1
|
Wang H, Lv P. Safety and efficacy of rivastigmine and memantine combined for treatment of patients with Alzheimer's disease: a retrospective study. Am J Transl Res 2025; 17:2240-2249. [PMID: 40226022 PMCID: PMC11982832 DOI: 10.62347/yopp4162] [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: 03/01/2024] [Accepted: 12/31/2024] [Indexed: 04/15/2025]
Abstract
OBJECTIVE To assess the effects and safety of combining rivastigmine hydrogen tartrate capsules with memantine tablets for Alzheimer's disease (AD). METHODS A retrospective study was conducted on AD patients admitted to The Third People's Hospital of Yongkang from November 2021 to June 2023. There were two groups: a single drug group (n=21) given only memantine tablets, and a combination group (n=39) treated with both rivastigmine hydrogen tartrate capsules and memantine tablets. Data were collected, including age, gender, education, overall response rate, adverse reaction rate, mini-mental state examination (MMSE), activity of daily living (ADL), behavioral pathology in Alzheimer's disease scale (BEHAVE-AD), serum tumor necrosis factor-α (TNF-α), serum interleukin-6 (IL-6) and serum Tau at baseline and at week 12. RESULTS In both groups, compared to baseline, at week 12, MMSE increased, while ADL, BEHAVE-AD, serum TNF-α, IL-6, and Tau decreased (all P<0.05). After treatment, compared with the single drug group at week 12, the combination group had a higher MMSE (t=2.519, P=0.015), better effectiveness (χ2=4.331, P=0.037), and lower ADL (t=2.418, P=0.019), BEHAVE-AD (t=3.231, P=0.002), TNF-α (t=3.496, P=0.001), IL-6 (t=2.513, P=0.015) and Tau (t=2.290, P=0.026) levels. CONCLUSION The combination of the two drugs was more effective in alleviating AD symptoms with comparable safety. It also showed an edge in suppressing pro-inflammatory cytokines and Tau in AD.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Geriatric Psychiatry, The Third People's Hospital of Yongkang No. 18 Feifeng Road, Xicheng Street, Yongkang City, Jinhua 321300, Zhejiang, China
| | - Pingping Lv
- Department of Geriatric Psychiatry, The Third People's Hospital of Yongkang No. 18 Feifeng Road, Xicheng Street, Yongkang City, Jinhua 321300, Zhejiang, China
| |
Collapse
|
2
|
Walia A, Shew MA, Durakovic N, Herzog JA, Cirrito JR, Yuede CM, Wick CC, Manis M, Holtzman DM, Buchman CA, Rutherford MA. Alzheimer's Disease-Related Analytes Amyloid-β and Tau in Perilymph: Correlation With Patient Age and Cognitive Score. Otolaryngol Head Neck Surg 2024; 171:1850-1858. [PMID: 39189154 PMCID: PMC11606756 DOI: 10.1002/ohn.942] [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: 04/26/2024] [Revised: 07/16/2024] [Accepted: 07/27/2024] [Indexed: 08/28/2024]
Abstract
OBJECTIVE To describe the collection methods for perilymph fluid biopsy during cochlear implantation, detect levels of amyloid β 42 and 40 (Aβ42 and Aβ40), and total tau (tTau) analytes with a high-precision assay, to compare these levels with patient age and Montreal Cognitive Assessment (MoCA) scores, and explore potential mechanisms and relationships with otic pathology. STUDY DESIGN Prospective study. SETTING Tertiary referral center. METHODS Perilymph was collected from 25 patients using polyimide tubing to avoid amyloid adherence to glass, and analyzed with a single-molecule array advanced digital enzyme-linked immunosorbent assay platform for Aβ40, Aβ42, and tTau. Cognition was assessed by MoCA. RESULTS Perilymph volumes ranged from ∼1 to 13 µL, with analyte concentrations spanning 2.67 to 1088.26 pg/mL. All samples had detectable levels of tTau, Aβ40, and Aβ42, with a significant positive correlation between Aβ42 and Aβ40 levels. Levels of Aβ42, Aβ40, and tTau were positively correlated with age, while MoCA scores were inversely correlated with age. tTau and Aβ42/Aβ40-ratios were significantly correlated with MoCA scores. CONCLUSION Alzheimer's disease-associated peptides Aβ42, Aβ40, and tau analytes are detectable in human perilymph at levels approximately 10-fold lower than those found in cerebrospinal fluid (CSF). These species increase with age and correlate with cognitive impairment indicators, suggesting their potential utility as biomarkers for cognitive impairment in patients undergoing cochlear implantation. Future research should investigate the origin of these analytes in the perilymph and their potential links to inner ear pathologies and hearing loss, as well as their relationships to CSF and plasma levels in individuals.
Collapse
Affiliation(s)
- Amit Walia
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Matthew A. Shew
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Nedim Durakovic
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Jacques A. Herzog
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - John R. Cirrito
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Carla M. Yuede
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Cameron C. Wick
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Melissa Manis
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Craig A. Buchman
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Mark A. Rutherford
- Department of Otolaryngology—Head and Neck Surgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
3
|
Zhang S, Wang F, Xie L, Xu J, Song X, Tao J, Chen J, Ma D, Yu X, Shi X, Yang Y. Sodium-glucose cotransporter 2 inhibition through henagliflozin ameliorates cognitive impairment in patients with type 2 diabetes. J Diabetes Investig 2024; 15:1596-1603. [PMID: 39254788 PMCID: PMC11527823 DOI: 10.1111/jdi.14306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
AIMS/INTRODUCTION To assess whether the sodium-glucose cotransporter 2 inhibitor, henagliflozin, improves cognitive impairment in patients with type 2 diabetes. MATERIALS AND METHODS We carried out a prospective study on 290 patients with type 2 diabetes and cognitive impairment. Montreal Cognitive Assessment scores and plasma phosphorylated tau181 levels were used to assess cognition. The association between henagliflozin use and changes in cognition was examined using multivariable logistic regression analysis. RESULTS Montreal Cognitive Assessment scores at enrollment and after 6 months were 21 (interquartile range [IQR]19-23) versus 22 (IQR 20-25; P < 0.0001) in all patients, 21 (IQR 19-23) versus 24 (IQR 22-26; P < 0.0001) in the henagliflozin group and 21 (IQR 19-22) versus 21 (IQR 19-23; P > 0.05) in the non-sodium-glucose cotransporter 2 inhibitor group. Logistic regression analysis showed that henagliflozin treatment was associated with Montreal Cognitive Assessment score improvement independent of potential confounders (odds ratio [OR] 3.670, 95% confidence interval [CI] 2.224-6.056, P < 0.0001). Additionally, plasma phosphorylated tau181 levels significantly decreased at 6-month follow up in all patients (OR 11.5, 95% CI 9.9-13.7 vs OR 10.1, 95% CI 7.8-12.9, P < 0.0001) and in the henagliflozin group (OR 11.5, 95% CI 10.3-13.0 vs OR 9.2, 95% CI 7.1-10.7, P < 0.0001), but not in the non-sodium-glucose cotransporter 2 inhibitor group. Henagliflozin treatment was independently associated with decreased phosphorylated tau181 levels (OR 3.670, 95% CI 1.598-4.213, P < 0.0001). CONCLUSIONS Henagliflozin treatment was independently associated with improvements in Montreal Cognitive Assessment scores and plasma phosphorylated tau181 levels, indicating significant beneficial effects on cognitive impairment in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Endocrinology, Tongji Medical College, Tongji HospitalHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
- Branch of National Clinical Research Center for Metabolic DiseaseWuhanHubei ProvinceChina
| | - Fen Wang
- Department of Endocrinology, Tongji Medical College, Tongji HospitalHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
- Branch of National Clinical Research Center for Metabolic DiseaseWuhanHubei ProvinceChina
| | - Lei Xie
- Department of Endocrinology, Tongji Medical College, Tongji HospitalHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
- Branch of National Clinical Research Center for Metabolic DiseaseWuhanHubei ProvinceChina
| | - Jialu Xu
- Department of Endocrinology, Tongji Medical College, Tongji HospitalHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
- Branch of National Clinical Research Center for Metabolic DiseaseWuhanHubei ProvinceChina
| | - Xiaoqing Song
- Department of Endocrinology, Tongji Medical College, Tongji HospitalHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
- Branch of National Clinical Research Center for Metabolic DiseaseWuhanHubei ProvinceChina
| | - Jing Tao
- Department of Endocrinology, Tongji Medical College, Tongji HospitalHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
- Branch of National Clinical Research Center for Metabolic DiseaseWuhanHubei ProvinceChina
| | - Juan Chen
- Department of Neurosurgery, Tongji Medical College, Tongji HospitalHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - Delin Ma
- Department of Endocrinology, Tongji Medical College, Tongji HospitalHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
- Branch of National Clinical Research Center for Metabolic DiseaseWuhanHubei ProvinceChina
| | - Xuefeng Yu
- Department of Endocrinology, Tongji Medical College, Tongji HospitalHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
- Branch of National Clinical Research Center for Metabolic DiseaseWuhanHubei ProvinceChina
| | - Xiaoli Shi
- Department of Endocrinology, Tongji Medical College, Tongji HospitalHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
- Branch of National Clinical Research Center for Metabolic DiseaseWuhanHubei ProvinceChina
| | - Yan Yang
- Department of Endocrinology, Tongji Medical College, Tongji HospitalHuazhong University of Science and TechnologyWuhanHubei ProvinceChina
- Branch of National Clinical Research Center for Metabolic DiseaseWuhanHubei ProvinceChina
| |
Collapse
|
4
|
Faris A, Ibrahim IM, Hadni H, Elhallaoui M. High-throughput virtual screening of phenylpyrimidine derivatives as selective JAK3 antagonists using computational methods. J Biomol Struct Dyn 2024; 42:7574-7599. [PMID: 37539779 DOI: 10.1080/07391102.2023.2240413] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/19/2023] [Indexed: 08/05/2023]
Abstract
In this study, we used phenylpyrimidine derivatives with known biological activity against JAK3, a critical tyrosine kinase enzyme involved in signaling pathways, to find similar compounds as potential treatments for rheumatoid arthritis. These inhibitors inhibited JAK3 activity by forming a covalent bond with the Cys909 residue, which resulted in a strong inhibitory effect. Phenylpyrimidine is considered a promising therapeutic target. For pharmacophore modeling, 39 phenylpyrimidine derivatives with high pIC50 (Exp) values were chosen. The best pharmacophore model produced 28 molecules, and the five-point common pharmacophore hypothesis from P HASE (DHRRR_1) revealed the requirement for a hydrogen bond donor feature, a hydrophobic group feature, and three aromatic ring features for further design. The validation of the pharmacophore model phase was performed through 3D-QSAR using partial least squares (P LS). The 3D-QSAR study produced two successful models, an atom-based model (R2 = 0.95; Q2 = 0.67) and a field-based model (R2 = 0.93; Q2 = 0.76), which were used to predict the biological activity of new compounds. The pharmacophore model successfully distinguished between active and inactive medications, discovered potential JAK3 inhibitors, and demonstrated validity with a ROC of 0. 77. ADME-Tox was used to eliminate compounds that might have adverse effects. The best pharmacokinetics and affinity derivatives were selected for covalent docking. A molecular dynamics simulation of the selected molecules and the protein complex was performed to confirm the stability of the interaction with JAK3, whereas MM/GBSA simulations further confirmed their binding affinity. By using the principle of retrosynthesis, we were able to map out a pathway for synthesizing these potential drug candidates. This study has the potential to offer valuable and practical insights for optimizing novel derivatives of phenylpyrimidine.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abdelmoujoud Faris
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Ibrahim M Ibrahim
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Hanine Hadni
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Menana Elhallaoui
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
5
|
Karger AB, Nasrallah IM, Braffett BH, Luchsinger JA, Ryan CM, Bebu I, Arends V, Habes M, Gubitosi-Klug RA, Chaytor N, Biessels GJ, Jacobson AM, DCCT/EDIC Research Group. Plasma Biomarkers of Brain Injury and Their Association With Brain MRI and Cognition in Type 1 Diabetes. Diabetes Care 2024; 47:1530-1538. [PMID: 38861647 PMCID: PMC11362129 DOI: 10.2337/dc24-0229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/30/2024] [Indexed: 06/13/2024]
Abstract
OBJECTIVE To evaluate associations between plasma biomarkers of brain injury and MRI and cognitive measures in participants with type 1 diabetes (T1D) from the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) study. RESEARCH DESIGN AND METHODS Plasma amyloid-β-40, amyloid-β-42, neurofilament light chain (NfL), phosphorylated Tau-181 (pTau-181), and glial fibrillary acidic protein (GFAP) were measured in 373 adults who participated in the DCCT/EDIC study. MRI assessments included total brain and white matter hyperintensity volumes, white matter mean fractional anisotropy, and indices of Alzheimer disease (AD)-like atrophy and predicted brain age. Cognitive measures included memory and psychomotor and mental efficiency tests and assessments of cognitive impairment. RESULTS Participants were 60 (range 44-74) years old with 38 (30-51) years' T1D duration. Higher NfL was associated with an increase in predicted brain age (0.51 years per 20% increase in NfL; P < 0.001) and a 19.5% increase in the odds of impaired cognition (P < 0.01). Higher NfL and pTau-181 were associated with lower psychomotor and mental efficiency (P < 0.001) but not poorer memory. Amyloid-β measures were not associated with study measures. A 1% increase in mean HbA1c was associated with a 14.6% higher NfL and 12.8% higher pTau-181 (P < 0.0001). CONCLUSIONS In this aging T1D cohort, biomarkers of brain injury did not demonstrate an AD-like profile. NfL emerged as a biomarker of interest in T1D because of its association with higher HbA1c, accelerated brain aging on MRI, and cognitive dysfunction. Our study suggests that early neurodegeneration in adults with T1D is likely due to non-AD/nonamyloid mechanisms.
Collapse
Affiliation(s)
- Amy B. Karger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Ilya M. Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | | | | | | | - Ionut Bebu
- The Biostatistics Center, George Washington University, Rockville, MD
| | - Valerie Arends
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Mohamad Habes
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonino, TX
| | - Rose A. Gubitosi-Klug
- Case Western Reserve University, Rainbow Babies and Children’s Hospital, Cleveland, OH
| | - Naomi Chaytor
- Department of Community and Behavioral Health, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA
| | - Geert J. Biessels
- Department of Neurology, University Medical Center Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Alan M. Jacobson
- New York University Grossman Long Island School of Medicine, New York University Langone Hospital-Long Island, Mineola, NY
| | | |
Collapse
|
6
|
Jiang YQ, Wei YP, Liu XP, Chen JS, Mao CJ, Jin BK. Strong cathode electroluminescence biosensor based on CeO 2 functionalized PCN-222@Ag NPs for sensitive detection of p-Tau-181 protein. J Colloid Interface Sci 2024; 665:144-151. [PMID: 38520931 DOI: 10.1016/j.jcis.2024.03.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/03/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
Electrochemiluminescence (ECL) biosensors provide a convenient and high sensitivity method for early disease diagnosis. However, creating luminophore arrays relying on powerful ECL signals remains a daunting task. Porphyrin-centered metal organic frameworks (MOFs) exhibit remarkable potential in ECL sensing applications. In this paper, based on a simple one-pot synthesis method, PCN-222@Ag NPs doped with CeO2 was synthesized to enhance the ECL performance. Due to the strong catalytic ability of CeO2, the ECL signal strength of the new material PCN-222@CeO2@Ag NPs is much higher than that of the PCN-222@Ag NPs and PCN-222. The luminous properties of PCN-222@CeO2@Ag NPs become more intense and stable due to the excellent electronic conductivity of Ag NPs. Based on the fact that CuS@PDA composite can quench the ECL signal of PCN-222@CeO2@Ag NPs, we constructed a novel sandwich ECL immune sensor for the detection of phosphorylated Tau 181 (p-Tau-181) protein. The ECL sensor has a great linear relationship with p-Tau-181 protein concentration, ranging from 1 pg/mL to 100 ng/mL. The detection limit is as low as 0.147 pg/mL. This work provides new ideas for developing sensitive ECL sensors for the p-Tau-181 protein, the marker of Alzheimer's disease.
Collapse
Affiliation(s)
- Yun-Qi Jiang
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China
| | - Yu-Ping Wei
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China
| | - Xing-Pei Liu
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China
| | - Jing-Shuai Chen
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China
| | - Chang-Jie Mao
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China.
| | - Bao-Kang Jin
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials (Ministry of Education), School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China; Key Laboratory of Functional Inorganic Materials of Anhui Province, School of Chemistry & Chemical Engineering, Anhui University, Hefei 230601, PR China
| |
Collapse
|
7
|
Ming T, Lan T, Yu M, Cheng S, Duan X, Wang H, Deng J, Kong D, Yang S, Shen Z. Advancements in Biosensors for Point-of-Care Testing of Nucleic Acid. Crit Rev Anal Chem 2024:1-16. [PMID: 38889541 DOI: 10.1080/10408347.2024.2366943] [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: 06/20/2024]
Abstract
Rapid, low-cost and high-specific diagnosis based on nucleic acid detection is pivotal in both detecting and controlling various infectious diseases, effectively curbing their spread. Moreover, the analysis of circulating DNA in whole blood has emerged as a promising noninvasive strategy for cancer diagnosis and monitoring. Although traditional nucleic acid detection methods are reliable, their time-consuming and intricate processes restrict their application in rapid field assays. Consequently, an urgent emphasis on point-of-care testing (POCT) of nucleic acids has arisen. POCT enables timely and efficient detection of specific sequences, acting as a deterrent against infection sources and potential tumor threats. To address this imperative need, it is essential to consolidate key aspects and chart future directions in POCT biosensors development. This review aims to provide an exhaustive and meticulous analysis of recent advancements in POCT devices for nucleic acid diagnosis. It will comprehensively compare these devices across crucial dimensions, encompassing their integrated structures, the synthesized nanomaterials harnessed, and the sophisticated detection principles employed. By conducting a rigorous evaluation of the current research landscape, this review will not only spotlight achievements but also identify limitations, offering valuable insights into the future trajectory of nucleic acid POCT biosensors. Through this comprehensive analysis, the review aspires to serve as an indispensable guide for fostering the development of more potent biosensors, consequently fostering precise and efficient POCT applications for nucleic acids.
Collapse
Affiliation(s)
- Tao Ming
- Institute of Transplantation Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Tingting Lan
- Institute of Transplantation Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Mingxing Yu
- Institute of Transplantation Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Shuhan Cheng
- Institute of Transplantation Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Xu Duan
- Institute of Transplantation Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Hong Wang
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Juan Deng
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Deling Kong
- Institute of Transplantation Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Shuang Yang
- Institute of Transplantation Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Zhongyang Shen
- Institute of Transplantation Medicine, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
8
|
Kim S, Ma X, Jeon MJ, Song S, Lee JS, Lee JU, Lee CN, Choi SH, Sim SJ. Distinct plasma phosphorylated-tau proteins profiling for the differential diagnosis of mild cognitive impairment and Alzheimer's disease by plasmonic asymmetric nanobridge-based biosensor. Biosens Bioelectron 2024; 250:116085. [PMID: 38295582 DOI: 10.1016/j.bios.2024.116085] [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: 09/05/2023] [Revised: 01/22/2024] [Accepted: 01/27/2024] [Indexed: 02/02/2024]
Abstract
The differential diagnosis between mild cognitive impairment (MCI) and Alzheimer's disease (AD) has been highly demanded for its effectiveness in preventing and contributing to early diagnosis of AD. To this end, we developed a single plasmonic asymmetric nanobridge (PAN)-based biosensor to differentially diagnose MCI and AD by quantitative profiling of phosphorylated tau proteins (p-tau) in clinical plasma samples, which revealed a significant correlation with AD development and progression. The PAN was designed to have a conductive junction and asymmetric structure, which was unable to be synthesized by the traditional thermodynamical methods. For its unique morphological characteristics, PAN features high electromagnetic field enhancement, enabling the biosensor to achieve high sensitivity, with a limit of detection in the attomolar regime for quantitative analysis of p-tau. By introducing support vector machine (SVM)-based machine learning algorithm, the improved diagnostic system was achieved for prediction of healthy controls, MCI, and AD groups with an accuracy of 94.47 % by detecting various p-tau species levels in human plasma. Thus, our proposed PAN-based plasmonic biosensor has a powerful potential in clinical utility for predicting the onset of AD progression in the asymptomatic phase.
Collapse
Affiliation(s)
- Soohyun Kim
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Xingyi Ma
- School of Science, Harbin Institute of Technology, Shenzhen, Guangdong, 518055, China
| | - Myeong Jin Jeon
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sojin Song
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jeong Seop Lee
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jong Uk Lee
- Department of Chemical Engineering, Sunchon National University, Jeollanam-do, 57922, Republic of Korea.
| | - Chan-Nyoung Lee
- Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| | - Seong Hye Choi
- Department of Neurology, Inha University College of Medicine, Incheon, 22332, Republic of Korea.
| | - Sang Jun Sim
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
9
|
Patel H, Wisely CE, Robbins CB, Parker D, Challa P, Grewal DS, Fekrat S. Aqueous and Plasma Levels of Phosphorylated Tau 181 in Individuals with Normal Cognition. J Alzheimers Dis 2024; 100:239-245. [PMID: 38848189 DOI: 10.3233/jad-240279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Background Plasma and cerebrospinal fluid (CSF) levels of p-tau181 have been associated with Alzheimer's disease (AD). The retina and vitreous have shown measurable quantities of phosphorylated tau 181 (p-tau181). The aqueous humor, which can be collected during cataract surgery, may have measurable concentrations of p-tau181. Objective To determine whether p-tau181 is detectable in the aqueous humor and if so, whether it is associated with other measures that might be consistent with AD such as higher plasma p-tau181 concentration and lower Montreal Cognitive Assessment (MoCA-BLIND version 7.1) score. Methods Aqueous humor samples, blood samples, and MoCA-BLIND scores were collected from patients who did not carry a clinical diagnosis of cognitive impairment at the time of cataract surgery. Aqueous p-tau181 concentrations and plasma p-tau181 concentrations were then measured using ultra-sensitive single-molecule assay ELISA technology. A rank-transformed mixed-effects multivariate regression model was used to determine associations between aqueous concentrations, plasma concentrations, and MoCA-BLIND scores. Results 16 eyes of 16 participants were enrolled with an average age of 71.6. Average MoCA-BLIND score was 20.6/22, average aqueous p-tau181 concentration was 6.4 pg/mL, and average plasma p-tau181 concentration was 3.1 pg/mL. Higher plasma p-tau181 was significantly associated with higher aqueous p-tau181 (p = 0.02). Aqueous p-tau181 and plasma p-tau181 were negatively associated with MoCA-BLIND scores (p = 0.005 and p = 0.001 respectively) in these patients. Conclusions Aqueous p-tau181 is positively correlated with plasma p-tau181 and is negatively correlated with MoCA-BLIND scores. Further study in individuals with mild cognitive impairment or AD characterized by cerebrospinal fluid and volumetric MRI metrics may yield further insights.
Collapse
Affiliation(s)
- Hemal Patel
- iMIND Study Group, Duke University School of Medicine, Durham, NC, USA
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - C Ellis Wisely
- iMIND Study Group, Duke University School of Medicine, Durham, NC, USA
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Cason B Robbins
- iMIND Study Group, Duke University School of Medicine, Durham, NC, USA
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Daniel Parker
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Pratap Challa
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Dilraj S Grewal
- iMIND Study Group, Duke University School of Medicine, Durham, NC, USA
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Sharon Fekrat
- iMIND Study Group, Duke University School of Medicine, Durham, NC, USA
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
10
|
Zhao X, Li L, Ma X, Li Y, Gao B, Luo W. The role of immune and inflammatory-related indicators in cognitive dysfunction and disease severity in patients with parkinson's disease. J Neural Transm (Vienna) 2024; 131:13-24. [PMID: 37864052 DOI: 10.1007/s00702-023-02704-8] [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: 06/07/2023] [Accepted: 09/27/2023] [Indexed: 10/22/2023]
Abstract
We aimed to explore the role of immune and inflammatory indicators in cognitive dysfunction and disease severity in patients with Parkinson's disease (PD). A total of 123 patients with Parkinson's disease were enrolled in the PD group and 49 healthy volunteers in the control group. The patients with PD were further divided into 2 subgroups by evaluating cognitive function using the Montreal Cognitive Assessment (MoCA) and Mini-Mental State Examination (MMSE): the normal cognitive function (PD-NCI) group and the mild cognitive impairment (PD-MCI) group. Moreover, the PD patients were also divided into 2 subgroups using the defined scale of the Hoehn and Yahr (H-Y) stage: the early-stage group and the middle- and late-stage group. Immune and inflammatory indicators, including serum Aβ1-42, Tau, CD4+, CD8+, CD3+, B lymphocytes cell, NK cell, Th17 cell, Treg cell, IL-6, IL-17, and TNF-α levels, were evaluated and analyzed to explore the potential correlation with the cognitive dysfunction and disease severity of PD. Among the 123 PD patients, 60 (48.8%) were diagnosed with mild cognitive impairment. Aβ1-42, CD4+, CD8+, CD3+, and Treg levels observed in the PD-NCI group were lower than the control group (P < 0.001), while higher than the PD-MCI group (P < 0.001). The levels of Tau, Th17, IL-6, IL-17, and TNF-α observed in the PD-NCI group were higher than the control group (P < 0.001), while lower than in the PD-MCI group (P < 0.01). Using the same method, the results of the early-stage group and the middle- and the late-stage group were the same as above. Logistic regression analysis and ROC curve estimation were performed and indicated that the variation of Tau, CD8+, Treg, TNF-α levels was associated with cognitive decline in PD patients, and may serve as markers of PD onset. Furthermore, the variation of Aβ1-42, IL-6, and TNF-α levels was found to correlate with the disease severity of PD. The immune and inflammatory-related indicators may represent an important factor in the pathogenesis of PD, cognitive dysfunction, and disease severity. The variation of Tau protein, CD8+, Treg, and TNF-α levels are associated with the cognitive dysfunction of PD, which may be considered as onset markers. Moreover, the variation of Aβ1-42, IL-6, and TNF-α levels can predict the progression of PD.
Collapse
Affiliation(s)
- Xudong Zhao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu Province, China
- Department of General Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu Province, China
| | - Lei Li
- Department of General Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu Province, China
| | - Xiuping Ma
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu Province, China
| | - Yang Li
- Department of Neurology, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, 313000, Zhejiang Province, China
| | - Beibei Gao
- Department of General Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu Province, China
| | - Weifeng Luo
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu Province, China.
| |
Collapse
|
11
|
Singh AK, Anwar M, Pradhan R, Ashar MS, Rai N, Dey S. Surface plasmon resonance based-optical biosensor: Emerging diagnostic tool for early detection of diseases. JOURNAL OF BIOPHOTONICS 2023:e202200380. [PMID: 36883612 DOI: 10.1002/jbio.202200380] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/01/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
The development of diagnostic tools remains at the center of the health care system. In recent times optical biosensors have been widely applied in the scientific community, especially for monitoring protein-protein or nucleic acid hybridization interactions. Optical biosensors-derived surface plasmon resonance (SPR) technology has appeared as a revolutionary technology at the current times. This review focuses on the research work in molecular biomarker evaluation using the technique based on SPR for translational clinical diagnosis. The review has covered both communicable and noncommunicable diseases by using different bio-fluids of the patient's sample for diagnosis of the diseases. An increasing number of SPR approaches have been developed in healthcare research and fundamental biological studies. The utility of SPR in the area of biosensing basically lies in its noninvasive diagnostic and prognostic feature due to its label-free high sensitivity and specificity properties. This makes SPR an invaluable tool with precise application in the recognition of different stages of the disease.
Collapse
Affiliation(s)
- Abhinay Kumar Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Masroor Anwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Rashmita Pradhan
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Mohd Suhail Ashar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Nitish Rai
- Department of Biotechnology, Mohanlal Sukhadia University (MLSU), Udaipur, Rajasthan, India
| | - Sharmistha Dey
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
12
|
Abed SS, Hamdan FB, Hussein MM, Al-Mayah QS. Plasma tau and neurofilament light chain as biomarkers of Alzheimer's disease and their relation to cognitive functions. J Med Life 2023; 16:284-289. [PMID: 36937471 PMCID: PMC10015560 DOI: 10.25122/jml-2022-0251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/03/2023] [Indexed: 03/21/2023] Open
Abstract
Alzheimer's disease (AD) dementia is the most frequent cause of neurodegenerative dementia. The cognitive and behavioral symptoms associated with this disorder often have overlapping characteristics, potentially resulting in delayed diagnosis or misdiagnosis. This study aimed to assess the level of peripheral blood neurofilament light chain (NfL) and total tau (t-tau) protein in AD patients and investigate their relationship with cognitive impairment. The study included 80 participants of both sexes between the ages of 60 to 85 years. The participants were divided into two groups, consisting of 40 individuals in the control group (mean age 75±6.6 years) who had no cognitive or functional impairments and 40 AD patients (mean age 74.98±5.03 years). This study utilized the DSM-5 diagnostic criteria for major or mild neurocognitive disorder attributed to Alzheimer's disease (AD). The clinical and biochemical features of all participants were documented, and the Alzheimer's disease Assessment Scale cognitive subscale (ADAS-cog) scores were evaluated. Sandwich ELISA was employed to determine serum NfL and t-tau protein levels. The median serum NfL and t-tau protein levels in AD patients were significantly higher than those of the controls (47.84 pg/ml versus 17.66 pg/ml and 12.05 pg/ml versus 11.13 pg/ml, respectively). Age was positively correlated with NfL, t-tau levels, and ADAS-cog. Although elevated NfL and t-tau protein levels may play a role in disease progression, their diagnostic value for AD was limited.
Collapse
Affiliation(s)
- Sadiruldeen Sami Abed
- Department of Pharmacy, Osol Aldeen University College, Baghdad, Iraq
- Corresponding Author: Sadiruldeen Sami Abed, Department of Pharmacy, Osol Aldeen University College, Baghdad, Iraq. E-mail:
| | - Farqad Bader Hamdan
- Department of Physiology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | | | | |
Collapse
|
13
|
Su J, Deng Y, Cai B, Teng S, Zhang S, Liu Y, Lin J, Yang Q, Zeng D, Zhao X, Chen T. PI3K polymorphism in patients with sporadic Parkinson's disease. Medicine (Baltimore) 2022; 101:e32349. [PMID: 36595764 PMCID: PMC9794324 DOI: 10.1097/md.0000000000032349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Parkinson's disease (PD) is a common irreversible neurodegenerative disease associated with cognitive impairment. To investigate the serum level of phosphatidylinositol-3-kinase (PI3K) and the distribution of the genotypes and alleles of 3 PI3K single-nucleotide polymorphisms (RS37,30,087, RS37,30,088, and RS37,30,089) in PD patients with different clinical characteristics. A total of 54 PD patients and 50 healthy individuals were recruited. The serum PI3K level was measured using the enzyme-linked immunosorbent assay. The severity of PD was assessed using the modified Hoehn-Yahr scale. The cognitive function of PD patients was evaluated using the Mini-Mental State Examination scale and the Montreal Cognitive Assessment. The distribution of the alleles and genotypes of PI3K single-nucleotide polymorphisms (SNPs) was calculated using the Hardy-Weinberg equilibrium. PD patients showed a significantly higher serum level of PI3K compared to healthy individuals. Increased serum PI3K level was observed in PD patients with more severe disease, longer disease duration, and impaired cognitive function. Additionally, no significant differences were observed in the distributions of the genotypes and alleles of 3 PI3K SNPs between PD patients with normal cognitive function and those with cognitive impairment. PD patients with different levels of disease severity, disease duration, and cognitive function had significantly different serum levels of PI3K. However, the PI3K SNPs in patients with normal cognitive function were not significantly different from those in patients with cognitive impairment. These findings contribute to a better understanding of the roles of PI3K and SNPs of the PI3K gene in PD.
Collapse
Affiliation(s)
- Jiali Su
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yidong Deng
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Benchi Cai
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Si Teng
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Shan Zhang
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yanhui Liu
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jie Lin
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Qiang Yang
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Danting Zeng
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Xiuying Zhao
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Tao Chen
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
- * Correspondence: Tao Chen, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19 Xiuhua Road, Haikou, Hainan 570311, China (e-mail: )
| |
Collapse
|
14
|
Plasma P-Tau181 for the Discrimination of Alzheimer’s Disease from Other Primary Dementing and/or Movement Disorders. Biomolecules 2022; 12:biom12081099. [PMID: 36008993 PMCID: PMC9405977 DOI: 10.3390/biom12081099] [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: 02/22/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
Blood phospho-tau181 may offer a useful biomarker for Alzheimer’s disease. However, the use of either serum or plasma phospho-tau181 and their diagnostic value are currently under intense investigation. In a pilot study, we measured both serum and plasma phospho-tau181 (pT181-Tau) by single molecule array (Simoa) in a group of patients with Alzheimer’s disease and a mixed group of patients with other primary dementing and/or movement disorders. Classical cerebrospinal fluid biomarkers were also measured. Plasma (but not serum) pT181-Tau showed a significant increase in Alzheimer’s disease and correlated significantly with cerebrospinal fluid amyloid and pT181-Tau. Receiver operating curve analysis revealed a significant discrimination of Alzheimer’s from non-Alzheimer’s disease patients, with an area under the curve of 0.83 and an excellent sensitivity but a moderate specificity. Plasma pT181-Tau is not an established diagnostic biomarker for Alzheimer’s disease, but it could become one in the future, or it may serve as a screening tool for specific cases of patients or presymptomatic subjects.
Collapse
|
15
|
Blood phospho-tau in Alzheimer disease: analysis, interpretation, and clinical utility. Nat Rev Neurol 2022; 18:400-418. [PMID: 35585226 DOI: 10.1038/s41582-022-00665-2] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 12/11/2022]
Abstract
Well-authenticated biomarkers can provide critical insights into the biological basis of Alzheimer disease (AD) to enable timely and accurate diagnosis, estimate future burden and support therapeutic trials. Current cerebrospinal fluid and molecular neuroimaging biomarkers fulfil these criteria but lack the scalability and simplicity necessary for widespread application. Blood biomarkers of adequate effectiveness have the potential to act as first-line diagnostic and prognostic tools, and offer the possibility of extensive population screening and use that is not limited to specialized centres. Accelerated progress in our understanding of the biochemistry of brain-derived tau protein and advances in ultrasensitive technologies have enabled the development of AD-specific phosphorylated tau (p-tau) biomarkers in blood. In this Review we discuss how new information on the molecular processing of brain p-tau and secretion of specific fragments into biofluids is informing blood biomarker development, enabling the evaluation of preanalytical factors that affect quantification, and informing harmonized protocols for blood handling. We also review the performance of blood p-tau biomarkers in the context of AD and discuss their potential contexts of use for clinical and research purposes. Finally, we highlight outstanding ethical, clinical and analytical challenges, and outline the steps that need to be taken to standardize inter-laboratory and inter-assay measurements.
Collapse
|
16
|
Frontera JA, Boutajangout A, Masurkar AV, Betensky RA, Ge Y, Vedvyas A, Debure L, Moreira A, Lewis A, Huang J, Thawani S, Balcer L, Galetta S, Wisniewski T. Comparison of serum neurodegenerative biomarkers among hospitalized COVID-19 patients versus non-COVID subjects with normal cognition, mild cognitive impairment, or Alzheimer's dementia. Alzheimers Dement 2022; 18:899-910. [PMID: 35023610 PMCID: PMC9011610 DOI: 10.1002/alz.12556] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Neurological complications among hospitalized COVID-19 patients may be associated with elevated neurodegenerative biomarkers. METHODS Among hospitalized COVID-19 patients without a history of dementia (N = 251), we compared serum total tau (t-tau), phosphorylated tau-181 (p-tau181), glial fibrillary acidic protein (GFAP), neurofilament light chain (NfL), ubiquitin carboxy-terminal hydrolase L1 (UCHL1), and amyloid beta (Aβ40,42) between patients with or without encephalopathy, in-hospital death versus survival, and discharge home versus other dispositions. COVID-19 patient biomarker levels were also compared to non-COVID cognitively normal, mild cognitive impairment (MCI), and Alzheimer's disease (AD) dementia controls (N = 161). RESULTS Admission t-tau, p-tau181, GFAP, and NfL were significantly elevated in patients with encephalopathy and in those who died in-hospital, while t-tau, GFAP, and NfL were significantly lower in those discharged home. These markers correlated with severity of COVID illness. NfL, GFAP, and UCHL1 were higher in COVID patients than in non-COVID controls with MCI or AD. DISCUSSION Neurodegenerative biomarkers were elevated to levels observed in AD dementia and associated with encephalopathy and worse outcomes among hospitalized COVID-19 patients.
Collapse
Affiliation(s)
| | | | | | | | - Yulin Ge
- New York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Alok Vedvyas
- New York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Ludovic Debure
- New York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Andre Moreira
- New York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Ariane Lewis
- New York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Joshua Huang
- New York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Sujata Thawani
- New York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Laura Balcer
- New York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Steven Galetta
- New York University Grossman School of MedicineNew YorkNew YorkUSA
| | | |
Collapse
|
17
|
Alawode DOT, Fox NC, Zetterberg H, Heslegrave AJ. Alzheimer’s Disease Biomarkers Revisited From the Amyloid Cascade Hypothesis Standpoint. Front Neurosci 2022; 16:837390. [PMID: 35573283 PMCID: PMC9091905 DOI: 10.3389/fnins.2022.837390] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/04/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease worldwide. Amyloid beta (Aβ) is one of the proteins which aggregate in AD, and its key role in the disease pathogenesis is highlighted in the amyloid cascade hypothesis, which states that the deposition of Aβ in the brain parenchyma is a crucial initiating step in the future development of AD. The sensitivity of instruments used to measure proteins in blood and cerebrospinal fluid has significantly improved, such that Aβ can now successfully be measured in plasma. However, due to the peripheral production of Aβ, there is significant overlap between diagnostic groups. The presence of pathological Aβ within the AD brain has several effects on the cells and surrounding tissue. Therefore, there is a possibility that using markers of tissue responses to Aβ may reveal more information about Aβ pathology and pathogenesis than looking at plasma Aβ alone. In this manuscript, using the amyloid cascade hypothesis as a starting point, we will delve into how the effect of Aβ on the surrounding tissue can be monitored using biomarkers. In particular, we will consider whether glial fibrillary acidic protein, triggering receptor expressed on myeloid cells 2, phosphorylated tau, and neurofilament light chain could be used to phenotype and quantify the tissue response against Aβ pathology in AD.
Collapse
Affiliation(s)
- Deborah O. T. Alawode
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- *Correspondence: Deborah O. T. Alawode,
| | - Nick C. Fox
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Amanda J. Heslegrave
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Amanda J. Heslegrave,
| |
Collapse
|
18
|
Wang T, Zhang X, Wang Y, Liu W, Wang L, Hao L, Ju M, Xiao R. High cholesterol and 27-hydroxycholesterol contribute to phosphorylation of tau protein by impairing autophagy causing learning and memory impairment in C57BL/6J mice. J Nutr Biochem 2022; 106:109016. [DOI: 10.1016/j.jnutbio.2022.109016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 01/05/2022] [Accepted: 03/03/2022] [Indexed: 12/15/2022]
|
19
|
Nabizadeh F, Pourhamzeh M, Khani S, Rezaei A, Ranjbaran F, Deravi N. Plasma phosphorylated-tau181 levels reflect white matter microstructural changes across Alzheimer's disease progression. Metab Brain Dis 2022; 37:761-771. [PMID: 35015198 DOI: 10.1007/s11011-022-00908-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/06/2022] [Indexed: 01/25/2023]
Abstract
Alzheimer's Disease (AD) is characterized by cognitive impairments that hinder daily activities and lead to personal and behavioral problems. Plasma hyperphosphorylated tau protein at threonine 181 (p-tau181) has recently emerged as a new sensitive tool for the diagnosis of AD patients. We herein investigated the association of plasma P-tau181 and white matter (WM) microstructural changes in AD. We obtained data from a large prospective cohort of elderly individuals participating in the Alzheimer's Disease Neuroimaging Initiative (ADNI), which included baseline measurements of plasma P-tau181 and imaging findings. A subset of 41 patients with AD, 119 patients with mild cognitive impairments (MCI), and 43 healthy controls (HC) was included in the study, all of whom had baseline blood P-tau181 levels and had also undergone Diffusion Tensor Imaging. The analysis revealed that the plasma level of P-tau181 has a positive correlation with changes in Mean Diffusivity (MD), Radial Diffusivity (RD), and Axial Diffusivity (AxD), but a negative with Fractional Anisotropy (FA) parameters in WM regions of all participants. There is also a significant association between WM microstructural changes in different regions and P-tau181 plasma measurements within each MCI, HC, and AD group. In conclusion, our findings clarified that plasma P-tau181 levels are associated with changes in WM integrity in AD. P-tau181 could improve the accuracy of diagnostic procedures and support the application of blood-based biomarkers to diagnose WM neurodegeneration. Longitudinal clinical studies are also needed to demonstrate the efficacy of the P-tau181 biomarker and predict its role in structural changes.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Mahsa Pourhamzeh
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Saghar Khani
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ayda Rezaei
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Ranjbaran
- School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Tau proteins in blood as biomarkers of Alzheimer's disease and other proteinopathies. J Neural Transm (Vienna) 2022; 129:239-259. [PMID: 35175385 DOI: 10.1007/s00702-022-02471-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/02/2022] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), the most common age-dependent neurodegenerative disorder, is characterized neuropathologically by extracellular Aβ plaques and intracellular tau neurofibrillary tangles. While in AD tau pathology probably follows early alterations in Aβ metabolism, it develops independently in the so-called primary tauopathies, the main form being frontotemporal lobar degeneration with tau pathology. Tau pathology in AD brain is reflected in the cerebrospinal fluid (CSF) by elevated levels of the two AD tau biomarkers total and phosphorylated tau, which are now used for routine diagnostic purposes. On the contrary, no established neurochemical biomarkers exist for tau pathology in primary tauopathies. Thanks to recent technological advances, total and phosphorylated tau can now be quantified also on peripheral blood, and accumulating evidence shows that measurement of plasma phosphorylated tau species (P-tau181, P-tau217, and P-tau231) has high performances in discriminating AD patients from cognitively unimpaired subjects but also from patients with other dementias. Moreover, plasma P-tau levels are associated with tracer uptake on tau- and amyloid-PET as well as with brain atrophy, cognitive measures and longitudinal changes of these parameters. These features, together with the low invasiveness, scalability, and ease of longitudinal sampling, which differentiate plasma P-tau species from their CSF counterparts, make these proteins promising peripheral biomarkers for AD in both research and clinical setting. This review discusses the recent developments in the field of plasma tau proteins as diagnostic, pathophysiological and prognostic biomarkers of Alzheimer's disease; additional findings from the fields of genetic forms of AD and of non-AD proteinopathies are also summarized.
Collapse
|
21
|
Eduarda Schneider M, Guillade L, Correa-Duarte MA, Moreira FT. Development of a biosensor for Phosphorylated Tau 181 Protein detection in Early-Stage Alzheimer’s Disease. Bioelectrochemistry 2022; 145:108057. [DOI: 10.1016/j.bioelechem.2022.108057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/24/2022]
|
22
|
Altuna-Azkargorta M, Mendioroz-Iriarte M. Blood biomarkers in Alzheimer's disease. NEUROLOGÍA (ENGLISH EDITION) 2021; 36:704-710. [PMID: 34752348 DOI: 10.1016/j.nrleng.2018.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/01/2018] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Early diagnosis of Alzheimer disease (AD) through the use of biomarkers could assist in the implementation and monitoring of early therapeutic interventions, and has the potential to significantly modify the course of the disease. DEVELOPMENT The classic cerebrospinal fluid and approved structural and functional neuroimaging biomarkers are of limited clinical application given their invasive nature and/or high cost. The identification of more accessible and less costly biomarkers, such as blood biomarkers, would increase their use in clinical practice. We review the available published evidence on the main blood biochemical biomarkers potentially useful for diagnosing AD. CONCLUSIONS Blood biomarkers are more cost- and time-effective than CSF biomarkers. However, immediate applicability in clinical practice is relatively unlikely. The main limitations come from the difficulty of measuring and standardising thresholds between different laboratories and the failure to replicate results. Of all the molecules studied, apoptosis and neurodegeneration biomarkers and the biomarker panels obtained through "omics" approaches, such as isolated or combined metabolomics, offer the most promising results.
Collapse
Affiliation(s)
- M Altuna-Azkargorta
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain.
| | - M Mendioroz-Iriarte
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; Servicio de Neurología, Complejo Hospitalario de Navarra, Pamplona, Spain
| |
Collapse
|
23
|
Chen TB, Lin KJ, Lin SY, Lee YJ, Lin YC, Wang CY, Chen JP, Wang PN. Prediction of Cerebral Amyloid Pathology Based on Plasma Amyloid and Tau Related Markers. Front Neurol 2021; 12:619388. [PMID: 34671305 PMCID: PMC8520900 DOI: 10.3389/fneur.2021.619388] [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: 10/20/2020] [Accepted: 09/07/2021] [Indexed: 11/26/2022] Open
Abstract
Background and Purpose: Pyroglutamate-modified β-amyloid peptide (AβpE) is crucial for AD pathophysiological process. The potential associations of plasma AβpE and total tau (t-tau) with brain Aβ burden and cognitive performance remain to be clarified. Methods: Forty-six subjects with unimpaired cognition, mild cognitive impairment, or very mild dementia were enrolled. Plasma levels of AβpE3−40, t-tau, and Aβ42 were quantified by immunomagnetic reduction (IMR) assays. We analyzed individual and combined biomarker correlations with neuropsychological scores and Aβ positivity determined by 18F-florbetapir positron emission tomography (PET). Results: Both plasma AβpE3−40 levels and AβpE3−40/t-tau ratios correlated negatively with short-term memory and global cognition scores, while correlating positively with PET standardized uptake value ratios (SUVRs). Among the biomarkers analyzed, the combination of AβpE3−40 in a ratio with t-tau had the best discriminatory ability for Aβ PET positivity. Likewise, logistic regression analysis showed that AβpE3−40/t-tau was a highly robust predictor of Aβ PET positivity after controlling for relevant demographic covariates. Conclusion: Plasma AβpE3−40/t-tau ratios correlate with cognitive function and cerebral Aβ burden. The suitability of AβpE3−40/t-tau as a candidate clinical biomarker of AD pathology in the brain should be examined further in larger studies.
Collapse
Affiliation(s)
- Ting-Bin Chen
- Department of Neurology, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan.,Dementia Center, Taichung Veterans General Hospital, Taichung, Taiwan.,Center for Geriatrics and Gerontology, Taichung Veterans General Hospital, Taichung, 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
| | - Szu-Ying Lin
- Taipei Municipal Gan-Dau Hospital, Taipei, Taiwan
| | - Yi-Jung Lee
- Division of Neurology, Department of Medicine, Taipei City Hospital Renai Branch, Taipei, Taiwan
| | - Yi-Cheng Lin
- Taipei Municipal Gan-Dau Hospital, Taipei, Taiwan.,School of Life Sciences, Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
| | - Chen-Yu Wang
- Division of General Neurology, Department of Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jun-Peng Chen
- Biostatistics Task Force of Taichung Veterans General Hospital, Taichung, Taiwan
| | - Pei-Ning Wang
- Division of General Neurology, Department of Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
24
|
Yuan A, Nixon RA. Neurofilament Proteins as Biomarkers to Monitor Neurological Diseases and the Efficacy of Therapies. Front Neurosci 2021; 15:689938. [PMID: 34646114 PMCID: PMC8503617 DOI: 10.3389/fnins.2021.689938] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/02/2021] [Indexed: 01/01/2023] Open
Abstract
Biomarkers of neurodegeneration and neuronal injury have the potential to improve diagnostic accuracy, disease monitoring, prognosis, and measure treatment efficacy. Neurofilament proteins (NfPs) are well suited as biomarkers in these contexts because they are major neuron-specific components that maintain structural integrity and are sensitive to neurodegeneration and neuronal injury across a wide range of neurologic diseases. Low levels of NfPs are constantly released from neurons into the extracellular space and ultimately reach the cerebrospinal fluid (CSF) and blood under physiological conditions throughout normal brain development, maturation, and aging. NfP levels in CSF and blood rise above normal in response to neuronal injury and neurodegeneration independently of cause. NfPs in CSF measured by lumbar puncture are about 40-fold more concentrated than in blood in healthy individuals. New ultra-sensitive methods now allow minimally invasive measurement of these low levels of NfPs in serum or plasma to track disease onset and progression in neurological disorders or nervous system injury and assess responses to therapeutic interventions. Any of the five Nf subunits - neurofilament light chain (NfL), neurofilament medium chain (NfM), neurofilament heavy chain (NfH), alpha-internexin (INA) and peripherin (PRPH) may be altered in a given neuropathological condition. In familial and sporadic Alzheimer's disease (AD), plasma NfL levels may rise as early as 22 years before clinical onset in familial AD and 10 years before sporadic AD. The major determinants of elevated levels of NfPs and degradation fragments in CSF and blood are the magnitude of damaged or degenerating axons of fiber tracks, the affected axon caliber sizes and the rate of release of NfP and fragments at different stages of a given neurological disease or condition directly or indirectly affecting central nervous system (CNS) and/or peripheral nervous system (PNS). NfPs are rapidly emerging as transformative blood biomarkers in neurology providing novel insights into a wide range of neurological diseases and advancing clinical trials. Here we summarize the current understanding of intracellular NfP physiology, pathophysiology and extracellular kinetics of NfPs in biofluids and review the value and limitations of NfPs and degradation fragments as biomarkers of neurodegeneration and neuronal injury.
Collapse
Affiliation(s)
- Aidong Yuan
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, NYU Neuroscience Institute, New York, NY, United States
| | - Ralph A. Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, NYU Neuroscience Institute, New York, NY, United States
- Department of Cell Biology, New York University Grossman School of Medicine, (NYU), Neuroscience Institute, New York, NY, United States
| |
Collapse
|
25
|
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
|
26
|
Alawode DOT, Heslegrave AJ, Ashton NJ, Karikari TK, Simrén J, Montoliu‐Gaya L, Pannee J, O´Connor A, Weston PSJ, Lantero‐Rodriguez J, Keshavan A, Snellman A, Gobom J, Paterson RW, Schott JM, Blennow K, Fox NC, Zetterberg H. Transitioning from cerebrospinal fluid to blood tests to facilitate diagnosis and disease monitoring in Alzheimer's disease. J Intern Med 2021; 290:583-601. [PMID: 34021943 PMCID: PMC8416781 DOI: 10.1111/joim.13332] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/18/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is increasingly prevalent worldwide, and disease-modifying treatments may soon be at hand; hence, now, more than ever, there is a need to develop techniques that allow earlier and more secure diagnosis. Current biomarker-based guidelines for AD diagnosis, which have replaced the historical symptom-based guidelines, rely heavily on neuroimaging and cerebrospinal fluid (CSF) sampling. While these have greatly improved the diagnostic accuracy of AD pathophysiology, they are less practical for application in primary care, population-based and epidemiological settings, or where resources are limited. In contrast, blood is a more accessible and cost-effective source of biomarkers in AD. In this review paper, using the recently proposed amyloid, tau and neurodegeneration [AT(N)] criteria as a framework towards a biological definition of AD, we discuss recent advances in biofluid-based biomarkers, with a particular emphasis on those with potential to be translated into blood-based biomarkers. We provide an overview of the research conducted both in CSF and in blood to draw conclusions on biomarkers that show promise. Given the evidence collated in this review, plasma neurofilament light chain (N) and phosphorylated tau (p-tau; T) show particular potential for translation into clinical practice. However, p-tau requires more comparisons to be conducted between its various epitopes before conclusions can be made as to which one most robustly differentiates AD from non-AD dementias. Plasma amyloid beta (A) would prove invaluable as an early screening modality, but it requires very precise tests and robust pre-analytical protocols.
Collapse
Affiliation(s)
- D. O. T. Alawode
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - A. J. Heslegrave
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - N. J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Department of Old Age PsychiatryInstitute of Psychiatry, Psychology & NeuroscienceKing’s College LondonLondonUK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS FoundationLondonUK
| | - T. K. Karikari
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Simrén
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - L. Montoliu‐Gaya
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Pannee
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - A. O´Connor
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - P. S. J. Weston
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. Lantero‐Rodriguez
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - A. Keshavan
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - A. Snellman
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Turku PET CentreUniversity of TurkuTurkuFinland
| | - J. Gobom
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - R. W. Paterson
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. M. Schott
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - K. Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - N. C. Fox
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - H. Zetterberg
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| |
Collapse
|
27
|
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
|
28
|
Mielke MM. Consideration of Sex Differences in the Measurement and Interpretation of Alzheimer Disease-Related Biofluid-Based Biomarkers. J Appl Lab Med 2021; 5:158-169. [PMID: 31811073 DOI: 10.1373/jalm.2019.030023] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/23/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND The development of cerebrospinal fluid and blood-based biomarkers for Alzheimer disease (AD) and related disorders is rapidly progressing. Such biomarkers may be used clinically to screen the population, to enhance diagnosis, or to help determine prognosis. Although the use of precision medicine methods has contributed to enhanced understanding of the AD pathophysiological changes and development of assays, one aspect not commonly considered is sex differences. CONTENT There are several ways in which sex can affect the concentration or interpretation of biofluid biomarkers. For some markers, concentrations will vary by sex. For others, the concentrations might not vary by sex, but the impact or interpretation may vary by sex depending on the context of use (e.g., diagnostic vs prognostic). Finally, for others, there will be no sex differences in concentrations or their interpretation. This review will first provide a basis for sex differences, including differences in brain structure and function, and the means by which these differences could contribute to sex differences in biomarker concentrations. Next, the current state of sex differences in AD-related biofluid markers (i.e., amyloid-β, phosphorylated τ, total τ, neurofilament light chain, and neurogranin) will be reviewed. Lastly, factors that can lead to the misinterpretation of observed sex differences in biomarkers (either providing evidence for or against) will be considered. SUMMARY This review is intended to provide an impetus to consider sex differences in the measurement and interpretation of AD-related biofluid-based biomarkers.
Collapse
Affiliation(s)
- Michelle M Mielke
- Departments of Health Sciences Research and Neurology, Mayo Clinic, Rochester, MN
| |
Collapse
|
29
|
Cano A, Turowski P, Ettcheto M, Duskey JT, Tosi G, Sánchez-López E, García ML, Camins A, Souto EB, Ruiz A, Marquié M, Boada M. Nanomedicine-based technologies and novel biomarkers for the diagnosis and treatment of Alzheimer's disease: from current to future challenges. J Nanobiotechnology 2021; 19:122. [PMID: 33926475 PMCID: PMC8086346 DOI: 10.1186/s12951-021-00864-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/17/2021] [Indexed: 02/07/2023] Open
Abstract
Increasing life expectancy has led to an aging population, which has consequently increased the prevalence of dementia. Alzheimer's disease (AD), the most common form of dementia worldwide, is estimated to make up 50-80% of all cases. AD cases are expected to reach 131 million by 2050, and this increasing prevalence will critically burden economies and health systems in the next decades. There is currently no treatment that can stop or reverse disease progression. In addition, the late diagnosis of AD constitutes a major obstacle to effective disease management. Therefore, improved diagnostic tools and new treatments for AD are urgently needed. In this review, we investigate and describe both well-established and recently discovered AD biomarkers that could potentially be used to detect AD at early stages and allow the monitoring of disease progression. Proteins such as NfL, MMPs, p-tau217, YKL-40, SNAP-25, VCAM-1, and Ng / BACE are some of the most promising biomarkers because of their successful use as diagnostic tools. In addition, we explore the most recent molecular strategies for an AD therapeutic approach and nanomedicine-based technologies, used to both target drugs to the brain and serve as devices for tracking disease progression diagnostic biomarkers. State-of-the-art nanoparticles, such as polymeric, lipid, and metal-based, are being widely investigated for their potential to improve the effectiveness of both conventional drugs and novel compounds for treating AD. The most recent studies on these nanodevices are deeply explained and discussed in this review.
Collapse
Affiliation(s)
- Amanda Cano
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain.
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain.
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain.
| | - Patric Turowski
- UCL Institute of Ophthalmology, University College of London, London, UK
| | - Miren Ettcheto
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Jason Thomas Duskey
- Nanotech Lab, Te.Far.T.I, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Umberto Veronesi Foundation, 20121, Milano, Italy
| | - Giovanni Tosi
- Nanotech Lab, Te.Far.T.I, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Maria Luisa García
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Antonio Camins
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Agustín Ruiz
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Marta Marquié
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
30
|
Shin MK, Vázquez-Rosa E, Koh Y, Dhar M, Chaubey K, Cintrón-Pérez CJ, Barker S, Miller E, Franke K, Noterman MF, Seth D, Allen RS, Motz CT, Rao SR, Skelton LA, Pardue MT, Fliesler SJ, Wang C, Tracy TE, Gan L, Liebl DJ, Savarraj JPJ, Torres GL, Ahnstedt H, McCullough LD, Kitagawa RS, Choi HA, Zhang P, Hou Y, Chiang CW, Li L, Ortiz F, Kilgore JA, Williams NS, Whitehair VC, Gefen T, Flanagan ME, Stamler JS, Jain MK, Kraus A, Cheng F, Reynolds JD, Pieper AA. Reducing acetylated tau is neuroprotective in brain injury. Cell 2021; 184:2715-2732.e23. [PMID: 33852912 PMCID: PMC8491234 DOI: 10.1016/j.cell.2021.03.032] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/21/2021] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
Traumatic brain injury (TBI) is the largest non-genetic, non-aging related risk factor for Alzheimer's disease (AD). We report here that TBI induces tau acetylation (ac-tau) at sites acetylated also in human AD brain. This is mediated by S-nitrosylated-GAPDH, which simultaneously inactivates Sirtuin1 deacetylase and activates p300/CBP acetyltransferase, increasing neuronal ac-tau. Subsequent tau mislocalization causes neurodegeneration and neurobehavioral impairment, and ac-tau accumulates in the blood. Blocking GAPDH S-nitrosylation, inhibiting p300/CBP, or stimulating Sirtuin1 all protect mice from neurodegeneration, neurobehavioral impairment, and blood and brain accumulation of ac-tau after TBI. Ac-tau is thus a therapeutic target and potential blood biomarker of TBI that may represent pathologic convergence between TBI and AD. Increased ac-tau in human AD brain is further augmented in AD patients with history of TBI, and patients receiving the p300/CBP inhibitors salsalate or diflunisal exhibit decreased incidence of AD and clinically diagnosed TBI.
Collapse
Affiliation(s)
- Min-Kyoo Shin
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Edwin Vázquez-Rosa
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Yeojung Koh
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Matasha Dhar
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Kalyani Chaubey
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Coral J Cintrón-Pérez
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sarah Barker
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Emiko Miller
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Kathryn Franke
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Maria F Noterman
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Divya Seth
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, US
| | - Cara T Motz
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, US
| | - Sriganesh Ramachandra Rao
- Departments of Ophthalmology and Biochemistry, and the Neuroscience Graduate Program, SUNY-University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
| | - Lara A Skelton
- Departments of Ophthalmology and Biochemistry, and the Neuroscience Graduate Program, SUNY-University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, US
| | - Steven J Fliesler
- Departments of Ophthalmology and Biochemistry, and the Neuroscience Graduate Program, SUNY-University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
| | - Chao Wang
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Daniel J Liebl
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jude P J Savarraj
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Glenda L Torres
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hilda Ahnstedt
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ryan S Kitagawa
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - H Alex Choi
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Pengyue Zhang
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chien-Wei Chiang
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Francisco Ortiz
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jessica A Kilgore
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Noelle S Williams
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Victoria C Whitehair
- MetroHealth Rehabilitation Institute, The MetroHealth System, Cleveland, OH; Department of Physical Medicine and Rehabilitation, Case Western Reserve University, School of Medicine, Cleveland, OH USA
| | - Tamar Gefen
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Jonathan S Stamler
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Mukesh K Jain
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Allison Kraus
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - James D Reynolds
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Departments of Anesthesiology & Perioperative Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Andrew A Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Weill Cornell Autism Research Program, Weill Cornell Medicine of Cornell University, New York, NY, USA; Department of Neuroscience, Case Western Reserve University, School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
31
|
Chong JR, Ashton NJ, Karikari TK, Tanaka T, Saridin FN, Reilhac A, Robins EG, Nai YH, Vrooman H, Hilal S, Zetterberg H, Blennow K, Lai MKP, Chen CP. Plasma P-tau181 to Aβ42 ratio is associated with brain amyloid burden and hippocampal atrophy in an Asian cohort of Alzheimer's disease patients with concomitant cerebrovascular disease. Alzheimers Dement 2021; 17:1649-1662. [PMID: 33792168 DOI: 10.1002/alz.12332] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/01/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022]
Abstract
INTRODUCTION There is increasing evidence that phosphorylated tau (P-tau181) is a specific biomarker for Alzheimer's disease (AD) pathology, but its potential utility in non-White patient cohorts and patients with concomitant cerebrovascular disease (CeVD) is unknown. METHODS Single molecule array (Simoa) measurements of plasma P-tau181, total tau, amyloid beta (Aβ)40 and Aβ42, as well as derived ratios were correlated with neuroimaging modalities indicating brain amyloid (Aβ+), hippocampal atrophy, and CeVD in a Singapore-based cohort of non-cognitively impaired (NCI; n = 43), cognitively impaired no dementia (CIND; n = 91), AD (n = 44), and vascular dementia (VaD; n = 22) subjects. RESULTS P-tau181/Aβ42 ratio showed the highest area under the curve (AUC) for Aβ+ (AUC = 0.889) and for discriminating between AD Aβ+ and VaD Aβ- subjects (AUC = 0.903). In addition, P-tau181/Aβ42 ratio was associated with hippocampal atrophy. None of the biomarkers was associated with CeVD. DISCUSSION Plasma P-tau181/Aβ42 ratio may be a noninvasive means of identifying AD with elevated brain amyloid in populations with concomitant CeVD.
Collapse
Affiliation(s)
- Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Psychology and Neuroscience, King's College London, Institute of Psychiatry, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK.,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Tomotaka Tanaka
- Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore.,Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Francis N Saridin
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
| | - Anthonin Reilhac
- Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Edward G Robins
- Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Technology and Research, Biopolis, Singapore Bioimaging Consortium, A*Star Agency for Science, Singapore
| | - Ying-Hwey Nai
- Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Henri Vrooman
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Saima Hilal
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Kent Ridge, Singapore
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute at UCL, London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory, Aging and Cognition Centre, National University Health Systems, Kent Ridge, Singapore
| |
Collapse
|
32
|
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: 31] [Impact Index Per Article: 7.8] [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
|
33
|
Nangare S, Patil P. Nanoarchitectured Bioconjugates and Bioreceptors Mediated Surface Plasmon Resonance Biosensor for In Vitro Diagnosis of Alzheimer’s Disease: Development and Future Prospects. Crit Rev Anal Chem 2021; 52:1139-1169. [DOI: 10.1080/10408347.2020.1864716] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Sopan Nangare
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Pravin Patil
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| |
Collapse
|
34
|
Wilczyńska K, Waszkiewicz N. Diagnostic Utility of Selected Serum Dementia Biomarkers: Amyloid β-40, Amyloid β-42, Tau Protein, and YKL-40: A Review. J Clin Med 2020; 9:jcm9113452. [PMID: 33121040 PMCID: PMC7692800 DOI: 10.3390/jcm9113452] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/16/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022] Open
Abstract
Introduction: Dementia is a group of disorders that causes dysfunctions in human cognitive and operating functions. Currently, it is not possible to conduct a fast, low-invasive dementia diagnostic process with the use of peripheral blood biomarkers, however, there is a great deal of research in progress covering this subject. Research on dementia biomarkers in serum validates anticipated health and economic benefits from early screening tests. Biomarkers are also essential for improving the process of developing new drugs. Methods: The result analysis, of current studies on selected biomarker concentrations (Aβ40, Aβ42, t-tau, and YKL-40) and their combination in the serum of patients with dementia and mild cognitive disorders, involved a search for papers available in Medline, PubMed, and Web of Science databases published from 2000 to 2020. Results: The results of conducted cross-sectional studies comparing Aβ40, Aβ42, and Aβ42/Aβ40 among people with cognitive disorders and a control group are incoherent. Most of the analyzed papers showed an increase in t-tau concentration in diagnosed Alzheimer’s disease (AD) patients’ serum, whereas results of mild cognitive impairment (MCI) groups did not differ from the control groups. In several papers on the concentration of YKL-40 and t-tau/Aβ42 ratio, the results were promising. To date, several studies have only covered the field of biomarker concentrations in dementia disorders other than AD. Conclusions: Insufficient amyloid marker test repeatability may result either from imperfection of the used laboratorial techniques or inadequate selection of control groups with their comorbidities. On the basis of current knowledge, t-tau, t-tau/Aβ42, and YKL-40 seem to be promising candidates as biomarkers of cognitive disorders in serum. YKL-40 seems to be a more useful biomarker in early MCI diagnostics, whereas t-tau can be used as a marker of progress of prodromal states in mild AD. Due to the insignificant number of studies conducted to date among patients with dementia disorders other than AD, it is not possible to make a sound assessment of their usefulness in dementia differential diagnostics.
Collapse
|
35
|
Serum FOXO3A: A ray of hope for early diagnosis of Alzheimer's disease. Mech Ageing Dev 2020; 190:111290. [PMID: 32603667 DOI: 10.1016/j.mad.2020.111290] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 01/07/2023]
Abstract
Diagnosis of Alzheimer's disease (AD) is often difficult because of distinct and subjective clinical features, especially in the early stage. FOXO3a protein present in the cognitive centre of brain in inferior temporal region and parahippocampus. FOXO3a can be a potential novel target against AD. AD, Mild Cognitive impairment (MCI) and Geriatric Control (GC) were recruited after diagnosis by clinical assessment, MRI, TauPET and FDG-PET. We have quantified serum FOXO3a by surface plasmon resonance (SPR) and compare with TauPET between of AD, MCI patients and GC. Serum FOXO3A was significantly lower in AD (1.42 ± 0.09 ng/μl) compare to MCI (1.61 ± 0.14 ng/μl) and GC (1.89 ± 0.07 ng/μl). However, the Tau was higher in AD both in serum and also in PET scan. Serum pTau was significantly over-expressed in AD (0.176 ± 0.03 ng/μl), compare to other groups; MCI (0.16 ± 0.014 ng/μl) and GC (0.15 ± 0.024 ng/μl). Serum FOXO3A could significantly differentiate AD vs MCI, MCI vs GC and AD vs GC. However, Tau protein could only differentiate AD vs GC but not MCI vs GC. Serum FOXO3A may serve as novel blood marker for early detection for AD and target for therapeutic intervention.
Collapse
|
36
|
Chen TB, Lai YH, Ke TL, Chen JP, Lee YJ, Lin SY, Lin PC, Wang PN, Cheng IH. Changes in Plasma Amyloid and Tau in a Longitudinal Study of Normal Aging, Mild Cognitive Impairment, and Alzheimer's Disease. Dement Geriatr Cogn Disord 2020; 48:180-195. [PMID: 31991443 DOI: 10.1159/000505435] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/15/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Changes in cerebrospinal fluid, neuroimaging, and cognitive functions have been used as diagnostic biomarkers of Alzheimer's disease (AD). This study aimed to investigate the temporal trajectories of plasma biomarkers in subjects with mild cognitive impairment (MCI) and patients with AD relative to healthy controls (HCs). METHODS In this longitudinal study, 82 participants (31 HCs, 33 MCI patients, and 18 AD patients) were enrolled. After 3 years, 7 HCs had transitioned to MCI and 10 subjects with MCI had converted to AD. We analyzed plasma amyloid beta (Aβ) and tau proteins at baseline and annually to correlate with biochemical data and neuropsychological scores. RESULTS Longitudinal data analysis showed an evolution of Aβ-related biomarkers over time within patients, whereas tau-related biomarkers differed primarily across diagnostic classifications. An initial steady increase in Aβ42 in the MCI stage was followed by a decrease just prior to clinical AD onset. Hyperphosphorylated tau protein levels correlated with cognitive decline in the MCI stage, but not in the AD stage. CONCLUSION Plasma Aβ and tau levels change in a dynamic, nonlinear, nonparallel manner over the AD continuum. Changes in plasma Aβ concentration are time-dependent, whereas changes in hyperphosphorylated tau protein levels paralleled the clinical progression of MCI. It remains to be clarified whether diagnostic efficiency can be improved by combining multiple plasma markers or combining plasma markers with other diagnostic biomarkers.
Collapse
Affiliation(s)
- Ting-Bin Chen
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan.,Department of Neurology, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan.,Dementia and Parkinson's Disease Integrated Center, Taichung Veterans General Hospital, Taichung, Taiwan.,Center for Geriatrics and Gerontology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yu-Hua Lai
- Department of Neurology, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Ting-Ling Ke
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Jun-Peng Chen
- Biostatistics Task Force of Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Jung Lee
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan.,Division of Neurology, Department of Medicine, Taipei City Hospital Renai Branch, Taipei, Taiwan
| | - Szu-Ying Lin
- Taipei Municipal Gan-Dau Hospital, Taipei, Taiwan
| | - Po-Chen Lin
- Division of General Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Pei-Ning Wang
- Division of General Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.,Aging and Health Research Center, National Yang-Ming University, Taipei, Taiwan.,Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Irene H Cheng
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan, .,Brain Research Center, National Yang-Ming University, Taipei, Taiwan,
| |
Collapse
|
37
|
Johnson TP, Sejvar J, Nutman TB, Nath A. The Pathogenesis of Nodding Syndrome. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2020; 15:395-417. [PMID: 31977293 DOI: 10.1146/annurev-pathmechdis-012419-032748] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nodding syndrome is a rare, enigmatic form of pediatric epilepsy that has occurred in an epidemic fashion beginning in the early 2000s in geographically distinct regions of Africa. Despite extensive investigation, the etiology of nodding syndrome remains unclear, although much progress has been made in understanding the pathogenesis of the disease, as well as in treatment and prevention. Nodding syndrome is recognized as a defined disease entity, but it is likely one manifestation along a continuum of Onchocerca volvulus-associated neurological complications. This review examines the epidemiology of nodding syndrome and its association with environmental factors. It provides a critical analysis of the data that support or contradict the leading hypotheses of the etiologies underlying the pathogenesis of the syndrome. It also highlights the important progress made in treating and preventing this devastating neurological disease and prioritizes important areas for future research.
Collapse
Affiliation(s)
- Tory P Johnson
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - James Sejvar
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia 30329-4027, USA
| | - Thomas B Nutman
- Helminth Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA;
| |
Collapse
|
38
|
Li LX, Yang T, Guo L, Wang DY, Tang CH, Li Q, Yang HM, Zhu J, Zhang LL. Serum tau levels are increased in patients with hyperthyroidism. Neurosci Lett 2020; 729:135003. [PMID: 32335219 DOI: 10.1016/j.neulet.2020.135003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 12/19/2022]
Abstract
Hyperthyroidism may cause cognitive decline and increases the risk of Alzheimer's disease (AD), the major form of dementia; however, the underlying mechanism of this relationship is unclear. AD is associated with increased serum levels of tau. In this study, we investigated the relationship between serum thyroid hormones (THs) and tau. Fifty participants diagnosed with hyperthyroidism and fifty euthyroid counterparts were included and received clinical examinations. Serum concentrations of thyroid-stimulating hormone (TSH), free thyroxine (FT4), free triiodothyronine (FT3) and tau protein were assessed. The total tau protein level was significantly higher in hyperthyroidism participants than in their euthyroid counterparts. The level of circulating total tau had a significant positive association with the serum concentrations of FT3 and FT4. Total tau level was increased in the low TSH group and the serum THs decreased with the increase of age. These findings reveal that peripheral THs are associated with the serum concentration of tau, which may be involved in the pathogenesis of AD, suggesting a potential therapeutic target of AD via hyperthyroidism therapy.
Collapse
Affiliation(s)
- Lun-Xi Li
- Department of Neurology, Army Medical University Daping Hospital, China
| | - Tong Yang
- Department of Neurology, Army Medical University Daping Hospital, China
| | - Lu Guo
- Department of Neurology, Army Medical University Daping Hospital, China
| | - Da-Yan Wang
- Department of Neurology, the Seventh Affiliated Hospital, Sun Yat-sen University, China
| | - Chun-Hua Tang
- Department of Neurology, Army Medical University Daping Hospital, China
| | - Qiong Li
- Department of Neurology, Army Medical University Daping Hospital, China
| | - Hai-Mei Yang
- Department of Neurology, Army Medical University Daping Hospital, China
| | - Jie Zhu
- Department of Neurology, Army Medical University Daping Hospital, China.
| | - Li-Li Zhang
- Department of Neurology, Army Medical University Daping Hospital, China
| |
Collapse
|
39
|
Hampel H, Vergallo A, Afshar M, Akman-Anderson L, Arenas J, Benda N, Batrla R, Broich K, Caraci F, Cuello AC, Emanuele E, Haberkamp M, Kiddle SJ, Lucía A, Mapstone M, Verdooner SR, Woodcock J, Lista S. Blood-based systems biology biomarkers for next-generation clinical trials in Alzheimer's disease
. DIALOGUES IN CLINICAL NEUROSCIENCE 2020. [PMID: 31636492 PMCID: PMC6787542 DOI: 10.31887/dcns.2019.21.2/hhampel] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD)-a complex disease showing multiple pathomechanistic alterations-is triggered by nonlinear dynamic interactions of genetic/epigenetic and environmental risk factors, which, ultimately, converge into a biologically heterogeneous disease. To tackle the burden of AD during early preclinical stages, accessible blood-based biomarkers are currently being developed. Specifically, next-generation clinical trials are expected to integrate positive and negative predictive blood-based biomarkers into study designs to evaluate, at the individual level, target druggability and potential drug resistance mechanisms. In this scenario, systems biology holds promise to accelerate validation and qualification for clinical trial contexts of use-including proof-of-mechanism, patient selection, assessment of treatment efficacy and safety rates, and prognostic evaluation. Albeit in their infancy, systems biology-based approaches are poised to identify relevant AD "signatures" through multifactorial and interindividual variability, allowing us to decipher disease pathophysiology and etiology. Hopefully, innovative biomarker-drug codevelopment strategies will be the road ahead towards effective disease-modifying drugs.
.
Collapse
Affiliation(s)
- Harald Hampel
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Andrea Vergallo
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Mohammad Afshar
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Leyla Akman-Anderson
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Joaquín Arenas
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Norbert Benda
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Richard Batrla
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Karl Broich
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Filippo Caraci
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - A Claudio Cuello
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Enzo Emanuele
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Marion Haberkamp
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Steven J Kiddle
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Alejandro Lucía
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Mark Mapstone
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Steven R Verdooner
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Janet Woodcock
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| | - Simone Lista
- Author affiliations: AXA Research Fund & Sorbonne University Chair, Paris, France; Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France; Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France (Harald Hampel, Andrea Vergallo, Simone Lista); Ariana Pharma, Paris, France (Mohammad Afshar); NeuroVision Imaging, Inc., Sacramento, California, USA (Leyla Akman-Anderson, Steven R. Verdooner); Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain (Joaquín Arenas, Alejandro Lucía); Biostatistics and Special Pharmacokinetics Unit/Research Division, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Norbert Benda); Roche Diagnostics International, Rotkreuz, Switzerland (Richard Batrla); Head and President, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Karl Broich); Department of Drug Sciences, University of Catania, Catania, Italy; IRCCS Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, Troina, Enna, Italy (Filippo Caraci); Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada (A. Claudio Cuello); 2E Science, Robbio, Pavia, Italy (Enzo Emanuele); Neurology/Psychiatry/Ophthalmology Unit, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany (Marion Haberkamp); MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK (Steven J. Kiddle); Universidad Europea de Madrid (Sports Science Department), Madrid, Spain (Alejandro Lucía); Department of Neurology, University of California Irvine School of Medicine, Irvine, California, USA (Mark Mapstone); Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA (Janet Woodcock). Address for correspondence: Professor Harald Hampel, MD, PhD, Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, 47 boulevard de l'hôpital, F-75013, Paris, France.
| |
Collapse
|
40
|
Milà-Alomà M, Suárez-Calvet M, Molinuevo JL. Latest advances in cerebrospinal fluid and blood biomarkers of Alzheimer's disease. Ther Adv Neurol Disord 2019; 12:1756286419888819. [PMID: 31897088 PMCID: PMC6920596 DOI: 10.1177/1756286419888819] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and its diagnosis has classically been based on clinical symptoms. Recently, a biological rather than a syndromic definition of the disease has been proposed that is based on biomarkers that reflect neuropathological changes. In AD, there are two main biomarker categories, namely neuroimaging and fluid biomarkers [cerebrospinal fluid (CSF) and blood]. As a complex and multifactorial disease, AD biomarkers are important for an accurate diagnosis and to stage the disease, assess the prognosis, test target engagement, and measure the response to treatment. In addition, biomarkers provide us with information that, even if it does not have a current clinical use, helps us to understand the mechanisms of the disease. In addition to the pathological hallmarks of AD, which include amyloid-β and tau deposition, there are multiple concomitant pathological events that play a key role in the disease. These include, but are not limited to, neurodegeneration, inflammation, vascular dysregulation or synaptic dysfunction. In addition, AD patients often have an accumulation of other proteins including α-synuclein and TDP-43, which may have a pathogenic effect on AD. In combination, there is a need to have biomarkers that reflect different aspects of AD pathogenesis and this will be important in the future to establish what are the most suitable applications for each of these AD-related biomarkers. It is unclear whether sex, gender, or both have an effect on the causes of AD. There may be differences in fluid biomarkers due to sex but this issue has often been neglected and warrants further research. In this review, we summarize the current state of the principal AD fluid biomarkers and discuss the effect of sex on these biomarkers.
Collapse
Affiliation(s)
- Marta Milà-Alomà
- Barcelonaβeta Brain Research Center (BBRC),
Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research
Institute), Barcelona
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC),
Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research
Institute), Barcelona
- Department of Neurology, Hospital del Mar,
Barcelona
| | - José Luís Molinuevo
- Scientific Director, Alzheimer’s Prevention
Program, Barcelonaβeta Brain Research Center, Wellington 30, Barcelona,
08005, Spain
- IMIM (Hospital del Mar Medical Research
Institute), Barcelona
- CIBER Fragilidad y Envejecimiento Saludable,
Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
41
|
Soler M, Huertas CS, Lechuga LM. Label-free plasmonic biosensors for point-of-care diagnostics: a review. Expert Rev Mol Diagn 2018; 19:71-81. [PMID: 30513011 DOI: 10.1080/14737159.2019.1554435] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: Optical biosensors, particularly those based on nanoplasmonics technology, have emerged in recent decades as a potential solution for disease diagnostics and therapy follow-up at the point-of-care (POC). These biosensor platforms could overcome some of the challenges faced in conventional diagnosis techniques offering label-free assays with immediate results and employing small and user-friendly devices. Areas covered: In this review, we will provide a critical overview of the recent advances in the development of nanoplasmonic biosensors for the POC diagnostics. We focus on those systems with demonstrated capabilities for integration in portable platforms, highlighting some of the most relevant diagnostics applications targeting proteins, nucleic acids, and cells as disease biomarkers. Expert commentary: Despite the attractive features of label-free nanoplasmonic sensors in terms of miniaturization and analytical robustness, the route toward an effective clinical implementation involves the integration of fully automated microfluidic systems for sample processing and analysis, and the optimization of surface biofunctionalization procedures. Additionally, the development of multiplexed sensors for high-throughput analysis and including specific neoantigens and novel biomarkers in detection panels will provide the means for delivering a powerful analytical technology for an accurate and improved medical diagnosis.
Collapse
Affiliation(s)
- Maria Soler
- a Nanobiosensors and Bioanalytical Applications Group , Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, BIST and CIBER-BBN , Bellaterra , Barcelona , Spain
| | - Cesar S Huertas
- a Nanobiosensors and Bioanalytical Applications Group , Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, BIST and CIBER-BBN , Bellaterra , Barcelona , Spain.,b School of Engineering , RMIT University , Melbourne , Australia
| | - Laura M Lechuga
- a Nanobiosensors and Bioanalytical Applications Group , Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, BIST and CIBER-BBN , Bellaterra , Barcelona , Spain
| |
Collapse
|
42
|
Molinuevo JL, Ayton S, Batrla R, Bednar MM, Bittner T, Cummings J, Fagan AM, Hampel H, Mielke MM, Mikulskis A, O'Bryant S, Scheltens P, Sevigny J, Shaw LM, Soares HD, Tong G, Trojanowski JQ, Zetterberg H, Blennow K. Current state of Alzheimer's fluid biomarkers. Acta Neuropathol 2018; 136:821-853. [PMID: 30488277 PMCID: PMC6280827 DOI: 10.1007/s00401-018-1932-x] [Citation(s) in RCA: 360] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 12/12/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease with a complex and heterogeneous pathophysiology. The number of people living with AD is predicted to increase; however, there are no disease-modifying therapies currently available and none have been successful in late-stage clinical trials. Fluid biomarkers measured in cerebrospinal fluid (CSF) or blood hold promise for enabling more effective drug development and establishing a more personalized medicine approach for AD diagnosis and treatment. Biomarkers used in drug development programmes should be qualified for a specific context of use (COU). These COUs include, but are not limited to, subject/patient selection, assessment of disease state and/or prognosis, assessment of mechanism of action, dose optimization, drug response monitoring, efficacy maximization, and toxicity/adverse reactions identification and minimization. The core AD CSF biomarkers Aβ42, t-tau, and p-tau are recognized by research guidelines for their diagnostic utility and are being considered for qualification for subject selection in clinical trials. However, there is a need to better understand their potential for other COUs, as well as identify additional fluid biomarkers reflecting other aspects of AD pathophysiology. Several novel fluid biomarkers have been proposed, but their role in AD pathology and their use as AD biomarkers have yet to be validated. In this review, we summarize some of the pathological mechanisms implicated in the sporadic AD and highlight the data for several established and novel fluid biomarkers (including BACE1, TREM2, YKL-40, IP-10, neurogranin, SNAP-25, synaptotagmin, α-synuclein, TDP-43, ferritin, VILIP-1, and NF-L) associated with each mechanism. We discuss the potential COUs for each biomarker.
Collapse
Affiliation(s)
- José Luis Molinuevo
- BarcelonaBeta Brain Research Center, Fundació Pasqual Maragall, Universitat Pompeu Fabra, Barcelona, Spain
- Unidad de Alzheimer y otros trastornos cognitivos, Hospital Clinic-IDIBAPS, Barcelona, Spain
| | - Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Richard Batrla
- Roche Centralised and Point of Care Solutions, Roche Diagnostics International, Rotkreuz, Switzerland
| | - Martin M Bednar
- Neuroscience Therapeutic Area Unit, Takeda Development Centre Americas Ltd, Cambridge, MA, USA
| | - Tobias Bittner
- Genentech, A Member of the Roche Group, Basel, Switzerland
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Harald Hampel
- AXA Research Fund and Sorbonne University Chair, Paris, France
- Sorbonne University, GRC No 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Brain and Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Michelle M Mielke
- Departments of Epidemiology and Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Sid O'Bryant
- Department of Pharmacology and Neuroscience; Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Philip Scheltens
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Jeffrey Sevigny
- Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA
| | - Holly D Soares
- Clinical Development Neurology, AbbVie, North Chicago, IL, USA
| | | | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal Campus, Sahlgrenska University Hospital, 431 80, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal Campus, Sahlgrenska University Hospital, 431 80, Mölndal, Sweden.
| |
Collapse
|
43
|
Alzheimer’s disease (AD) therapeutics – 1: Repeated clinical failures continue to question the amyloid hypothesis of AD and the current understanding of AD causality. Biochem Pharmacol 2018; 158:359-375. [DOI: 10.1016/j.bcp.2018.09.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/24/2018] [Indexed: 12/17/2022]
|
44
|
Zheng H, Cheng B, Li Y, Li X, Chen X, Zhang YW. TREM2 in Alzheimer's Disease: Microglial Survival and Energy Metabolism. Front Aging Neurosci 2018; 10:395. [PMID: 30532704 PMCID: PMC6265312 DOI: 10.3389/fnagi.2018.00395] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of age-related dementia among the elderly population. Recent genetic studies have identified rare variants of the gene encoding the triggering receptor expressed on myeloid cells-2 (TREM2) as significant genetic risk factors in late-onset AD (LOAD). TREM2 is specifically expressed in brain microglia and modulates microglial functions in response to key AD pathologies such as amyloid-β (Aβ) plaques and tau tangles. In this review article, we discuss recent research progress in our understanding on the role of TREM2 in microglia and its relevance to AD pathologies. In addition, we discuss evidence describing new TREM2 ligands and the role of TREM2 signaling in microglial survival and energy metabolism. A comprehensive understanding of TREM2 function in the pathogenesis of AD offers a unique opportunity to explore the potential of this microglial receptor as an alternative target in AD therapy.
Collapse
Affiliation(s)
- Honghua Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China.,Shenzhen Research Institute, Xiamen University, Shenzhen, China
| | - Baoying Cheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China
| | - Yanfang Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China.,Shenzhen Research Institute, Xiamen University, Shenzhen, China
| | - Xin Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China
| | - Xiaofen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China.,Shenzhen Research Institute, Xiamen University, Shenzhen, China
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China
| |
Collapse
|
45
|
Neuroprotection by ethanolic extract of Syzygium aromaticum in Alzheimer's disease like pathology via maintaining oxidative balance through SIRT1 pathway. Exp Gerontol 2018; 110:277-283. [PMID: 29959974 DOI: 10.1016/j.exger.2018.06.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/15/2018] [Accepted: 06/25/2018] [Indexed: 11/17/2022]
Abstract
The oxidative stress plays a key role in Alzheimer's disease (AD) and Sirtuin (SIRT1) is potential mediator of oxidative pathway. This study explored the role of Syzygium aromaticum on SIRT1 and oxidative balance in amyloid beta induced toxicity. Anti-oxidative capacity of Syzygium aromaticum was performed in Aβ25-35 induced neurotoxicity in neuronal cells. Superoxide dismutase, Catalase and Glutathione enzyme activity were determined by the treatment of Syzygium aromaticum. Both recombinant and endogenous SIRT1 activity were performed in its presence. The expression of γ-secretase and SIRT1 were evaluated by western blot. Syzygium aromaticum was capable to scavenge ROS and elevate the percentage of anti-oxidant enzymes. It also activated and elevated the level of SIRT1 and downregulated γ-secretase level. These findings show a holistic approach towards the neurodegenerative disease management by Syzygium aromaticum which could lead to the formulation of new drug for AD. This Ayurvedic product can give a healthy aging with no side effects and also be cost effectives. It may meet unmet medical needs of current relevance.
Collapse
|
46
|
Salama M, Shalash A, Magdy A, Makar M, Roushdy T, Elbalkimy M, Elrassas H, Elkafrawy P, Mohamed W, Abou Donia MB. Tubulin and Tau: Possible targets for diagnosis of Parkinson's and Alzheimer's diseases. PLoS One 2018; 13:e0196436. [PMID: 29742117 PMCID: PMC5942772 DOI: 10.1371/journal.pone.0196436] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 04/12/2018] [Indexed: 11/29/2022] Open
Abstract
Neurodegenerative diseases including Alzheimer's disease (AD) and Parkinson's disease (PD) are characterized by progressive neuronal loss and pathological accumulation of some proteins. Developing new biomarkers for both diseases is highly important for the early diagnosis and possible development of neuro-protective strategies. Serum antibodies (AIAs) against neuronal proteins are potential biomarkers for AD and PD that may be formed in response to their release into systemic circulation after brain damage. In the present study, two AIAs (tubulin and tau) were measured in sera of patients of PD and AD, compared to healthy controls. Results showed that both antibodies were elevated in patients with PD and AD compared to match controls. Curiously, the profile of elevation of antibodies was different in both diseases. In PD cases, tubulin and tau AIAs levels were similar. On the other hand, AD patients showed more elevation of tau AIAs compared to tubulin. Our current results suggested that AIAs panel could be able to identify cases with neuro-degeneration when compared with healthy subjects. More interestingly, it is possible to differentiate between PD and AD cases through identifying specific AIAs profile for each neurodegenerative states.
Collapse
Affiliation(s)
- Mohamed Salama
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ali Shalash
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Alshimaa Magdy
- Biochemistry Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Marianne Makar
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Tamer Roushdy
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mahmoud Elbalkimy
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hanan Elrassas
- Okasha Institute of Psychiatry, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Wael Mohamed
- Department of Pharmacology, Faculty of Medicine, Menoufia University, Shebeen Elkoum, Egypt
- Basic Medical Science Department, Kulliyyah of Medicine, International Islamic University Malaysia, Kuantan Pahang, Malaysia
| | - Mohamed B. Abou Donia
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
47
|
Altuna-Azkargorta M, Mendioroz-Iriarte M. Blood biomarkers in Alzheimer's disease. Neurologia 2018; 36:S0213-4853(18)30091-4. [PMID: 29752036 DOI: 10.1016/j.nrl.2018.03.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/20/2018] [Accepted: 03/01/2018] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION The early diagnosis of Alzheimer's disease (AD) via the use of biomarkers could facilitate the implementation and monitoring of early therapeutic interventions with the potential capacity to significantly modify the course of the disease. DEVELOPMENT Classic cerebrospinal fluid biomarkers and approved structural and functional neuroimaging have a limited clinical application given their invasive nature and/or high cost. The identification of more accessible and less costly biomarkers, such as blood biomarkers, would facilitate application in clinical practice. We present a literature review of the main blood biochemical biomarkers with potential use for diagnosing Alzheimer's disease. CONCLUSIONS Blood biomarkers are cost and time effective with regard to cerebrospinal fluid biomarkers. However, the immediate applicability of blood biochemical biomarkers in clinical practice is not very likely. The main limitations come from the difficulties in measuring and standardising thresholds between different laboratories and in failures to replicate results. Among all the molecules studied, apoptosis and neurodegeneration biomarkers and the biomarker panels obtained through omics approaches, such as isolated or combined metabolomics, offer the most promising results.
Collapse
Affiliation(s)
- M Altuna-Azkargorta
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, España.
| | - M Mendioroz-Iriarte
- Laboratorio de Neuroepigenética, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, España; Servicio de Neurología, Complejo Hospitalario de Navarra, Pamplona, España
| |
Collapse
|
48
|
Motamedi V, Kanefsky R, Matsangas P, Mithani S, Jeromin A, Brock MS, Mysliwiec V, Gill J. Elevated tau and interleukin-6 concentrations in adults with obstructive sleep apnea. Sleep Med 2018; 43:71-76. [DOI: 10.1016/j.sleep.2017.11.1121] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/04/2017] [Accepted: 11/10/2017] [Indexed: 11/26/2022]
|
49
|
Vu Nu TT, Tran NHT, Nam E, Nguyen TT, Yoon WJ, Cho S, Kim J, Chang KA, Ju H. Blood-based immunoassay of tau proteins for early diagnosis of Alzheimer's disease using surface plasmon resonance fiber sensors. RSC Adv 2018; 8:7855-7862. [PMID: 35539129 PMCID: PMC9078509 DOI: 10.1039/c7ra11637c] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 02/06/2018] [Indexed: 11/21/2022] Open
Abstract
We present the immunoassay of tau proteins (total tau and phosphorylated tau) in human sera using surface plasmon resonance (SPR) fiber sensors. This assay aimed at harvesting the advantages of using both SPR fiber sensors and a blood-based assay to demonstrate label-free point-of-care-testing (POCT) patient-friendly assay in a compact format for the early diagnosis of Alzheimer's disease (AD). For conducting the assay, we used human sera of 40 subjects divided into halves, which were grouped into AD patients and control groups according to a number of neuropsychological tests. We found that on an average, the concentrations of both total tau and phosphorylated tau proteins (all known to be higher in cerebrospinal fluid (CSF) and the brain) turned out to be higher in human sera of AD patients than in controls. The limits of detection of total tau and phosphorylated tau proteins were 2.4 pg mL−1 and 1.6 pg mL−1, respectively. In particular, it was found that the AD group exhibited average concentration of total tau proteins 6-fold higher than the control group, while concentration of phosphorylated tau proteins was 3-fold higher than that of the control. We can attribute this inhomogeneity between both types of tau proteins (in terms of increase of control-to-AD in average concentration) to un-phosphorylated tau proteins being more likely to be produced in blood than phosphorylated tau proteins, which possibly is one of the potential key elements playing an important role in AD progress. Blood-based early diagnosis of Alzheimer's disease using a plasmonic fiber sensor that detects immunoreaction of tau proteins.![]()
Collapse
Affiliation(s)
- Truong Thi Vu Nu
- Department of Nano-Physics
- Gachon University
- Seongnam-si
- Republic of Korea
- GachonBionano Research Institute
| | - Nhu Hoa Thi Tran
- Department of Nano-Physics
- Gachon University
- Seongnam-si
- Republic of Korea
- GachonBionano Research Institute
| | - Eunjoo Nam
- Department of Pharmacology
- College of Medicine
- Neuroscience Research Institute
- Gachon University
- Incheon
| | - Tan Tai Nguyen
- Department of Materials Science
- School of Basic Science
- TraVinh University
- TraVinh City
- Vietnam
| | - Won Jung Yoon
- Department of Chemical and Bioengineering
- Gachon University
- Seongnam-si
- Republic of Korea
| | - Sungbo Cho
- Gachon Advanced Institute for Health Science and Technology
- Gachon University
- Incheon 21999
- Republic of Korea
- Department of Biomedical Engineering
| | - Jungsuk Kim
- Gachon Advanced Institute for Health Science and Technology
- Gachon University
- Incheon 21999
- Republic of Korea
- Department of Biomedical Engineering
| | - Keun-A. Chang
- Department of Pharmacology
- College of Medicine
- Neuroscience Research Institute
- Gachon University
- Incheon
| | - Heongkyu Ju
- Department of Nano-Physics
- Gachon University
- Seongnam-si
- Republic of Korea
- GachonBionano Research Institute
| |
Collapse
|
50
|
Wang H, Stewart T, Toledo JB, Ginghina C, Tang L, Atik A, Aro P, Shaw LM, Trojanowski JQ, Galasko DR, Edland S, Jensen PH, Shi M, Zhang J, for The Alzheimer’s Disease Neuroimaging Initiative. A Longitudinal Study of Total and Phosphorylated α-Synuclein with Other Biomarkers in Cerebrospinal Fluid of Alzheimer's Disease and Mild Cognitive Impairment. J Alzheimers Dis 2018; 61:1541-1553. [PMID: 29376878 PMCID: PMC5828159 DOI: 10.3233/jad-171013] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) features a dynamic sequence of amyloid deposition, neurodegeneration, and cognitive impairment. A significant fraction of AD brains also displays Lewy body pathology, suggesting that addition of classically Parkinson's disease-related proteins to the AD biomarker panel may be of value. To determine whether addition of cerebrospinal fluid (CSF) total α-synuclein and its form phosphorylated at S129 (pS129) to the AD biomarker panel [Amyloid-β1-42 (Aβ42), tau, and phosphorylated tau (p-tau181)] improves its performance, we examined CSF samples collected longitudinally up to 7 years as part of the Alzheimer's Disease Neuroimaging Initiative. From 87 AD, 177 mild cognitive impairment (MCI), and 104 age-matched healthy controls, 792 baseline and longitudinal CSF samples were tested for total α-synuclein, pS129, Aβ42, tau, and p-tau181. pS129, but not total α-synuclein, was weakly associated with diagnosis at baseline when t-tau/Aβ42 was included in the statistical model (β= 0.0026, p = 0.041, 95% CI [(0.0001)-(0.005)]). CSF α-synuclein predicted Alzheimer's Disease Assessment Scale-Cognitive (β= -0.59, p = 0.0015, 95% CI [(-0.96)-(-0.23)]), memory (β= 0.4, p = 0.00025, 95% CI [(0.16)-(0.59)]), and executive (0.62,<0.0001, 95% CI [(0.31)-(0.93)]) function composite scores, and progression from MCI to AD (β= 0.019, p = 0.0011, 95% CI [(0.002)-(0.20)]). pS129 was associated with executive function (β= -2.55, p = 0.0085, 95% CI [(-4.45)-(-0.66)]). Lower values in the mismatch between α-synuclein and p-tau181 predicted faster cognitive decline (β= 0.64, p = 0.0012, 95% CI [(0.48)-(0.84)]). Longitudinal biomarker changes did not differ between groups, and may not reflect AD progression. The α-synuclein-p-tau181-Mismatch could better predict longitudinal cognitive changes than classical AD markers alone, and its pathological correlates should be investigated further.
Collapse
Affiliation(s)
- Hua Wang
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
- Department of Pathology, Peking University Health Science Centre and Third Hospital, Beijing, China
| | - Tessandra Stewart
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
| | - Jon B. Toledo
- Department of Pathology & Laboratory Medicine, Institute on Aging, Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Carmen Ginghina
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
| | - Lu Tang
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
| | - Anzari Atik
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
| | - Patrick Aro
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Q. Trojanowski
- Department of Pathology & Laboratory Medicine, Institute on Aging, Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Alzheimer’s Disease Core Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Udall Parkinson’s Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas R. Galasko
- Shiley-Marcos Alzheimer’s Disease Research Center, Department of Neurosciences, University of California, San Diego, CA USA
| | - Steven Edland
- Shiley-Marcos Alzheimer’s Disease Research Center, Department of Neurosciences, University of California, San Diego, CA USA
- Division of Biostatistics and Bioinformatics, University of California San Diego, San Diego, United States
| | - Poul H. Jensen
- University of Aarhus, DANDRITE—Danish Research Institute of Translational Neuroscience & Department of Biomedicine, Aarhus, Denmark
| | - Min Shi
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
| | - Jing Zhang
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
- Department of Pathology, Peking University Health Science Centre and Third Hospital, Beijing, China
| | | |
Collapse
|