an insidious age-related neurodegenerative disease and is presently considered a global public health threat. Its main histological hallmarks are the Aβ senile plaques and the P-tau neurofibrillary tangles, while clinically it is marked by a progressive cognitive decline that reflects the underlying synaptic loss and neurodegeneration. Many of the drug therapies targeting the two pathological hallmarks namely Aβ and P-tau have been proven futile. This is probably attributed to the initiation of therapy at a stage where cognitive alterations are already obvious. In other words, the underlying neuropathological changes are at a stage where these drugs lack any therapeutic value in reversing the damage. Therefore, there is an urgent need to start treatment in the very early stage where these changes can be reversed, and hence, early diagnosis is of primordial importance. To this aim, the use of robust and informative biomarkers that could provide accurate diagnosis preferably at an earlier phase of the disease is of the essence. To date, several biomarkers have been established that, to a different extent, allow researchers and clinicians to evaluate, diagnose, and more specially exclude other related pathologies. In this study, we extensively reviewed data on the currently explored biomarkers in terms of AD pathology-specific and non-specific biomarkers and highlighted the recent developments in the diagnostic and theragnostic domains. In the end, we have presented a separate elaboration on aspects of future perspectives and concluding remarks.
Collapse
|
25
|
Laffoon SB, Doecke JD, Roberts AM, Vance JA, Reeves BD, Pertile KK, Rumble RL, Fowler CJ, Trounson B, Ames D, Martins R, Bush AI, Masters CL, Grieco PA, Dratz EA, Roberts BR. Analysis of plasma proteins using 2D gels and novel fluorescent probes: in search of blood based biomarkers for Alzheimer's disease. Proteome Sci 2022; 20:2. [PMID: 35081972 PMCID: PMC8790928 DOI: 10.1186/s12953-021-00185-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/23/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The Australian Imaging and Biomarker Lifestyle (AIBL) study of aging is designed to aid the discovery of biomarkers. The current study aimed to discover differentially expressed plasma proteins that could yield a blood-based screening tool for Alzheimer's disease. METHODS The concentration of proteins in plasma covers a vast range of 12 orders of magnitude. Therefore, to search for medium to low abundant biomarkers and elucidate mechanisms of AD, we immuno-depleted the most abundant plasma proteins and pre-fractionated the remaining proteins by HPLC, prior to two-dimensional gel electrophoresis. The relative levels of approximately 3400 protein species resolved on the 2D gels were compared using in-gel differential analysis with spectrally resolved fluorescent protein detection dyes (Zdyes™). Here we report on analysis of pooled plasma samples from an initial screen of a sex-matched cohort of 72 probable AD patients and 72 healthy controls from the baseline time point of AIBL. RESULTS We report significant changes in variants of apolipoprotein E, haptoglobin, α1 anti-trypsin, inter-α trypsin inhibitor, histidine-rich glycoprotein, and a protein of unknown identity. α1 anti-trypsin and α1 anti-chymotrypsin demonstrated plasma concentrations that were dependent on APOE ε4 allele dose. Our analysis also identified an association with the level of Vitamin D binding protein fragments and complement factor I with sex. We then conducted a preliminary validation study, on unique individual samples compared to the discovery cohort, using a targeted LC-MS/MS assay on a subset of discovered biomarkers. We found that targets that displayed a high degree of isoform specific changes in the 2D gels were not changed in the targeted MS assay which reports on the total level of the biomarker. CONCLUSIONS This demonstrates that further development of mass spectrometry assays is needed to capture the isoform complexity that exists in theses biological samples. However, this study indicates that a peripheral protein signature has potential to aid in the characterization of AD.
Collapse
Affiliation(s)
- Scott B. Laffoon
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne Dementia Research Centre, Parkville, VIC 3010 Australia
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59715 USA
- Cooperative Research Centre for Mental Health, Carlton South, VIC Australia
| | - James D. Doecke
- Australian e-Health Research Centre, CSIRO and Cooperative Research Centre of Mental Health, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4029 Australia
| | - Anne M. Roberts
- Department of Biochemistry, Emory School of Medicine, 4001 Rollins Research Building, Atlanta, GA 30322 USA
- Department of Neurology, Emory School of Medicine, 4001 Rollins Research Building, Atlanta, GA 30322 USA
| | | | - Benjamin D. Reeves
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59715 USA
| | - Kelly K. Pertile
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne Dementia Research Centre, Parkville, VIC 3010 Australia
| | - Rebecca L. Rumble
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne Dementia Research Centre, Parkville, VIC 3010 Australia
| | - Chris J. Fowler
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne Dementia Research Centre, Parkville, VIC 3010 Australia
| | - Brett Trounson
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne Dementia Research Centre, Parkville, VIC 3010 Australia
| | - David Ames
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne Dementia Research Centre, Parkville, VIC 3010 Australia
| | - Ralph Martins
- Cooperative Research Centre for Mental Health, Carlton South, VIC Australia
- School of Medical Sciences, Edith Cowan University, Joondalup, WA Australia
- Department of Biomedical Sciences, Macquarie University, North Ryde, NSW Australia
| | - Ashley I. Bush
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne Dementia Research Centre, Parkville, VIC 3010 Australia
- Cooperative Research Centre for Mental Health, Carlton South, VIC Australia
| | - Colin L. Masters
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne Dementia Research Centre, Parkville, VIC 3010 Australia
- Cooperative Research Centre for Mental Health, Carlton South, VIC Australia
| | - Paul A. Grieco
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59715 USA
| | - Edward A. Dratz
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59715 USA
| | - Blaine R. Roberts
- Department of Biochemistry, Emory School of Medicine, 4001 Rollins Research Building, Atlanta, GA 30322 USA
- Department of Neurology, Emory School of Medicine, 4001 Rollins Research Building, Atlanta, GA 30322 USA
| |
Collapse
|
26
|
Evidence of plasma biomarkers indicating high risk of dementia in cognitively normal subjects. Sci Rep 2022; 12:1192. [PMID: 35075194 PMCID: PMC8786959 DOI: 10.1038/s41598-022-05177-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/07/2022] [Indexed: 11/08/2022] Open
Abstract
Subjects with comorbidities are at risk for neurodegeneration. There is a lack of a direct relationship between comorbidities and neurodegeneration. In this study, immunomagnetic reduction (IMR) assays were utilized to assay plasma Aβ1-42 and total tau protein (T-Tau) levels in poststroke (PS, n = 27), family history of Alzheimer's disease (ADFH, n = 35), diabetes (n = 21), end-stage renal disease (ESRD, n = 41), obstructive sleep apnea (OSA, n = 20), Alzheimer's disease (AD, n = 65). Thirty-seven healthy controls (HCs) were enrolled. The measured concentrations of plasma Aβ1-42 were 14.26 ± 1.42, 15.43 ± 1.76, 15.52 ± 1.60, 16.15 ± 1.05, 16.52 ± 0.59, 15.97 ± 0.54 and 20.06 ± 3.09 pg/mL in HC, PS, ADFH, diabetes, ESRD, OSA and AD groups, respectively. The corresponding concentrations of plasma T-Tau were 15.13 ± 3.62, 19.29 ± 8.01, 17.93 ± 6.26, 19.74 ± 2.92, 21.54 ± 2.72, 20.17 ± 2.77 and 41.24 ± 14.64 pg/mL. The plasma levels of Aβ1-42 and T-Tau in were significantly higher in the PS, ADFH, diabetes, ESRD and OSA groups than controls (Aβ1-42 in PS: 15.43 ± 1.76 pg/mL vs. 14.26 ± 1.42 pg/mL, p < 0.005; T-Tau in PS: 19.29 ± 8.01 vs. 15.13 ± 3.62 pg/mL, p < 0.005, Aβ1-42 in ADFH: 15.52 ± 1.60 pg/mL vs. 14.26 ± 1.42 pg/mL, p < 0.001; T-Tau in ADFH: 17.93 ± 6.26 vs. 15.13 ± 3.62 pg/mL, p < 0.005, Aβ1-42 in diabetes: 16.15 ± 1.05 pg/mL vs. 14.26 ± 1.42 pg/mL, p < 0.001; T-Tau in diabetes: 19.74 ± 2.92 vs. 15.13 ± 3.62 pg/mL, p < 0.001, Aβ1-42 in ESRD: 16.52 ± 0.59 pg/mL vs. 14.26 ± 1.42 pg/mL, p < 0.001; T-Tau in ESRD: 21.54 ± 2.72 vs. 15.13 ± 3.62 pg/mL, p < 0.001, Aβ1-42 in OSA: 15.97 ± 0.54 pg/mL vs. 14.26 ± 1.42 pg/mL, p < 0.001; T-Tau in OSA: 20.17 ± 2.77 vs. 15.13 ± 3.62 pg/mL, p < 0.001). This evidence indicates the high risk for dementia in these groups from the perspective of plasma biomarkers.
Collapse
|
27
|
Zheng Y, Ji B, Chen S, Zhou R, Ni R. The impact of uremic toxins on Alzheimer's disease. Curr Alzheimer Res 2022; 19:104-118. [PMID: 35048807 DOI: 10.2174/1567205019666220120113305] [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: 08/18/2021] [Revised: 11/27/2021] [Accepted: 12/14/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia, pathologically characterized by accumulation of senile plaques and neurofibrillary tangles. Chronic kidney disease (CKD) is highly prevalent in elderly population closely associated with occurrence of dementia. Recent epidemiological and experimental studies suggest a potential association of CKD with AD. Both diseases share a panel of identical risk factors, such as type 2 diabetes; and hypertension. However, the relationship between CKD and AD is unclear. Lower clearance of a panel of uremic toxin including cystatin-C, guanidine, and adiponectin due to CKD is implied to contribute to AD pathogenesis. In this review we summarize the current evidence from epidemiological, experimental and clinical studies on the potential contribution of uremic toxins to AD pathogenesis. We describe outstanding questions and propose an outlook on the link between uremic toxins and AD.
Collapse
Affiliation(s)
- Yuqi Zheng
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Sijun Chen
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rong Zhou
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
Hao N, Wang Z, Liu P, Becker R, Yang S, Yang K, Pei Z, Zhang P, Xia J, Shen L, Wang L, Welsh-Bohmer KA, Sanders L, Lee LP, Huang TJ. Acoustofluidic multimodal diagnostic system for Alzheimer's disease. Biosens Bioelectron 2022; 196:113730. [PMID: 34736099 PMCID: PMC8643320 DOI: 10.1016/j.bios.2021.113730] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/13/2021] [Accepted: 10/23/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative brain disorder that affects tens of millions of older adults worldwide and has significant economic and societal impacts. Despite its prevalence and severity, early diagnosis of AD remains a considerable challenge. Here we report an integrated acoustofluidics-based diagnostic system (ADx), which combines triple functions of acoustics, microfluidics, and orthogonal biosensors for clinically accurate, sensitive, and rapid detection of AD biomarkers from human plasma. We design and fabricate a surface acoustic wave-based acoustofluidic separation device to isolate and purify AD biomarkers to increase the signal-to-noise ratio. Multimodal biosensors within the integrated ADx are fabricated by in-situ patterning of the ZnO nanorod array and deposition of Ag nanoparticles onto the ZnO nanorods for surface-enhanced Raman scattering (SERS) and electrochemical immunosensors. We obtain the label-free detections of SERS and electrochemical immunoassay of clinical plasma samples from AD patients and healthy controls with high sensitivity and specificity. We believe that this efficient integration provides promising solutions for the early diagnosis of AD.
Collapse
Affiliation(s)
- Nanjing Hao
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Zeyu Wang
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Pengzhan Liu
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Ryan Becker
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Shujie Yang
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Kaichun Yang
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Zhichao Pei
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Peiran Zhang
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Jianping Xia
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Liang Shen
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Lin Wang
- Ascent Bio-Nano Technologies, Inc., Morrisville, NC, 27560, USA
| | | | - Laurie Sanders
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Luke P Lee
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA, 94720, USA; Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Tony Jun Huang
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
29
|
Wu Y, Wang Z, Yin J, Yang B, Fan J, Cheng Z. Association Plasma Aβ42 Levels with Alzheimer's Disease and Its Influencing Factors in Chinese Elderly Population. Neuropsychiatr Dis Treat 2022; 18:1831-1841. [PMID: 36043117 PMCID: PMC9420413 DOI: 10.2147/ndt.s374722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/17/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Intracerebral Aβ protein deposition is an important pathological mechanism of Alzheimer's disease (AD) and is one of the indicators of early diagnosis of AD. However, invasive lumbar puncture and Aβ PET are difficult to perform in primary units, resulting delays in early diagnosis of AD. In recent years, it has been found that plasma Aβ can reflect the pathological state of AD in early stage, but the results are not consistent. The objective of this study was to explore the association between plasma Aβ42 levels and AD cognitive impairment and its influencing factors in Chinese elderly population, so as to provide guidance for the clinical application of plasma Aβ42 as a blood biomarker of AD. METHODS This is a cross-sectional study based on the community population. Plasma samples were collected from 604 healthy controls (HC), 508 mild cognitive impairment (MCI) and 202 dementia with Alzheimer's type (DAT) patients from three cities. We analyzed the correlation between plasma Aβ42 levels and cognitive function and the influence of confounding factors on the relationship between plasma Aβ42 levels and AD. The independent influencing factors of plasma Aβ42 levels were determined by covariance and linear regression analysis. RESULTS Our results suggest that there is a special linear relationship between plasma Aβ42 and cognitive impairment of AD in Chinese elderly population, with Aβ42 levels slightly decreased in early AD and significantly increased in moderate-to-severe AD (P<0.01). There are many factors influencing the association between plasma Aβ42 levels and AD cognitive impairment, and sample source, gender and BMI are independent influencing factors of plasma Aβ42. CONCLUSION This indentifies that plasma Aβ42 may be a peripheral biomarker for AD screening in Chinese elderly population, but it is necessary to establish standardized detection methods and establish different demarcation criteria for various influencing factors.
Collapse
Affiliation(s)
- Yue Wu
- Department of Geriatric Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi, People's Republic of China
| | - Zhiqiang Wang
- Department of Clinical Psychology, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi, People's Republic of China
| | - Jiajun Yin
- Brain Science Basic Laboratory, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi, People's Republic of China
| | - Bixiu Yang
- Department of Clinical Psychology, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi, People's Republic of China
| | - Jie Fan
- Department of Geriatric Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi, People's Republic of China
| | - Zaohuo Cheng
- Department of Geriatric Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi, People's Republic of China
| |
Collapse
|
30
|
Huang KL, Hsiao IT, Chang TY, Yang SY, Chang YJ, Wu HC, Liu CH, Wu YM, Lin KJ, Ho MY, Lee TH. Neurodegeneration and Vascular Burden on Cognition After Midlife: A Plasma and Neuroimaging Biomarker Study. Front Hum Neurosci 2022; 15:735063. [PMID: 34970128 PMCID: PMC8712753 DOI: 10.3389/fnhum.2021.735063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Objectives: Neurodegeneration and vascular burden are the two most common causes of post-stroke cognitive impairment. However, the interrelationship between the plasma beta-amyloid (Aβ) and tau protein, cortical atrophy and brain amyloid accumulation on PET imaging in stroke patients is undetermined. We aimed to explore: (1) the relationships of cortical thickness and amyloid burden on PET with plasma Aβ40, Aβ42, tau protein and their composite scores in stroke patients; and (2) the associations of post-stroke cognitive presentations with these plasma and neuroimaging biomarkers. Methods: The prospective project recruited first-ever ischemic stroke patients around 3 months after stroke onset. The plasma Aβ40, Aβ42, and total tau protein were measured with the immunomagnetic reduction method. Cortical thickness was evaluated on MRI, and cortical amyloid plaque deposition was evaluated by 18F-florbetapir PET. Cognition was evaluated with Mini-Mental State Examination (MMSE), Geriatric Depression Scale (GDS), Dementia Rating Scale-2 (DRS-2). Results: The study recruited 24 stroke patients and 13 normal controls. The plasma tau and tau*Aβ42 levels were correlated with mean cortical thickness after age adjustment. The Aβ42/Aβ40 ratio was correlated with global cortical 18F-florbetapir uptake value. The DRS-2 and GDS scores were associated with mean cortical thickness and plasma biomarkers, including Aβ42/Aβ40, tau, tau*Aβ42, tau/Aβ42, and tau/Aβ40 levels, in stroke patients. Conclusion: Plasma Aβ, tau, and their composite scores were associated with cognitive performance 3 months after stroke, and these plasma biomarkers were correlated with corresponding imaging biomarkers of neurodegeneration. Further longitudinal studies with a larger sample size are warranted to replicate the study results.
Collapse
Affiliation(s)
- Kuo-Lun Huang
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ing-Tsung Hsiao
- Department of Nuclear Medicine and Molecular Imaging Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Healthy Aging Research Center and Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Yu Chang
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | | | - Yeu-Jhy Chang
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsiu-Chuan Wu
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Hung Liu
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Ming Wu
- Department of Radiology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Molecular Imaging Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Healthy Aging Research Center and Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Meng-Yang Ho
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Behavioral Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Tsong-Hai Lee
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
31
|
Serum Glycoproteomics and Identification of Potential Mechanisms Underlying Alzheimer’s Disease. Behav Neurol 2021; 2021:1434076. [PMID: 34931130 PMCID: PMC8684523 DOI: 10.1155/2021/1434076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/04/2021] [Accepted: 11/16/2021] [Indexed: 02/07/2023] Open
Abstract
Objectives. This study compares glycoproteomes in Thai Alzheimer’s disease (AD) patients with those of cognitively normal individuals. Methods. Study participants included outpatients with clinically diagnosed AD (
) and healthy controls without cognitive impairment (
). Blood samples were collected from all participants for biochemical analysis and for
(APOE) genotyping by real-time TaqMan PCR assays. Comparative serum glycoproteomic profiling by liquid chromatography-tandem mass spectrometry was then performed to identify differentially abundant proteins with functional relevance. Results. Statistical differences in age, educational level, and APOE ɛ3/ɛ4 and ɛ4/ɛ4 haplotype frequencies were found between the AD and control groups. The frequency of the APOE ɛ4 allele was significantly higher in the AD group than in the control group. In total, 871 glycoproteins were identified, including 266 and 259 unique proteins in control and AD groups, respectively. There were 49 and 297 upregulated and downregulated glycoproteins, respectively, in AD samples compared with the controls. Unique AD glycoproteins were associated with numerous pathways, including Alzheimer’s disease-presenilin pathway (16.6%), inflammation pathway mediated by chemokine and cytokine signaling (9.2%), Wnt signaling pathway (8.2%), and apoptosis signaling pathway (6.7%). Conclusion. Functions and pathways associated with protein-protein interactions were identified in AD. Significant changes in these proteins can indicate the molecular mechanisms involved in the pathogenesis of AD, and they have the potential to serve as AD biomarkers. Such findings could allow us to better understand AD pathology.
Collapse
|
32
|
Hu CJ, Chiu MJ, Pai MC, Yan SH, Wang PN, Chiu PY, Lin CH, Chen TF, Yang FC, Huang KL, Hsu YT, Hou YC, Lin WC, Lu CH, Huang LK, Yang SY. Assessment of High Risk for Alzheimer's Disease Using Plasma Biomarkers in Subjects with Normal Cognition in Taiwan: A Preliminary Study. J Alzheimers Dis Rep 2021; 5:761-770. [PMID: 34870102 PMCID: PMC8609520 DOI: 10.3233/adr-210310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background In Alzheimer's disease (AD), cognitive impairment begins 10-15 years later than neurodegeneration in the brain. Plasma biomarkers are promising candidates for assessing neurodegeneration in people with normal cognition. It has been reported that subjects with the concentration of plasma amyloid-β 1-42×total tau protein higher than 455 pg2/ml2 are assessed as having a high risk of amnesic mild impairment or AD, denoted as high risk of AD (HRAD). Objective The prevalence of high-risk for dementia in cognitively normal controls is explored by assaying plasma biomarkers. Methods 422 subjects with normal cognition were enrolled around Taiwan. Plasma Aβ1-40, Aβ1-42, and T-Tau levels were assayed using immunomagnetic reduction to assess the risk of dementia. Results The results showed that 4.6% of young adults (age: 20-44 years), 8.5% of middle-aged adults (age: 45-64 years), and 7.3% of elderly adults (age: 65-90 years) had HRAD. The percentage of individuals with HRAD dramatically increased in middle-aged and elderly adults compared to young adults. Conclusion The percentage of HRAD in cognitively normal subjects are approximately 10%, which reveals that the potentially public-health problem of AD in normal population. Although the subject having abnormal levels of Aβ or tau is not definitely going on to develop cognitive declines or AD, the risk of suffering cognitive impairment in future is relatively high. Suitable managements are suggested for these high-risk cognitively normal population. Worth noting, attention should be paid to preventing cognitive impairment due to AD, not only in elderly adults but also middle-aged adults.
Collapse
Affiliation(s)
- Chaur-Jong Hu
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurology, Dementia Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Psychology, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Ming-Chyi Pai
- Division of Behavioral Neurology, Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sui-Hing Yan
- Department of Neurology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Pei-Ning Wang
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Neurology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Pai-Yi Chiu
- Department of Neurology, Show Chwan Memorial Hospital, Chunghwa, Taiwan.,MR-guided Focus Ultrasound Center, Chang Bin Show Chwan Memorial Hospital, Chunghwa, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Fu-Chi Yang
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Lun Huang
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Ting Hsu
- Department of Neurology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yi-Chou Hou
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Internal Medicine, Cardinal Tien Hospital, New Taipei City, Taiwan.,School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wei-Che Lin
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Cheng-Hsien Lu
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Li-Kai Huang
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurology, Dementia Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | | |
Collapse
|
33
|
Jiao B, Liu H, Guo L, Liao X, Zhou Y, Weng L, Xiao X, Zhou L, Wang X, Jiang Y, Yang Q, Zhu Y, Zhou L, Zhang W, Wang J, Yan X, Tang B, Shen L. Performance of Plasma Amyloid β, Total Tau, and Neurofilament Light Chain in the Identification of Probable Alzheimer's Disease in South China. Front Aging Neurosci 2021; 13:749649. [PMID: 34776933 PMCID: PMC8579066 DOI: 10.3389/fnagi.2021.749649] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/24/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Alzheimer's disease (AD) is the most common type of dementia and has no effective treatment to date. It is essential to develop a minimally invasive blood-based biomarker as a tool for screening the general population, but the efficacy remains controversial. This cross-sectional study aimed to evaluate the ability of plasma biomarkers, including amyloid β (Aβ), total tau (t-tau), and neurofilament light chain (NfL), to detect probable AD in the South Chinese population. Methods: A total of 277 patients with a clinical diagnosis of probable AD and 153 healthy controls with normal cognitive function (CN) were enrolled in this study. The levels of plasma Aβ42, Aβ40, t-tau, and NfL were detected using ultra-sensitive immune-based assays (SIMOA). Lumbar puncture was conducted in 89 patients with AD to detect Aβ42, Aβ40, t-tau, and phosphorylated (p)-tau levels in the cerebrospinal fluid (CSF) and to evaluate the consistency between plasma and CSF biomarkers through correlation analysis. Finally, the diagnostic value of plasma biomarkers was further assessed by constructing a receiver operating characteristic (ROC) curve. Results: After adjusting for age, sex, and the apolipoprotein E (APOE) alleles, compared to the CN group, the plasma t-tau, and NfL were significantly increased in the AD group (p < 0.01, Bonferroni correction). Correlation analysis showed that only the plasma t-tau level was positively correlated with the CSF t-tau levels (r = 0.319, p = 0.003). The diagnostic model combining plasma t-tau and NfL levels, and age, sex, and APOE alleles, showed the best performance for the identification of probable AD [area under the curve (AUC) = 0.89, sensitivity = 82.31%, specificity = 83.66%]. Conclusion: Blood biomarkers can effectively distinguish patients with probable AD from controls and may be a non-invasive and efficient method for AD pre-screening.
Collapse
Affiliation(s)
- Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Hui Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lina Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxin Liao
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yafang Zhou
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Ling Weng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Xuewen Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yaling Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qijie Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Zhou
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Weiwei Zhang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Xinxiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| |
Collapse
|
34
|
Yun G, Kim HJ, Kim HG, Lee KM, Hong IK, Kim SH, Rhee HY, Jahng GH, Yoon SS, Park KC, Hwang KS, Lee JS. Association Between Plasma Amyloid-β and Neuropsychological Performance in Patients With Cognitive Decline. Front Aging Neurosci 2021; 13:736937. [PMID: 34759814 PMCID: PMC8573146 DOI: 10.3389/fnagi.2021.736937] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/07/2021] [Indexed: 01/10/2023] Open
Abstract
Objective: To investigate the association between plasma amyloid-β (Aβ) levels and neuropsychological performance in patients with cognitive decline using a highly sensitive nano-biosensing platform. Methods: We prospectively recruited 44 patients with cognitive decline who underwent plasma Aβ analysis, amyloid positron emission tomography (PET) scanning, and detailed neuropsychological tests. Patients were classified into a normal control (NC, n = 25) or Alzheimer’s disease (AD, n = 19) group based on amyloid PET positivity. Multiple linear regression was performed to determine whether plasma Aβ (Aβ40, Aβ42, and Aβ42/40) levels were associated with neuropsychological test results. Results: The plasma levels of Aβ42/40 were significantly different between the NC and AD groups and were the best predictor of amyloid PET positivity by receiver operating characteristic curve analysis [area under the curve of 0.952 (95% confidence interval, 0.892–1.000)]. Although there were significant differences in the neuropsychological performance of cognitive domains (language, visuospatial, verbal/visual memory, and frontal/executive functions) between the NC and AD groups, higher levels of plasma Aβ42/40 were negatively correlated only with verbal and visual memory performance. Conclusion: Our results demonstrated that plasma Aβ analysis using a nano-biosensing platform could be a useful tool for diagnosing AD and assessing memory performance in patients with cognitive decline.
Collapse
Affiliation(s)
- Gyihyaon Yun
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Hye Jin Kim
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Hyug-Gi Kim
- Department of Radiology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Kyung Mi Lee
- Department of Radiology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Il Ki Hong
- Department of Nuclear Medicine, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Sang Hoon Kim
- Department of Otorhinolaryngology, Head and Neck Surgery, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Hak Young Rhee
- Department of Neurology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Geon-Ho Jahng
- Department of Radiology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Sung Sang Yoon
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Key-Chung Park
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Kyo Seon Hwang
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Jin San Lee
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, South Korea
| |
Collapse
|
35
|
Park S, Kim Y. Bias-generating factors in biofluid amyloid-β measurements for Alzheimer's disease diagnosis. Biomed Eng Lett 2021; 11:287-295. [PMID: 34616582 DOI: 10.1007/s13534-021-00201-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia worldwide, yet the dearth of readily accessible diagnostic biomarkers is a substantial hindrance towards progressing to effective preventive and therapeutic approaches. Due to a long delay between cerebral amyloid-β (Aβ) accumulation and the onset of cognitive impairments, biomarkers that reflect Aβ pathology and enable routine screening for disease progression are of urgent need for application in the clinical diagnosis of AD. According to accumulating evidences, cerebrospinal fluid (CSF) and plasma offer windows to the brain as they allow monitoring of biochemical changes in the brain. Considering the high availability and accuracy in depicting Aβ deposition in the brain, Aβ levels in CSF and plasma are regarded as promising fluid biomarkers for the diagnosis of AD patients at an early stage. However, clinical data with intra- and interindividual variations in the concentrations of CSF and plasma Aβ implicate the need to reevaluate current Aβ detection methods and establish a standardized operating procedure. Therefore, this review introduces three bias-generating factors in biofluid Aβ measurement that may hamper the accurate Aβ quantification and how such complications can be overcome for the widespread implementation of fluid Aβ detection in clinical practice.
Collapse
Affiliation(s)
- Sohui Park
- Department of Pharmacy, Department of Integrative Biotechnology and Translational Medicine, and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983 Republic of Korea
| | - YoungSoo Kim
- Department of Pharmacy, Department of Integrative Biotechnology and Translational Medicine, and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983 Republic of Korea
| |
Collapse
|
36
|
Koychev I, Jansen K, Dette A, Shi L, Holling H. Blood-Based ATN Biomarkers of Alzheimer's Disease: A Meta-Analysis. J Alzheimers Dis 2021; 79:177-195. [PMID: 33252080 DOI: 10.3233/jad-200900] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The Amyloid Tau Neurodegeneration (ATN) framework was proposed to define the biological state underpinning Alzheimer's disease (AD). Blood-based biomarkers offer a scalable alternative to the costly and invasive currently available biomarkers. OBJECTIVE In this meta-analysis we sought to assess the diagnostic performance of plasma amyloid (Aβ40, Aβ42, Aβ42/40 ratio), tangle (p-tau181), and neurodegeneration (total tau [t-tau], neurofilament light [NfL]) biomarkers. METHODS Electronic databases were screened for studies reporting biomarker concentrations for AD and control cohorts. Biomarker performance was examined by random-effect meta-analyses based on the ratio between biomarker concentrations in patients and controls. RESULTS 83 studies published between 1996 and 2020 were included in the analyses. Aβ42/40 ratio as well as Aβ42 discriminated AD patients from controls when using novel platforms such as immunomagnetic reduction (IMR). We found significant differences in ptau-181 concentration for studies based on single molecule array (Simoa), but not for studies based on IMR or ELISA. T-tau was significantly different between AD patients and control in IMR and Simoa but not in ELISA-based studies. In contrast, NfL differentiated between groups across platforms. Exosome studies showed strong separation between patients and controls for Aβ42, t-tau, and p-tau181. CONCLUSION Currently available assays for sampling plasma ATN biomarkers appear to differentiate between AD patients and controls. Novel assay methodologies have given the field a significant boost for testing these biomarkers, such as IMR for Aβ, Simoa for p-tau181. Enriching samples through extracellular vesicles shows promise but requires further validation.
Collapse
Affiliation(s)
- Ivan Koychev
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Katrin Jansen
- Department of Psychology, University of Münster, Münster, Germany
| | - Alina Dette
- Department of Psychology, University of Münster, Münster, Germany
| | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Heinz Holling
- Department of Psychology, University of Münster, Münster, Germany
| |
Collapse
|
37
|
Liu HC, Chen HH, Ho CS, Chang JF, Lin CC, Chiu MJ, Chen TF, Hu CJ, Yan SH, Sun Y, Yang SY. Investigation of the Number of Tests Required for Assaying Plasma Biomarkers Associated with Alzheimer's Disease Using Immunomagnetic Reduction. Neurol Ther 2021; 10:1015-1028. [PMID: 34515952 PMCID: PMC8571465 DOI: 10.1007/s40120-021-00280-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/25/2021] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Concentrations of plasma biomarkers associated with Alzheimer's disease have been reported to be as low as several tens of picograms/milliliter (pg/ml). However, in assays measuring these biomarkers, it is likely that repeated measurements are necessary to obtain reliable values. METHODS We performed assays as a single test or as duplicate, quadruplicate, fivefold and tenfold repeated tests, on samples spiked with different concentrations of amyloid β 1-40 (Aβ1-40; 1-1000 pg/ml), Aβ1-42 (1-30,000 pg/ml) and total Tau protein (T-Tau; 0.1-1000 pg/ml), with the aim to to calculate the coefficients of variation (CVs). RESULTS The results demonstrated common changes in the CVs with changes in the number of tests for a given sample: the CVs decreased with increases in the number of tests from one to ten. All CV values were distributed within the range of 0.35 to 15.5%; as such, the CV values were all lower than the acceptable value of 20%. CONCLUSION Based on this study, a single assay of Aβ1-40, Aβ1-42 and T-Tau, respectively, provides reliable results in terms of the measurement of that plasma biomarker.
Collapse
Affiliation(s)
| | | | | | | | | | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Taipei Medical University Shuang-Ho Hospital, New Taipei City, 235, Taiwan
| | - Sui-Hing Yan
- Department of Neurology, Far Eastern Memorial Hospital, New Taipei City, 220, Taiwan
| | - Yu Sun
- Department of Neurology, En Chu Kong Hospital, New Taipei City, 237, Taiwan
| | | |
Collapse
|
38
|
Campese N, Beatino MF, Del Gamba C, Belli E, Giampietri L, Del Prete E, Galgani A, Vergallo A, Siciliano G, Ceravolo R, Hampel H, Baldacci F. Ultrasensitive techniques and protein misfolding amplification assays for biomarker-guided reconceptualization of Alzheimer's and other neurodegenerative diseases. Expert Rev Neurother 2021; 21:949-967. [PMID: 34365867 DOI: 10.1080/14737175.2021.1965879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The clinical validation and qualification of biomarkers reflecting the complex pathophysiology of neurodegenerative diseases (NDDs) is a fundamental challenge for current drug discovery and development and next-generation clinical practice. Novel ultrasensitive detection techniques and protein misfolding amplification assays hold the potential to optimize and accelerate this process. AREAS COVERED Here we perform a PubMed-based state of the art review and perspective report on blood-based ultrasensitive detection techniques and protein misfolding amplification assays for biomarkers discovery and development in NDDs. EXPERT OPINION Ultrasensitive assays represent innovative solutions for blood-based assessments during the entire Alzheimer's disease (AD) biological and clinical continuum, for contexts of use (COU) such as prediction, detection, early diagnosis, and prognosis of AD. Moreover, cerebrospinal fluid (CSF)-based misfolding amplification assays show encouraging performance in detecting α-synucleinopathies in prodromal or at-high-risk individuals and may serve as tools for patients' stratification by the presence of α-synuclein pathology. Further clinical research will help overcome current methodological limitations, also through exploring multiple accessible bodily matrices. Eventually, integrative longitudinal studies will support precise definitions for appropriate COU across NDDs.
Collapse
Affiliation(s)
- Nicole Campese
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Claudia Del Gamba
- Neurology Unit, Nuovo Ospedale Santo Stefano, Via Suor Niccolina Infermiera 20, Prato, Italy
| | - Elisabetta Belli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Linda Giampietri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eleonora Del Prete
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alessandro Galgani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Vergallo
- Sorbonne University, GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié- Salpêtrière Hospital, Boulevard De L'hôpital, Paris, France
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Harald Hampel
- Sorbonne University, GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié- Salpêtrière Hospital, Boulevard De L'hôpital, Paris, France
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Sorbonne University, GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié- Salpêtrière Hospital, Boulevard De L'hôpital, Paris, France
| |
Collapse
|
39
|
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
|
40
|
Hou YC, Huang CL, Lu CL, Zheng CM, Lin YF, Lu KC, Chung YL, Chen RM. The Role of Plasma Neurofilament Light Protein for Assessing Cognitive Impairment in Patients With End-Stage Renal Disease. Front Aging Neurosci 2021; 13:657794. [PMID: 34122041 PMCID: PMC8192845 DOI: 10.3389/fnagi.2021.657794] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 05/03/2021] [Indexed: 12/26/2022] Open
Abstract
Introduction: End-stage renal disease (ESRD) is defined as the irreversible loss of renal function, necessitating renal replacement therapy. Patients with ESRD tend to have more risk factors for cognitive impairment than the general population, including hypertension, accumulative uremic toxin, anemia, and old age. The association between these risk factors and the pathologic protein was lacking. Blood-based assays for detecting pathologic protein, such as amyloid beta (Aβ), total tau protein, and neurofilament light chain (NfL), have the advantages of being less invasive and more cost-effective for diagnosing patients with cognitive impairment. The aim of the study is to validate if the common neurologic biomarkers were different in ESRD patients and to differentiate if the specific biomarkers could correlate with specific correctable risk factors. Methods: In total, 67 participants aged >45 years were enrolled. The definition of ESRD was receiving maintenance hemodialysis for >3 months. Cognitive impairment was defined as a Mini-Mental State Examination score of <24. The participants were divided into groups for ESRD with and without cognitive impairment. The blood-based biomarkers (tau protein, Aβ1/40, Aβ1/42, and NfL) were analyzed through immunomagnetic reduction assay. Other biochemical and hematologic data were obtained simultaneously. Summary of results: The study enrolled 43 patients with ESRD who did not have cognitive impairment and 24 patients with ESRD who had cognitive impairment [Mini-Mental State Examination (MMSE): 27.60 ± 1.80 vs. 16.84 ± 6.40, p < 0.05]. Among the blood-based biomarkers, NfL was marginally higher in the ESRD with cognitive impairment group than in the ESRD without cognitive impairment group (10.41 ± 3.26 vs. 8.74 ± 2.81 pg/mL, p = 0.037). The concentrations of tau protein, amyloid β 1/42, and amyloid β 1/40 (p = 0.504, 0.393, and 0.952, respectively) were similar between the two groups. The area under the curve of NfL to distinguish cognitively impaired and unimpaired ESRD patients was 0.687 (95% confidence interval: 0.548-0.825, p = 0.034). There was no correlation between the concentration of NfL and MMSE among total population (r = -0.153, p = 0.277), patients with (r = 0.137, p = 0.583) or without cognitive impairment (r = 0.155, p = 0.333). Conclusion: Patients with ESRD who had cognitive impairment had marginally higher plasma NfL concentrations. NfL concentration was not correlated with the biochemical parameters, total MMSE among total population or individual groups with or without cognitive impairment. The concentrations of Aβ1/40, Aβ1/42, and tau were similar between the groups.
Collapse
Affiliation(s)
- Yi-Chou Hou
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Internal Medicine, Cardinal Tien Hospital, New Taipei City, Taiwan.,School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chuen-Lin Huang
- Department of Medical Research, Cardinal Tien Hospital, New Taipei City, Taiwan.,Department of Physiology and Biophysics, National Defense Medical Center, Graduate Institute of Physiology, Taipei, Taiwan
| | - Chien-Lin Lu
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.,Department of Nephrology, Fu Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Cai-Mei Zheng
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Medical University-Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Yuh-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Medical University-Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan.,National Defense Medical Center, Graduate Institute of Medical Sciences, Taipei, Taiwan
| | - Kuo-Cheng Lu
- Department of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Ya-Lin Chung
- Department of Medical Laboratory, Cardinal-Tien Hospital, New Taipei City, Taiwan
| | - Ruei-Ming Chen
- TMU Research Center of Cancer Translational Medicine, Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
41
|
Soares Martins T, Magalhães S, Rosa IM, Vogelgsang J, Wiltfang J, Delgadillo I, Catita J, da Cruz E Silva OAB, Nunes A, Henriques AG. Potential of FTIR Spectroscopy Applied to Exosomes for Alzheimer's Disease Discrimination: A Pilot Study. J Alzheimers Dis 2021; 74:391-405. [PMID: 32039849 DOI: 10.3233/jad-191034] [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: 12/23/2022]
Abstract
Alzheimer's disease (AD) diagnosis is based on psychological and imaging tests but can also include monitoring cerebrospinal fluid (CSF) biomarkers. However, CSF based-neurochemical approaches are expensive and invasive, limiting their use to well-equipped settings. In contrast, blood-based biomarkers are minimally invasive, cost-effective, and a widely accessible alternative. Blood-derived exosomes have recently emerged as a reliable AD biomarker source, carrying disease-specific cargo. Fourier-transformed infrared (FTIR) spectroscopy meets the criteria for an ideal diagnostic methodology since it is rapid, easy to implement, and has high reproducibility. This metabolome-based technique is useful for diagnosing a broad range of diseases, although to our knowledge, no reports for FTIR spectroscopy applied to exosomes in AD exist. In this ground-breaking pilot study, FTIR spectra of serum and serum-derived exosomes from two independent cohorts were acquired and analyzed using multivariate analysis. The regional UA-cohort includes 9 individuals, clinically diagnosed with AD, mean age of 78.7 years old; and the UMG-cohort comprises 12 individuals, clinically diagnosed with AD (based on molecular and/or imaging data), mean age of 73.2 years old. Unsupervised principal component analysis of FTIR spectra of serum-derived exosomes revealed higher discriminatory value for AD cases when compared to serum as a whole. Consistently, the partial least-squares analysis revealed that serum-derived exosomes present higher correlations than serum. In addition, the second derivative peak area calculation also revealed significant differences among Controls and AD cases. The results obtained suggest that this methodology can discriminate cases from Controls and thus be potential useful to assist in AD clinical diagnosis.
Collapse
Affiliation(s)
- Tânia Soares Martins
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Sandra Magalhães
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.,CICECO -Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Ilka Martins Rosa
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Jonathan Vogelgsang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, Goettingen, Germany.,Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Jens Wiltfang
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.,Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, Goettingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
| | | | - José Catita
- CEBIMED-Faculty of Health Sciences; University Fernando Pessoa, Porto, Portugal.,Paralab SA, Gondomar, Portugal
| | - Odete A B da Cruz E Silva
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.,The Discovery CTR, University of Aveiro Campus, Aveiro, Portugal
| | - Alexandra Nunes
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Ana Gabriela Henriques
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
42
|
Potter H, Woodcock JH, Boyd TD, Coughlan CM, O'Shaughnessy JR, Borges MT, Thaker AA, Raj BA, Adamszuk K, Scott D, Adame V, Anton P, Chial HJ, Gray H, Daniels J, Stocker ME, Sillau SH. Safety and efficacy of sargramostim (GM-CSF) in the treatment of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12158. [PMID: 33778150 PMCID: PMC7988877 DOI: 10.1002/trc2.12158] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Inflammatory markers have long been observed in the brain, cerebrospinal fluid (CSF), and plasma of Alzheimer's disease (AD) patients, suggesting that inflammation contributes to AD and might be a therapeutic target. However, non-steroidal anti-inflammatory drug trials in AD and mild cognitive impairment (MCI) failed to show benefit. Our previous work seeking to understand why people with the inflammatory disease rheumatoid arthritis are protected from AD found that short-term treatment of transgenic AD mice with the pro-inflammatory cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) led to an increase in activated microglia, a 50% reduction in amyloid load, an increase in synaptic area, and improvement in spatial memory to normal. These results called into question the consensus view that inflammation is solely detrimental in AD. Here, we tested our hypothesis that modulation of the innate immune system might similarly be used to treat AD in humans by investigating the ability of GM-CSF/sargramostim to safely ameliorate AD symptoms/pathology. METHODS A randomized, double-blind, placebo-controlled trial was conducted in mild-to-moderate AD participants (NCT01409915). Treatments (20 participants/group) occurred 5 days/week for 3 weeks plus two follow-up (FU) visits (FU1 at 45 days and FU2 at 90 days) with neurological, neuropsychological, blood biomarker, and imaging assessments. RESULTS Sargramostim treatment expectedly changed innate immune system markers, with no drug-related serious adverse events or amyloid-related imaging abnormalities. At end of treatment (EOT), the Mini-Mental State Examination score of the sargramostim group increased compared to baseline (P = .0074) and compared to placebo (P = .0370); the treatment effect persisted at FU1 (P = .0272). Plasma markers of amyloid beta (Aβ40 [decreased in AD]) increased 10% (P = .0105); plasma markers of neurodegeneration (total tau and UCH-L1) decreased 24% (P = .0174) and 42% (P = .0019), respectively, after sargramostim treatment compared to placebo. DISCUSSION The innate immune system is a viable target for therapeutic intervention in AD. An extended treatment trial testing the long-term safety and efficacy of GM-CSF/sargramostim in AD is warranted.
Collapse
Affiliation(s)
- Huntington Potter
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Jonathan H. Woodcock
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Timothy D. Boyd
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Christina M. Coughlan
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - John R. O'Shaughnessy
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Manuel T. Borges
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Department of RadiologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Ashesh A. Thaker
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Department of RadiologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | | | | | | | - Vanesa Adame
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Paige Anton
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Heidi J. Chial
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Helen Gray
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Joseph Daniels
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Michelle E. Stocker
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Stefan H. Sillau
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| |
Collapse
|
43
|
Ding X, Zhang S, Jiang L, Wang L, Li T, Lei P. Ultrasensitive assays for detection of plasma tau and phosphorylated tau 181 in Alzheimer's disease: a systematic review and meta-analysis. Transl Neurodegener 2021; 10:10. [PMID: 33712071 PMCID: PMC7953695 DOI: 10.1186/s40035-021-00234-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/24/2021] [Indexed: 02/08/2023] Open
Abstract
A lack of convenient and reliable biomarkers for diagnosis and prognosis is a common challenge for neurodegenerative diseases such as Alzheimer's disease (AD). Recent advancement in ultrasensitive protein assays has allowed the quantification of tau and phosphorylated tau proteins in peripheral plasma. Here we identified 66 eligible studies reporting quantification of plasma tau and phosphorylated tau 181 (ptau181) using four ultrasensitive methods. Meta-analysis of these studies confirmed that the AD patients had significantly higher plasma tau and ptau181 levels compared with controls, and that the plasma tau and ptau181 could predict AD with high-accuracy area under curve of the Receiver Operating Characteristic. Therefore, plasma tau and plasma ptau181 can be considered as biomarkers for AD diagnosis.
Collapse
Affiliation(s)
- Xulong Ding
- Department of Neurology and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuting Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lijun Jiang
- Mental Health Center and West China Brain Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lu Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Li
- Mental Health Center and West China Brain Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
44
|
Campese N, Palermo G, Del Gamba C, Beatino MF, Galgani A, Belli E, Del Prete E, Della Vecchia A, Vergallo A, Siciliano G, Ceravolo R, Hampel H, Baldacci F. Progress regarding the context-of-use of tau as biomarker of Alzheimer's disease and other neurodegenerative diseases. Expert Rev Proteomics 2021; 18:27-48. [PMID: 33545008 DOI: 10.1080/14789450.2021.1886929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Tau protein misfolding and accumulation in toxic species is a critical pathophysiological process of Alzheimer's disease (AD) and other neurodegenerative disorders (NDDs). Tau biomarkers, namely cerebrospinal fluid (CSF) total-tau (t-tau), 181-phosphorylated tau (p-tau), and tau-PET tracers, have been recently embedded in the diagnostic criteria for AD. Nevertheless, the role of tau as a diagnostic and prognostic biomarker for other NDDs remains controversial.Areas covered: We performed a systematical PubMed-based review of the most recent advances in tau-related biomarkers for NDDs. We focused on papers published from 2015 to 2020 assessing the diagnostic or prognostic value of each biomarker.Expert opinion: The assessment of tau biomarkers in alternative easily accessible matrices, through the development of ultrasensitive techniques, represents the most significant perspective for AD-biomarker research. In NDDs, novel tau isoforms (e.g. p-tau217) or proteolytic fragments (e.g. N-terminal fragments) may represent candidate diagnostic and prognostic biomarkers and may help monitoring disease progression. Protein misfolding amplification assays, allowing the identification of different tau strains (e.g. 3 R- vs. 4 R-tau) in CSF, may constitute a breakthrough for the in vivo stratification of NDDs. Tau-PET may help tracking the spatial-temporal evolution of tau pathophysiology in AD but its application outside the AD-spectrum deserves further studies.
Collapse
Affiliation(s)
- Nicole Campese
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giovanni Palermo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Claudia Del Gamba
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Alessandro Galgani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elisabetta Belli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eleonora Del Prete
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Andrea Vergallo
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Harald Hampel
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| |
Collapse
|
45
|
Tsai CL, Erickson KI, Sun HS, Kuo YM, Pai MC. A cross-sectional examination of a family history of Alzheimer's disease and ApoE epsilon 4 on physical fitness, molecular biomarkers, and neurocognitive performance. Physiol Behav 2020; 230:113268. [PMID: 33383402 DOI: 10.1016/j.physbeh.2020.113268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/30/2020] [Accepted: 11/21/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE The present study examined whether the ɛ4 allele of the apolipoprotein E (ApoE) gene impacts molecular biomarkers and neurocognitive performance among individuals at genetic risk for developing Alzheimer's disease (AD). The correlations between physical fitness and molecular/neurocognitive indices were also explored. METHODS Fasting blood samples were collected from 162 individuals with a family history of AD (ADFH). There were twenty-two carriers of the ApoE-4 variant (ApoE-4 group). For comparison purposes we randomly selected 22 non-ɛ4 carriers (non-ApoE-4 group) from the ADFH individuals. Circulating inflammatory cytokines (e.g., TNF-α, IL-1β, IL-6, IL-8, and IL-15), neuroprotective growth factors (e.g., BDNF, IGF-1, IGF-2, VEGF, and FGF-2), and Amyloid-β peptides (e.g., Aβ1-40 and Aβ1-42), neurocognitive performance [e.g., behavior and brain even-related potentials (ERP)] during a task-switching paradigm, as well as physical fitness scores were measured. RESULTS The ApoE-4 group relative to the non-ApoE-4 group was similar with respect to molecular biomarkers, physical fitness, and most measures of neurocognitive performance. However, ADFH individuals that were ɛ4 carriers exhibited significantly higher local switching accuracy costs, worse accuracy as well as smaller ERP P3 amplitudes for the memory-switching condition. Importantly, cardiorespiratory fitness levels were significantly correlated with accuracy for most task-switching conditions, and levels of BDNF, Aβ1-40, and Aβ1-42 collapsed across the two groups even when controlling for the age co-variable, while the ApoE-4 group revealed similar pattern of results. CONCLUSIONS These data suggest that individuals with ADFH that were carriers of the ApoE-4 variant performed worse on the task-switching paradigm and that this could be due to compromised task-set and memory updating processes. Physical exercise interventions aimed to enhance cardiorespiratory fitness levels could be a potential AD prevention strategy for ameliorating cognitive function and reducing the accumulation of the Aβ peptides in this high risk group.
Collapse
Affiliation(s)
- Chia-Liang Tsai
- Institute of Physical Education, Health and Leisure Studies, National Cheng Kung University, Taiwan.
| | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh, USA; Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Western Australia
| | - H-Sunny Sun
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Taiwan
| | - Yu-Min Kuo
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Taiwan
| | - Ming-Chyi Pai
- Division of Behavioral Neurology, Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Taiwan; Alzheimer's Disease Research Center, National Cheng Kung University Hospital, Taiwan.
| |
Collapse
|
46
|
Baldacci F, Lista S, Manca ML, Chiesa PA, Cavedo E, Lemercier P, Zetterberg H, Blennow K, Habert MO, Potier MC, Dubois B, Vergallo A, Hampel H. Age and sex impact plasma NFL and t-Tau trajectories in individuals with subjective memory complaints: a 3-year follow-up study. ALZHEIMERS RESEARCH & THERAPY 2020; 12:147. [PMID: 33183357 PMCID: PMC7663867 DOI: 10.1186/s13195-020-00704-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022]
Abstract
Background Plasma neurofilament light (NFL) and total Tau (t-Tau) proteins are candidate biomarkers for early stages of Alzheimer’s disease (AD). The impact of biological factors on their plasma concentrations in individuals with subjective memory complaints (SMC) has been poorly explored. We longitudinally investigate the effect of sex, age, APOE ε4 allele, comorbidities, brain amyloid-β (Aβ) burden, and cognitive scores on plasma NFL and t-Tau concentrations in cognitively healthy individuals with SMC, a condition associated with AD development. Methods Three hundred sixteen and 79 individuals, respectively, have baseline and three-time point assessments (at baseline, 1-year, and 3-year follow-up) of the two biomarkers. Plasma biomarkers were measured with an ultrasensitive assay in a mono-center cohort (INSIGHT-preAD study). Results We show an effect of age on plasma NFL, with women having a higher increase of plasma t-Tau concentrations compared to men, over time. The APOE ε4 allele does not affect the biomarker concentrations while plasma vitamin B12 deficiency is associated with higher plasma t-Tau concentrations. Both biomarkers are correlated and increase over time. Baseline NFL is related to the rate of Aβ deposition at 2-year follow-up in the left-posterior cingulate and the inferior parietal gyri. Baseline plasma NFL and the rate of change of plasma t-Tau are inversely associated with cognitive score. Conclusion We find that plasma NFL and t-Tau longitudinal trajectories are affected by age and female sex, respectively, in SMC individuals. Exploring the influence of biological variables on AD biomarkers is crucial for their clinical validation in blood.
Collapse
Affiliation(s)
- Filippo Baldacci
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, F-75013, Paris, France. .,Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56125, Pisa, Italy.
| | - Simone Lista
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, F-75013, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France.,Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Institute of Memory and Alzheimer's Disease (IM2A), F-75013, Paris, France
| | - Maria Laura Manca
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56125, Pisa, Italy.,Department of Mathematics, University of Pisa, Pisa, Italy
| | - Patrizia A Chiesa
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, F-75013, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France.,Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Institute of Memory and Alzheimer's Disease (IM2A), F-75013, Paris, France
| | - Enrica Cavedo
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, F-75013, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France.,Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Institute of Memory and Alzheimer's Disease (IM2A), F-75013, Paris, France.,Qynapse, Paris, France
| | - Pablo Lemercier
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, F-75013, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France.,Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Institute of Memory and Alzheimer's Disease (IM2A), F-75013, Paris, France
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Marie-Odile Habert
- Laboratoire d'Imagerie Biomédicale, Sorbonne University, CNRS, INSERM, F-75013, Paris, France.,Centre pour l'Acquisition et le Traitement des Images (www.cati-neuroimaging.com), Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Département de Médecine Nucléaire, F-75013, Paris, France
| | - Marie Claude Potier
- ICM Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, 47 Bd de l'Hôpital, F-75013, Paris, France
| | - Bruno Dubois
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, F-75013, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France.,Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Institute of Memory and Alzheimer's Disease (IM2A), F-75013, Paris, France
| | - Andrea Vergallo
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, F-75013, Paris, France
| | - Harald Hampel
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, F-75013, Paris, France
| | | | | |
Collapse
|
47
|
Fluid Candidate Biomarkers for Alzheimer's Disease: A Precision Medicine Approach. J Pers Med 2020; 10:jpm10040221. [PMID: 33187336 PMCID: PMC7712586 DOI: 10.3390/jpm10040221] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022] Open
Abstract
A plethora of dynamic pathophysiological mechanisms underpins highly heterogeneous phenotypes in the field of dementia, particularly in Alzheimer's disease (AD). In such a faceted scenario, a biomarker-guided approach, through the implementation of specific fluid biomarkers individually reflecting distinct molecular pathways in the brain, may help establish a proper clinical diagnosis, even in its preclinical stages. Recently, ultrasensitive assays may detect different neurodegenerative mechanisms in blood earlier. ß-amyloid (Aß) peptides, phosphorylated-tau (p-tau), and neurofilament light chain (NFL) measured in blood are gaining momentum as candidate biomarkers for AD. P-tau is currently the more convincing plasma biomarker for the diagnostic workup of AD. The clinical role of plasma Aβ peptides should be better elucidated with further studies that also compare the accuracy of the different ultrasensitive techniques. Blood NFL is promising as a proxy of neurodegeneration process tout court. Protein misfolding amplification assays can accurately detect α-synuclein in cerebrospinal fluid (CSF), thus representing advancement in the pathologic stratification of AD. In CSF, neurogranin and YKL-40 are further candidate biomarkers tracking synaptic disruption and neuroinflammation, which are additional key pathophysiological pathways related to AD genesis. Advanced statistical analysis using clinical scores and biomarker data to bring together individuals with AD from large heterogeneous cohorts into consistent clusters may promote the discovery of pathophysiological causes and detection of tailored treatments.
Collapse
|
48
|
Tsai CL, Pai MC. Circulating levels of Irisin in obese individuals at genetic risk for Alzheimer's disease: Correlations with amyloid-β, metabolic, and neurocognitive indices. Behav Brain Res 2020; 400:113013. [PMID: 33186636 DOI: 10.1016/j.bbr.2020.113013] [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: 07/28/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022]
Abstract
Irisin is involved in various metabolic pathways and is suggested to be a potential agent capable of preventing onset of Alzheimer's disease (AD) and ameliorating AD neuropathology and cognitive deficits. In the present study, the serum levels of Irisin and Amyloid-β (Aβ) peptides and the neurocognitive performance among obese individuals at genetic risk for AD were investigated. The correlations between Irisin and AD-related neuropathological and neurocognitive indices were also explored. Thirty-two individuals with a family history of AD (ADFH) and obesity (ADFH-obesity group) and 32 controls (ADFH-non-obesity group) were recruited. Circulating levels of Irisin, Aβ peptides, and metabolic biomarkers, as well as neurocognitive performance [e.g., behavior and brain even-related potentials (ERP)] were measured during a visuospatial working memory task. Although the ADFH-obesity group exhibited comparable reaction times, ERP N2 latency and amplitudes, and P3 latency as compared to the ADFH-non-obesity group when performing the cognitive task, they exhibited significantly lower rates of accuracy and smaller P3 amplitudes in the higher memory-load condition, even when controlling for the blood pressure and cardiorespiratory fitness co-variables. The serum levels of leptin, insulin, and glucose, and HOMA-IR were significantly higher in the ADFH-obesity group relative to the ADFH-non-obesity group, but this was not the case for the levels of Aβ1-40 and Aβ1-42. The Irisin levels approached between-group significance. Partial correlations adjusting for cardiorespiratory fitness and blood pressure showed that Irisin levels were positively associated with neurophysiological (i.e., P3 amplitude) performance in the ADFH-obesity group. The Irisin levels were not significantly correlated with the levels of Aβ1-40 and Aβ1-42. The present findings suggest that ADFH individuals with obesity exhibited neurocognitive deficits when performing the visuospatial working memory task, and serum Irisin levels could be one of the influencing factors. However, the relationship between the circulating levels of Irisin and Aβ peptides needs more evidence to support this assumption.
Collapse
Affiliation(s)
- Chia-Liang Tsai
- Institute of Physical Education, Health and Leisure Studies, National Cheng Kung University, Taiwan.
| | - Ming-Chyi Pai
- Division of Behavioral Neurology, Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Taiwan; Alzheimer's Disease Research Center, National Cheng Kung University Hospital, Taiwan
| |
Collapse
|
49
|
Yang SY, Liu HC, Chen WP. Immunomagnetic Reduction Detects Plasma Aβ 1-42 Levels as a Potential Dominant Indicator Predicting Cognitive Decline. Neurol Ther 2020; 9:435-442. [PMID: 33090326 PMCID: PMC7606390 DOI: 10.1007/s40120-020-00215-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 09/23/2020] [Indexed: 11/03/2022] Open
Abstract
Although the concentrations of Alzheimer’s disease (AD) biomarkers Aβ1–40, Aβ1–42 and tau protein are very low in human plasma, ultrasensitive assays such as immunomagnetic reduction (IMR) are able to precisely quantify them. Review articles have described the detailed working mechanism of IMR and revealed the feasibility of detecting early-stage AD by assaying these plasma biomarkers with IMR. In this review, we aimed to compare the significance of these plasma biomarkers in predicting cognitive decline in patients with Down syndrome, stroke, or amnestic mild cognitive impairment based on findings in the literature. We found that plasma Aβ1–42 might play the predominant role in predicting cognitive decline in these patients.
Collapse
Affiliation(s)
- Shieh-Yueh Yang
- MagQu Co., Ltd., New Taipei City, 231, Taiwan. .,MagQu LLC, Surprise, AZ, 85378, USA.
| | | | | |
Collapse
|
50
|
Tsai CL, Liang CS, Yang CP, Lee JT, Ho TH, Su MW, Lin GY, Lin YK, Chu HT, Hsu YW, Yang FC. Indicators of rapid cognitive decline in amnestic mild cognitive impairment: The role of plasma biomarkers using magnetically labeled immunoassays. J Psychiatr Res 2020; 129:66-72. [PMID: 32592947 DOI: 10.1016/j.jpsychires.2020.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/22/2020] [Accepted: 06/01/2020] [Indexed: 11/29/2022]
Abstract
Plasma levels of biomarkers change with the progression of Alzheimer's disease (AD), which involves the accumulation of pathological amyloid β (Aβ) and Tau protein tangles. However, few studies have investigated the association between plasma biomarkers and rapid cognitive decline in patients with amnestic mild cognitive impairment (aMCI) and AD. A total of 10 healthy controls, 24 patients with aMCI, and 19 patients with AD were enrolled. All participants underwent twice Mini-Mental State Examination (MMSE), with a mean 1.2 year interval. Immunomagnetic reduction was utilized to evaluate levels of plasma biomarkers, including amyloid β 1-40 (Aβ1-40), Aβ1-42, total Tau protein, phosphorylated Tau protein (Threonine 181), and α-synuclein (α-Syn). The correlations between plasma levels of biomarkers and MMSE change were examined. Our analysis reveals that current higher plasma levels of Aβ1-42 and α-Syn with the cut-off value of plasma Aβ1-42 >17.26 pg/mL and α-Syn >105 fg/mL had a moderate-to-high discriminatory capacity (area under the curve >0.70) for identifying cognitive deterioration in patients with aMCI. Our results thus suggest that plasma levels of Aβ1-42 and α-Syn may be considered as useful markers to assess the severity of global cognitive decline in patients with aMCI.
Collapse
Affiliation(s)
- Chia-Lin Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Sung Liang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Pai Yang
- Department of Neurology, Kuang Tien General Hospital, Taichung, Taiwan; Department of Nutrition, Huang-Kuang University, Taichung, Taiwan
| | - Jiunn-Tay Lee
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tsung-Han Ho
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Wei Su
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Guan-Yu Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Kai Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsuan-Te Chu
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Wei Hsu
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Fu-Chi Yang
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|