1
|
Mathias K, Machado RS, Cardoso T, Tiscoski ADB, Piacentini N, Prophiro JS, Generoso JS, Barichello T, Petronilho F. The Blood-Cerebrospinal Fluid Barrier Dysfunction in Brain Disorders and Stroke: Why, How, What For? Neuromolecular Med 2024; 26:38. [PMID: 39278883 DOI: 10.1007/s12017-024-08806-0] [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: 08/08/2024] [Accepted: 09/03/2024] [Indexed: 09/18/2024]
Abstract
Ischemic stroke (IS) results in the interruption of blood flow to the brain, which can cause significant damage. The pathophysiological mechanisms of IS include ionic imbalances, oxidative stress, neuroinflammation, and impairment of brain barriers. Brain barriers, such as the blood-brain barrier (BBB) and the blood-cerebrospinal fluid (CSF) barrier (B-CSF), protect the brain from harmful substances by regulating the neurochemical environment. Although the BBB is widely recognized for its crucial role in protecting the brain and its involvement in conditions such as stroke, the B-CSF requires further study. The B-CSF plays a fundamental role in regulating the CSF environment and maintaining the homeostasis of the central nervous system (CNS). However, the impact of B-CSF impairment during pathological events such as IS is not yet fully understood. In conditions like IS and other neurological disorders, the B-CSF can become compromised, allowing the entry of inflammatory substances and increasing neuronal damage. Understanding and preserving the integrity of the B-CSF are crucial for mitigating damage and facilitating recovery after ischemic stroke, highlighting its fundamental role in regulating the CNS during adverse neurological conditions.
Collapse
Affiliation(s)
- Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Richard Simon Machado
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Taise Cardoso
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Anita Dal Bó Tiscoski
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Natália Piacentini
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Josiane Somariva Prophiro
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Jaqueline Silva Generoso
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Tatiana Barichello
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, 77054, USA
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil.
- Laboratory of Experimental Neurology, University of Extremo Sul Catarinense, Criciuma, SC, Brazil.
| |
Collapse
|
2
|
Chen R, Wang X, Li N, Golubnitschaja O, Zhan X. Body fluid multiomics in 3PM-guided ischemic stroke management: health risk assessment, targeted protection against health-to-disease transition, and cost-effective personalized approach are envisaged. EPMA J 2024; 15:415-452. [PMID: 39239108 PMCID: PMC11371995 DOI: 10.1007/s13167-024-00376-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
Because of its rapid progression and frequently poor prognosis, stroke is the third major cause of death in Europe and the first one in China. Many independent studies demonstrated sufficient space for prevention interventions in the primary care of ischemic stroke defined as the most cost-effective protection of vulnerable subpopulations against health-to-disease transition. Although several studies identified molecular patterns specific for IS in body fluids, none of these approaches has yet been incorporated into IS treatment guidelines. The advantages and disadvantages of individual body fluids are thoroughly analyzed throughout the paper. For example, multiomics based on a minimally invasive approach utilizing blood and its components is recommended for real-time monitoring, due to the particularly high level of dynamics of the blood as a body system. On the other hand, tear fluid as a more stable system is recommended for a non-invasive and patient-friendly holistic approach appropriate for health risk assessment and innovative screening programs in cost-effective IS management. This article details aspects essential to promote the practical implementation of highlighted achievements in 3PM-guided IS management. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00376-2.
Collapse
Affiliation(s)
- Ruofei Chen
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 P. R. China
| | - Xiaoyan Wang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 P. R. China
| | - Na Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 P. R. China
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, University Hospital Bonn, Venusberg Campus 1, Rheinische Friedrich-Wilhelms-University of Bonn, Bonn, 53127 Germany
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, Shandong 250117 P. R. China
| |
Collapse
|
3
|
Ahmad S, Yang W, Orellana A, Frölich L, de Rojas I, Cano A, Boada M, Hernández I, Hausner L, Harms AC, Bakker MHM, Cabrera-Socorro A, Amin N, Ramírez A, Ruiz A, Van Duijn CM, Hankemeier T. Association of oxidative stress and inflammatory metabolites with Alzheimer's disease cerebrospinal fluid biomarkers in mild cognitive impairment. Alzheimers Res Ther 2024; 16:171. [PMID: 39080778 PMCID: PMC11287840 DOI: 10.1186/s13195-024-01542-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND Isoprostanes and prostaglandins are biomarkers for oxidative stress and inflammation. Their role in Alzheimer's disease (AD) pathophysiology is yet unknown. In the current study, we aim to identify the association of isoprostanes and prostaglandins with the Amyloid, Tau, Neurodegeneration (ATN) biomarkers (Aβ-42, p-tau, and t-tau) of AD pathophysiology in mild cognitive impairment (MCI) subjects. METHODS Targeted metabolomics profiling was performed using liquid chromatography-mass spectrometry (LCMS) in 147 paired plasma-CSF samples from the Ace Alzheimer Center Barcelona and 58 CSF samples of MCI patients from the Mannheim/Heidelberg cohort. Linear regression was used to evaluate the association of metabolites with CSF levels of ATN biomarkers in the overall sample and stratified by Aβ-42 pathology and APOE genotype. We further evaluated the role of metabolites in MCI to AD dementia progression. RESULTS Increased CSF levels of PGF2α, 8,12-iso-iPF2α VI, and 5-iPF2α VI were significantly associated (False discovery rate (FDR) < 0.05) with higher p-tau levels. Additionally, 8,12-iso-iPF2α VI was associated with increased total tau levels in CSF. In MCI due to AD, PGF2α was associated with both p-tau and total tau, whereases 8,12-iso-iPF2α VI was specifically associated with p-tau levels. In APOE stratified analysis, association of PGF2α with p-tau and t-tau was observed in only APOE ε4 carriers while 5-iPF2α VI showed association with both p-tau and t-tau in APOE ε33 carriers. CSF levels of 8,12- iso-iPF2α VI showed association with p-tau and t-tau in APOE ε33/APOE ε4 carriers and with t-tau in APOE ε3 carriers. None of the metabolites showed evidence of association with MCI to AD progression. CONCLUSIONS Oxidative stress (8,12-iso-iPF2α VI) and inflammatory (PGF2α) biomarkers are correlated with biomarkers of AD pathology during the prodromal stage of AD and relation of PGF2α with tau pathology markers may be influenced by APOE genotype.
Collapse
Affiliation(s)
- Shahzad Ahmad
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
- Oxford-GSK Institute of Molecular and Computational Medicine (IMCM), Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Wei Yang
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Adelina Orellana
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Amanda Cano
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Hernández
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Lucrezia Hausner
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
| | - Amy C Harms
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Margot H M Bakker
- Discovery Research, AbbVie Deutschland GmbH & Co. KG, 67061, KnollstrasseLudwigshafen, Germany
| | | | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands
- Nuffield Department of Population Health, University of Oxford, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Old Road Campus, , Headington-Oxford, OX3 7FZ, UK
| | - Alfredo Ramírez
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931, Cologne, Germany
- Department of Psychiatry and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, USA
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Cornelia M Van Duijn
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands.
- Nuffield Department of Population Health, University of Oxford, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Old Road Campus, , Headington-Oxford, OX3 7FZ, UK.
| | - Thomas Hankemeier
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands.
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
4
|
Li QY, Hu HY, Zhang GW, Hu H, Ou YN, Huang LY, Wang AY, Gao PY, Ma LY, Tan L, Yu JT. Associations between cardiometabolic multimorbidity and cerebrospinal fluid biomarkers of Alzheimer's disease pathology in cognitively intact adults: the CABLE study. Alzheimers Res Ther 2024; 16:28. [PMID: 38321520 PMCID: PMC10848421 DOI: 10.1186/s13195-024-01396-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/21/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Cardiometabolic multimorbidity is associated with an increased risk of dementia, but the pathogenic mechanisms linking them remain largely undefined. We aimed to assess the associations of cardiometabolic multimorbidity with cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease (AD) pathology to enhance our understanding of the underlying mechanisms linking cardiometabolic multimorbidity and AD. METHODS This study included 1464 cognitively intact participants from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) database. Cardiometabolic diseases (CMD) are a group of interrelated disorders such as hypertension, diabetes, heart diseases (HD), and stroke. Based on the CMD status, participants were categorized as CMD-free, single CMD, or CMD multimorbidity. CMD multimorbidity is defined as the coexistence of two or more CMDs. The associations of cardiometabolic multimorbidity and CSF biomarkers were examined using multivariable linear regression models with demographic characteristics, the APOE ε4 allele, and lifestyle factors as covariates. Subgroup analyses stratified by age, sex, and APOE ε4 status were also performed. RESULTS A total of 1464 individuals (mean age, 61.80 years; age range, 40-89 years) were included. The markers of phosphorylated tau-related processes (CSF P-tau181: β = 0.165, P = 0.037) and neuronal injury (CSF T-tau: β = 0.065, P = 0.033) were significantly increased in subjects with CMD multimorbidity (versus CMD-free), but not in those with single CMD. The association between CMD multimorbidity with CSF T-tau levels remained significant after controlling for Aβ42 levels. Additionally, significantly elevated tau-related biomarkers were observed in patients with specific CMD combinations (i.e., hypertension and diabetes, hypertension and HD), especially in long disease courses. CONCLUSIONS The presence of cardiometabolic multimorbidity was associated with tau phosphorylation and neuronal injury in cognitively normal populations. CMD multimorbidity might be a potential independent target to alleviate tau-related pathologies that can cause cognitive impairment.
Collapse
Affiliation(s)
- Qiong-Yao Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Gao-Wen Zhang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - An-Yi Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Pei-Yang Gao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Li-Yun Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, No. 12 Wulumuqi Road, Shanghai, China.
| |
Collapse
|
5
|
Naik A, Adeleye O, Koester SW, Winkler EA, Hartke JN, Karahalios K, Mihaljevic S, Rani A, Raikwar S, Rulney JD, Desai SM, Scherschinski L, Ducruet AF, Albuquerque FC, Lawton MT, Catapano JS, Jadhav AP, Jha RM. Cerebrospinal Fluid Biomarkers for Diagnosis and the Prognostication of Acute Ischemic Stroke: A Systematic Review. Int J Mol Sci 2023; 24:10902. [PMID: 37446092 DOI: 10.3390/ijms241310902] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Despite the high incidence and burden of stroke, biological biomarkers are not used routinely in clinical practice to diagnose, determine progression, or prognosticate outcomes of acute ischemic stroke (AIS). Because of its direct interface with neural tissue, cerebrospinal fluid (CSF) is a potentially valuable source for biomarker development. This systematic review was conducted using three databases. All trials investigating clinical and preclinical models for CSF biomarkers for AIS diagnosis, prognostication, and severity grading were included, yielding 22 human trials and five animal studies for analysis. In total, 21 biomarkers and other multiomic proteomic markers were identified. S100B, inflammatory markers (including tumor necrosis factor-alpha and interleukin 6), and free fatty acids were the most frequently studied biomarkers. The review showed that CSF is an effective medium for biomarker acquisition for AIS. Although CSF is not routinely clinically obtained, a potential benefit of CSF studies is identifying valuable biomarkers from the pathophysiologic microenvironment that ultimately inform optimization of targeted low-abundance assays from peripheral biofluid samples (e.g., plasma). Several important catabolic and anabolic markers can serve as effective measures of diagnosis, etiology identification, prognostication, and severity grading. Trials with large cohorts studying the efficacy of biomarkers in altering clinical management are still needed.
Collapse
Affiliation(s)
- Anant Naik
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Champaign, IL 61820, USA
| | - Olufunmilola Adeleye
- Mayo Clinic Alix School of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Stefan W Koester
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Ethan A Winkler
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Joelle N Hartke
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Katherine Karahalios
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Sandra Mihaljevic
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Anupama Rani
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Sudhanshu Raikwar
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Jarrod D Rulney
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Shashvat M Desai
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Lea Scherschinski
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Andrew F Ducruet
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Felipe C Albuquerque
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Michael T Lawton
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Joshua S Catapano
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Ashutosh P Jadhav
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Ruchira M Jha
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA
| |
Collapse
|
6
|
Morphometric imaging biomarker identifies Alzheimer's disease even among mixed dementia patients. Sci Rep 2022; 12:17675. [PMID: 36319674 PMCID: PMC9626495 DOI: 10.1038/s41598-022-21796-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
A definitive diagnosis of Alzheimer's disease (AD), even in the presence of co-morbid neuropathology (occurring in > 50% of AD cases), is a significant unmet medical need that has obstructed the discovery of effective AD therapeutics. An AD-biomarker, the Morphometric Imaging (MI) assay on cultured skin fibroblasts, was used in a double-blind, allcomers (ages 55-90) trial of 3 patient cohorts: AD dementia patients, N = 25, all autopsy confirmed, non-AD dementia patients, N = 21-all autopsy or genetically confirmed; and non-demented control (AHC) patients N = 27. Fibroblasts cells isolated from 3-mm skin punch biopsies were cultured on a 3-D Matrigel matrix with movement dynamics quantified by image analysis. From counts of all aggregates (N) in a pre-defined field image and measures of the average area (A) of aggregates per image, the number-to-area ratios in a natural logarithmic form Ln(A/N) were determined for all patient samples. AD cell lines formed fewer large aggregates (cells clustered together) than non-AD or AHC cell lines. The cut-off value of Ln(A/N) = 6.98 was determined from the biomarker values of non-demented apparently healthy control (AHC) cases. Unequivocal validation by autopsy, genetics, and/or dementia criteria was possible for all 73 patient samples. The samples were collected from multiple centers-four US centers and one center in Japan. The study found no effect of center-to-center variation in fibroblast isolation, cell growth, or cell aggregation values (Ln(A/N)). The autopsy-confirmed MI Biomarker distinguished AD from non-AD dementia (non-ADD) patients and correctly diagnosed AD even in the presence of other co-morbid pathologies at autopsy (True Positive = 25, False Negative = 0, False Positive = 0, True Negative = 21, and Accuracy = 100%. Sensitivity and specificity were calculated as 100% (95% CI = 84 to 100.00%). From these findings, the MI assay appears to detect AD with great accuracy-even with abundant co-morbidity.
Collapse
|
7
|
Huang KL, Hsiao IT, Chang TY, Yang SY, Chang YJ, Wu HC, Liu CH, Wu YM, Lin KJ, Ho MY, Lee TH. Neurodegeneration and Vascular Burden on Cognition After Midlife: A Plasma and Neuroimaging Biomarker Study. Front Hum Neurosci 2022; 15:735063. [PMID: 34970128 PMCID: PMC8712753 DOI: 10.3389/fnhum.2021.735063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Objectives: Neurodegeneration and vascular burden are the two most common causes of post-stroke cognitive impairment. However, the interrelationship between the plasma beta-amyloid (Aβ) and tau protein, cortical atrophy and brain amyloid accumulation on PET imaging in stroke patients is undetermined. We aimed to explore: (1) the relationships of cortical thickness and amyloid burden on PET with plasma Aβ40, Aβ42, tau protein and their composite scores in stroke patients; and (2) the associations of post-stroke cognitive presentations with these plasma and neuroimaging biomarkers. Methods: The prospective project recruited first-ever ischemic stroke patients around 3 months after stroke onset. The plasma Aβ40, Aβ42, and total tau protein were measured with the immunomagnetic reduction method. Cortical thickness was evaluated on MRI, and cortical amyloid plaque deposition was evaluated by 18F-florbetapir PET. Cognition was evaluated with Mini-Mental State Examination (MMSE), Geriatric Depression Scale (GDS), Dementia Rating Scale-2 (DRS-2). Results: The study recruited 24 stroke patients and 13 normal controls. The plasma tau and tau*Aβ42 levels were correlated with mean cortical thickness after age adjustment. The Aβ42/Aβ40 ratio was correlated with global cortical 18F-florbetapir uptake value. The DRS-2 and GDS scores were associated with mean cortical thickness and plasma biomarkers, including Aβ42/Aβ40, tau, tau*Aβ42, tau/Aβ42, and tau/Aβ40 levels, in stroke patients. Conclusion: Plasma Aβ, tau, and their composite scores were associated with cognitive performance 3 months after stroke, and these plasma biomarkers were correlated with corresponding imaging biomarkers of neurodegeneration. Further longitudinal studies with a larger sample size are warranted to replicate the study results.
Collapse
Affiliation(s)
- Kuo-Lun Huang
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ing-Tsung Hsiao
- Department of Nuclear Medicine and Molecular Imaging Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Healthy Aging Research Center and Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Yu Chang
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | | | - Yeu-Jhy Chang
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsiu-Chuan Wu
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Hung Liu
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Ming Wu
- Department of Radiology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Molecular Imaging Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Healthy Aging Research Center and Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Meng-Yang Ho
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Behavioral Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Tsong-Hai Lee
- Department of Neurology, Linkou Chang Gung Memorial Hospital, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
8
|
Surinkaew P, Apaijai N, Sawaddiruk P, Jaiwongkam T, Kerdphoo S, Chattipakorn N, Chattipakorn SC. Mitochondrial Fusion Promoter Alleviates Brain Damage in Rats with Cardiac Ischemia/Reperfusion Injury. J Alzheimers Dis 2021; 77:993-1003. [PMID: 32804148 DOI: 10.3233/jad-200495] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cardiac ischemia/reperfusion (I/R) injury induces brain damage through increased blood-brain barrier (BBB) breakdown, microglial hyperactivity, pro-inflammatory cytokines, amyloid-β deposition, loss of dendritic spines, brain mitochondrial dysfunction, and imbalanced mitochondrial dynamics. Previous studies demonstrated that mitochondrial fusion promoter reduced cardiac damage from cardiac I/R injury; however, following cardiac I/R injury, the roles of mitochondrial dynamics on the brain have not been investigated. OBJECTIVE To investigate the effects of pharmacological modulation using mitochondrial fusion promoter (M1) in the brain of rats following cardiac I/R injury. METHODS Twenty-four male Wistar rats were separated into two groups; 1) sham-operation (n = 8) and 2) cardiac I/R injury (n = 16). Rats in the cardiac I/R injury group were randomly received either normal saline solution as a vehicle or a mitochondrial fusion promoter (M1, 2 mg/kg) intravenously. Both treatments were given to the rats 15 minutes before cardiac I/R injury. At the end of the reperfusion protocol, the brain was rapidly removed to investigate brain mitochondrial function, mitochondrial dynamics proteins, microglial activity, and Alzheimer's disease (AD) related proteins. RESULTS Cardiac I/R injury induced brain mitochondrial dynamics imbalance as indicated by reduced mitochondrial fusion proteins expression without alteration in mitochondrial fission, brain mitochondrial dysfunction, BBB breakdown, increased macrophage infiltration, apoptosis, and AD-related proteins. Pretreatment with M1 effectively increased the expression of mitofusin 2, a mitochondrial outer membrane fusion protein, reduced brain mitochondrial dysfunction, BBB breakdown, macrophage infiltration, apoptosis, and AD-related proteins in rats following cardiac I/R injury. CONCLUSION This mitochondrial fusion promoter significantly protected rats with cardiac I/R injury against brain damage.
Collapse
Affiliation(s)
- Poomarin Surinkaew
- Department of Anesthesiology, Lamphun Hospital, Lamphun, Thailand.,Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Passakorn Sawaddiruk
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thidarat Jaiwongkam
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sasiwan Kerdphoo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
9
|
Onatsu J, Vanninen R, JÄkÄlÄ P, Mustonen P, Pulkki K, Korhonen M, Hedman M, HÖglund K, Blennow K, Zetterberg H, Herukka SK, Taina M. Tau, S100B and NSE as Blood Biomarkers in Acute Cerebrovascular Events. In Vivo 2021; 34:2577-2586. [PMID: 32871787 DOI: 10.21873/invivo.12075] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM We aimed to analyze the diagnostic value of total tau (T-tau), S-100 calcium-binding protein B (S100B) and neuron-specific enolase (NSE) as blood-based biomarkers in acute ischemic stroke (AIS) or transient ischemic attack (TIA), and their correlation with symptom severity, infarct size, etiology and outcome. PATIENTS AND METHODS A total of 102 patients with stroke and 35 with TIA were analyzed. Subacute (63.8±50.1 h) plasma T-tau was measured with the single-molecule array (Simoa) method and NSE and S100B were evaluated for comparison. We evaluated biomarkers associations with: (i) diagnosis of AIS or TIA, (ii) cerebral infarction volume in the brain computed tomography, (iii) stroke etiology, (iv) clinical stroke severity and (iv) functional outcome after three months. RESULTS T-tau was higher in patients with stroke [1.0 pg/ml (IQR=0.3-2.2)] than with TIA [0.5 pg/ml (IQR=0.2-1.0), p=0.02]. The levels of S100B were also increased in stroke [0.082 μg/l (IQR=0.049-0.157)] patients compared to TIA patients [0.045 μg/l (IQR=0.03-0.073), p<0.001]. However, when the results were adjusted for confounders, significance was lost. Serum levels of NSE among patients with AIS [11.85 μg/l (IQR=9.30-16.14)] compared to those with TIA [10.96 μg/l (IQR=7.98-15.33), p=0.30] were equal. T-tau and S100B concentrations significantly correlated with cerebral infarction volume (r=0.412, p<0.001) and (r=0.597, p<0.001), also after corrections (p<0.001). mRS scores at three-month follow-up correlated with T-tau (r=0.248, p=0.016) and S100B concentrations (r=0.205, p=0.045). CONCLUSION For the diagnosis of TIA vs. AIS, blood T-tau and S100B concentrations discriminated only modestly. Additionally, groups were not separable after measuring of T-tau and S100B levels in the blood. T-tau and S100B concentrations correlated with the infarct size, but were not alone predictive for functional outcome at 3 months.
Collapse
Affiliation(s)
- Juha Onatsu
- Department of Neurology, NeuroCenter, Kuopio University Hospital, Kuopio, Finland .,Unit of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ritva Vanninen
- Department of Clinical Radiology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland.,Department of Clinical Radiology, University of Eastern Finland, Kuopio, Finland
| | - Pekka JÄkÄlÄ
- Department of Neurology, NeuroCenter, Kuopio University Hospital, Kuopio, Finland.,Unit of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Pirjo Mustonen
- Department of Cardiology, Keski-Suomi Central Hospital, Jyväskylä, Finland
| | - Kari Pulkki
- Department of Clinical Chemistry, University of Eastern Finland, Kuopio, Finland.,Eastern Finland Laboratory Centre, Kuopio, Finland
| | - Miika Korhonen
- Department of Clinical Radiology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland
| | - Marja Hedman
- Department of Clinical Radiology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland
| | - Kina HÖglund
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, U.K.,UK Dementia Research Institute, London, U.K
| | - Sanna-Kaisa Herukka
- Department of Neurology, NeuroCenter, Kuopio University Hospital, Kuopio, Finland.,Unit of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Mikko Taina
- Department of Clinical Radiology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland.,Department of Clinical Radiology, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
10
|
Mohamed SA, Herrmann K, Adlung A, Paschke N, Hausner L, FrÖlich L, Schad L, Groden C, Kerl HU. Evaluation of Sodium ( 23Na) MR-imaging as a Biomarker and Predictor for Neurodegenerative Changes in Patients With Alzheimer's Disease. In Vivo 2021; 35:429-435. [PMID: 33402493 DOI: 10.21873/invivo.12275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND/AIM Sodium (23Na) MR imaging is a noninvasive MRI technique that has been shown to be sensitive to visualize biochemical information about tissue viability, their cell integrity, and cell function in various studies. The aim of this study was to evaluate differences in regional brain 23Na signal intensity between Alzheimer's disease (AD) and healthy controls to preliminarily evaluate the capability of 23Na imaging as a biomarker for AD. PATIENTS AND METHODS A total of 14 patients diagnosed with AD were included: 12 in the state of dementia and 2 with mild cognitive impairment (MCI), and 12 healthy controls (HC); they were all scanned on a 3T clinical scanner with a double tuned 1H/23Na birdcage head coil. After normalizing the signal intensity with that of the vitreous humor, relative tissue sodium concentration (rTSC) was measured after automated segmentation in the hippocampus, amygdala, basal ganglia, white matter (WM) and grey matter (GM) in both cerebral hemispheres. RESULTS Patients with AD showed a significant increase in rTSC in comparison to healthy controls in the following brain regions: WM 13.6%; p=0.007, hippocampus 12.9%; p=0.003, amygdala 18.9%; p=0.0007. CONCLUSION 23Na-MRI has the potential to be developed as a useful biomarker for the diagnosis of AD.
Collapse
Affiliation(s)
- Sherif A Mohamed
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany;
| | - Katrin Herrmann
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anne Adlung
- Department of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nadia Paschke
- Department of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lucrezia Hausner
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lutz FrÖlich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lothar Schad
- Department of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Groden
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hans Ulrich Kerl
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
11
|
Siafaka PI, Özcan Bülbül E, Dilsiz P, Karantas ID, Okur ME, Üstündağ Okur N. Detecting and targeting neurodegenerative disorders using electrospun nanofibrous matrices: current status and applications. J Drug Target 2021; 29:476-490. [PMID: 33269637 DOI: 10.1080/1061186x.2020.1859516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neurodegeneration is defined as the progressive atrophy and loss of function of neurons; it is present in neurodegenerative disorders such as Multiple Sclerosis, Alzheimer's, Huntington's, and Parkinson's diseases. The detection of such disorders is performed by various imaging modalities while their therapeutic management is quite challenging. Besides, the pathogenesis of neurodegenerative disorders is still under ongoing research due to complex and multi-factorial mechanisms. Currently, targeting the specific proteins responsible for neurodegeneration is of great interest to many researchers. Furthermore, nanotechnology-based approaches for targeting the affected neurons became an emerging field of interest. Nanostructures of various forms have been developed aiming to act as therapeutics for neurodegeneration, in which electrospun nanofibers seem to play an important role as biomedical products for both detection and management of the diseases. Electrospinning is an intriguing method able to produce nanofibers with a wide range of sizes and morphological characteristics. Such nanofibrous matrices can be delivered through different administration routes to target various diseases. In this review, the most recent advancements in electrospun nanofibrous systems that target or detect multiple neurodegenerative diseases have been enlightened and an introduction to the general aspects of neurodegenerative diseases and the electrospinning process has been made. Finally, future perspectives of neurodegeneration targeting were also discussed.
Collapse
Affiliation(s)
- Panoraia I Siafaka
- School of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ece Özcan Bülbül
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istinye University, Istanbul, Turkey
| | - Pelin Dilsiz
- Department of Physiology, School of Medicine, Regenerative and Restorative Medical Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey.,Faculty of Pharmacy, Altınbaş University, Istanbul, Turkey
| | | | - Mehmet Evren Okur
- Department of Pharmacology, Faculty of Pharmacy, University of Health Sciences, Istanbul, Turkey
| | - Neslihan Üstündağ Okur
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
12
|
Ahmad S, Orellana A, Kohler I, Frölich L, de Rojas I, Gil S, Boada M, Hernández I, Hausner L, Bakker MHM, Cabrera-Socorro A, Amin N, Ramírez A, Ruiz A, Hankemeier T, Van Duijn CM. Association of lysophosphatidic acids with cerebrospinal fluid biomarkers and progression to Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2020; 12:124. [PMID: 33008436 PMCID: PMC7532619 DOI: 10.1186/s13195-020-00680-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/09/2020] [Indexed: 01/15/2023]
Abstract
Background Lysophosphatidic acids (LPAs) are bioactive signaling phospholipids that have been implicated in Alzheimer’s disease (AD). It is largely unknown whether LPAs are associated with AD pathology and progression from mild cognitive impairment (MCI) to AD. Methods The current study was performed on cerebrospinal fluid (CSF) and plasma samples of 182 MCI patients from two independent cohorts. We profiled LPA-derived metabolites using liquid chromatography-mass spectrometry. We evaluated the association of LPAs with CSF biomarkers of AD, Aβ-42, p-tau, and total tau levels overall and stratified by APOE genotype and with MCI to AD progression. Results Five LPAs (C16:0, C16:1, C22:4, C22:6, and isomer-LPA C22:5) showed significant positive association with CSF biomarkers of AD, Aβ-42, p-tau, and total tau, while LPA C14:0 and C20:1 associated only with Aβ-42 and alkyl-LPA C18:1, and LPA C20:1 associated with tau pathology biomarkers. Association of cyclic-LPA C16:0 and two LPAs (C20:4, C22:4) with Aβ-42 levels was found only in APOE ε4 carriers. Furthermore, LPA C16:0 and C16:1 also showed association with MCI to AD dementia progression, but results did not replicate in an independent cohort. Conclusions Our findings provide evidence that LPAs may contribute to early AD pathogenesis. Future studies are needed to determine whether LPAs play a role in upstream of AD pathology or are downstream markers of neurodegeneration.
Collapse
Affiliation(s)
- Shahzad Ahmad
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands.
| | - Adelina Orellana
- Research Center and Memory Clinic Fundació ACE, Institut Català de Neurociències, Aplicades. Universitat Internacional de Catalunya, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabelle Kohler
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
| | - Itziar de Rojas
- Research Center and Memory Clinic Fundació ACE, Institut Català de Neurociències, Aplicades. Universitat Internacional de Catalunya, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Gil
- Research Center and Memory Clinic Fundació ACE, Institut Català de Neurociències, Aplicades. Universitat Internacional de Catalunya, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic Fundació ACE, Institut Català de Neurociències, Aplicades. Universitat Internacional de Catalunya, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Hernández
- Research Center and Memory Clinic Fundació ACE, Institut Català de Neurociències, Aplicades. Universitat Internacional de Catalunya, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Lucrezia Hausner
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany.,Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany
| | - Margot H M Bakker
- Discovery Research, AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061, Ludwigshafen, Germany
| | | | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Alfredo Ramírez
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany.,Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
| | - Agustín Ruiz
- Research Center and Memory Clinic Fundació ACE, Institut Català de Neurociències, Aplicades. Universitat Internacional de Catalunya, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Thomas Hankemeier
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands. .,Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| | - Cornelia M Van Duijn
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands. .,Nuffield Department of Population Health, Oxford University, Oxford, UK.
| |
Collapse
|
13
|
Dindelegan CM, Faur D, Purza L, Bumbu A, Sabau M. Distress in neurocognitive disorders due to Alzheimer's disease and stroke. Exp Ther Med 2020; 20:2501-2509. [PMID: 32765742 DOI: 10.3892/etm.2020.8806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/06/2020] [Indexed: 12/25/2022] Open
Abstract
Patients with neurocognitive disorders experience subjectively the concept of quality of life; this is the reason why researchers avoid approaching this concept and prefer to focus attention on the emotional profile of the caregivers. Many studies highlight the efforts both emotional and financial made by caregivers in case of patients diagnosed with neurocognitive disorders. The present study shows the differences between the patients diagnosed with neurocognitive disorder due to Alzheimer's disease and patients diagnosed with stroke, as well as the Romanian norms for the short form of Geriatric Depression Scale. The study group consisted of the clinical population (N=45), selected according to the inclusion/exclusion criteria, following the principles of Helsinki Declaration for Ethical Medical Research. The study was conducted at the Neuropsychiatry section of the Municipal Clinical Hospital, Dr Gavril Curteanu, Oradea, Romania. The results showed significant differences between the two types of patients in terms of quality of life, t(43)=-7.99, P=0.001, affective distress, t(43)=5.10, P=0.001 and perceived stress, t(43)=3.81, P=0.001. The internal consistency of the scale is high, the coefficient KR-20 being 0.86.
Collapse
Affiliation(s)
- Camelia Maria Dindelegan
- Psychology Department, Faculty of Social Humanistic Science, University of Oradea, 410087 Oradea, Romania
| | - Darian Faur
- Psychology Department, Faculty of Social Humanistic Science, University of Oradea, 410087 Oradea, Romania
| | - Lavinia Purza
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Adrian Bumbu
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania
| | - Monica Sabau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania.,Clinical Department of Neurology, Emergency Clinical County Hospital, 410169 Oradea, Romania
| |
Collapse
|
14
|
Vinciguerra L, Lanza G, Puglisi V, Fisicaro F, Pennisi M, Bella R, Cantone M. Update on the Neurobiology of Vascular Cognitive Impairment: From Lab to Clinic. Int J Mol Sci 2020; 21:2977. [PMID: 32340195 PMCID: PMC7215552 DOI: 10.3390/ijms21082977] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023] Open
Abstract
In the last years, there has been a significant growth in the literature exploring the pathophysiology of vascular cognitive impairment (VCI). As an "umbrella term" encompassing any degree of vascular-related cognitive decline, VCI is deemed to be the most common cognitive disorder in the elderly, with a significant impact on social and healthcare expenses. Interestingly, some of the molecular, biochemical, and electrophysiological abnormalities detected in VCI seem to correlate with disease process and progression, eventually promoting an adaptive plasticity in some patients and a maladaptive, dysfunctional response in others. However, the exact relationships between vascular lesion, cognition, and neuroplasticity are not completely understood. Recent findings point out also the possibility to identify a panel of markers able to predict cognitive deterioration in the so-called "brain at risk" for vascular or mixed dementia. This will be of pivotal importance when designing trials of disease-modifying drugs or non-pharmacological approaches, including non-invasive neuromodulatory techniques. Taken together, these advances could make VCI a potentially preventable cause of both vascular and degenerative dementia in late life. This review provides a timely update on the recent serological, cerebrospinal fluid, histopathological, imaging, and neurophysiological studies on this "cutting-edge" topic, including the limitations, future perspectives and translational implications in the diagnosis and management of VCI patients.
Collapse
Affiliation(s)
- Luisa Vinciguerra
- Department of Neurology and Stroke Unit, ASST Cremona, 26100 Cremona, Italy; (L.V.); (V.P.)
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy
- Department of Neurology IC, Oasi Research Institute – IRCCS, 94018 Troina, Italy
| | - Valentina Puglisi
- Department of Neurology and Stroke Unit, ASST Cremona, 26100 Cremona, Italy; (L.V.); (V.P.)
| | - Francesco Fisicaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.F.); (M.P.)
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.F.); (M.P.)
| | - Rita Bella
- Department of Medical and Surgical Sciences and Advanced Technologies, University of Catania, 95123 Catania, Italy;
| | - Mariagiovanna Cantone
- Department of Neurology, Sant’Elia Hospital, ASP Caltanissetta, 93100 Caltanissetta, Italy;
| |
Collapse
|
15
|
Update on the Neurobiology of Vascular Cognitive Impairment: From Lab to Clinic. Int J Mol Sci 2020. [PMID: 32340195 DOI: 10.3390/ijms21082977.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In the last years, there has been a significant growth in the literature exploring the pathophysiology of vascular cognitive impairment (VCI). As an "umbrella term" encompassing any degree of vascular-related cognitive decline, VCI is deemed to be the most common cognitive disorder in the elderly, with a significant impact on social and healthcare expenses. Interestingly, some of the molecular, biochemical, and electrophysiological abnormalities detected in VCI seem to correlate with disease process and progression, eventually promoting an adaptive plasticity in some patients and a maladaptive, dysfunctional response in others. However, the exact relationships between vascular lesion, cognition, and neuroplasticity are not completely understood. Recent findings point out also the possibility to identify a panel of markers able to predict cognitive deterioration in the so-called "brain at risk" for vascular or mixed dementia. This will be of pivotal importance when designing trials of disease-modifying drugs or non-pharmacological approaches, including non-invasive neuromodulatory techniques. Taken together, these advances could make VCI a potentially preventable cause of both vascular and degenerative dementia in late life. This review provides a timely update on the recent serological, cerebrospinal fluid, histopathological, imaging, and neurophysiological studies on this "cutting-edge" topic, including the limitations, future perspectives and translational implications in the diagnosis and management of VCI patients.
Collapse
|
16
|
Bekrater-Bodmann R, Löffler A, Silvoni S, Frölich L, Hausner L, Desch S, Kleinböhl D, Flor H. Tablet-based sensorimotor home-training system for amnestic mild cognitive impairments in the elderly: design of a randomised clinical trial. BMJ Open 2019; 9:e028632. [PMID: 31377702 PMCID: PMC6687000 DOI: 10.1136/bmjopen-2018-028632] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 06/13/2019] [Accepted: 07/05/2019] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Dementia (particularly Alzheimer's disease, AD) is a major cause of impaired cognitive functions in the elderly. Amnestic mild cognitive impairment (aMCI) is a prodromal stage of AD, if substantiated by Alzheimer biomarkers. A neuroscientific model of pathological ageing emphasises the loss of brain plasticity, sensorimotor capacities and subsequent cognitive decline. A mechanistic treatment targeting dysfunctional plastic changes associated with ageing should be efficacious in delaying AD. In this trial, we aim to evaluate the effectiveness of a newly developed sensorimotor training, delivered at home, combined with personalised reinforcement, on the progression of aMCI-related cognitive impairments. METHODS AND ANALYSIS In a randomised trial, we will compare two aMCI groups (30 subjects each), randomly allocated to a sensorimotor or a cognitive control training. Both trainings consist of an adaptive algorithm, and will last 3 months each. We hypothesise that both trainings will have positive effects on cognitive function with the sensorimotor training being superior compared with the control training based on its improvement in basic perceptual skills underlying memory encoding and retrieval. The primary outcome is episodic memory function, improved hippocampal function during memory tasks will be a secondary outcome. As further exploratory outcomes, we expect improved segregation in sensory and motor maps, better sensory discrimination only in the sensorimotor training and reduced transition to dementia (examined after completion of this study). We expect the experimental training to be evaluated more positively by the users compared with the cognitive training, resulting in reduced rates of discontinuation. ETHICS AND DISSEMINATION The Ethics Committee of the Medical Faculty Mannheim, Heidelberg University, approved the study (2015-543N-MA), which adheres to the Declaration of Helsinki. The results will be published in a peer-reviewed journal. Access to raw data is available on request. TRIAL REGISTRATION NUMBER DRKS00012748.
Collapse
Affiliation(s)
- Robin Bekrater-Bodmann
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Annette Löffler
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefano Silvoni
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lucrezia Hausner
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Simon Desch
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dieter Kleinböhl
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Herta Flor
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Psychology, School of Social Sciences, Mannheim University, Mannheim, Germany
| |
Collapse
|
17
|
Sandelius Å, Cullen NC, Källén Å, Rosengren L, Jensen C, Kostanjevecki V, Vandijck M, Zetterberg H, Blennow K. Transient increase in CSF GAP-43 concentration after ischemic stroke. BMC Neurol 2018; 18:202. [PMID: 30526557 PMCID: PMC6284302 DOI: 10.1186/s12883-018-1210-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/29/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Cerebrospinal fluid (CSF) biomarkers reflect ongoing processes in the brain. Growth-associated protein 43 (GAP-43) is highly upregulated in brain tissue shortly after experimental ischemia suggesting the CSF GAP-43 concentration may be altered in ischemic brain disorders. CSF GAP-43 concentration is elevated in Alzheimer's disease patients; however, patients suffering from stroke have not been studied previously. METHODS The concentration of GAP-43 was measured in longitudinal CSF samples from 28 stroke patients prospectively collected on days 0-1, 2-4, 7-9, 3 weeks, and 3-5 months after ischemia and cross-sectionally in 19 controls. The stroke patients were clinically evaluated using a stroke severity score system. The extent of the brain lesion, including injury size and degrees of white matter lesions and atrophy were evaluated by CT and magnetic resonance imaging. RESULTS Increased GAP-43 concentration was detected from day 7-9 to 3 weeks after stroke, compared to day 1-4 and to levels in the control group (P = 0.02 and P = 0.007). At 3-5 months after stroke GAP-43 returned to admission levels. The initial increase in GAP-43 during the nine first days was associated to stroke severity, the degree of white matter lesions and atrophy and correlated positively with infarct size (rs = 0.65, P = 0.001). CONCLUSIONS The transient increase of CSF GAP-43 is important to take into account when used as a biomarker for other neurodegenerative diseases such as Alzheimer's disease. Furthermore, GAP-43 may be a marker of neuronal responses after stroke and additional studies confirming the potential of CSF GAP-43 to reflect severity and outcome of stroke in larger cohorts are warranted.
Collapse
Affiliation(s)
- Åsa Sandelius
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden. .,Department of Psychiatry and Neurochemistry, Sahlgrenska University Hospital/Mölndal, S-431 80, Mölndal, Sweden.
| | - Nicholas C Cullen
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Åsa Källén
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Lars Rosengren
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience and Rehabilitation, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Crister Jensen
- Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute, WC1N, London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden. .,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden. .,Department of Psychiatry and Neurochemistry, Sahlgrenska University Hospital/Mölndal, S-431 80, Mölndal, Sweden.
| |
Collapse
|
18
|
Visualization of ischemic stroke-related changes on 18F-THK-5351 positron emission tomography. EJNMMI Res 2018; 8:62. [PMID: 30014313 PMCID: PMC6047954 DOI: 10.1186/s13550-018-0417-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022] Open
Abstract
Background The 18F-THK-5351 radiotracer has been used to detect the in vivo tau protein distribution in patients with tauopathy, such as Alzheimer’s disease and corticobasal syndrome. In addition, 18F-THK-5351 can also monitor neuroinflammatory process due to high affinity to astrogliosis. We aimed to explore 18F-THK-5351 distribution patterns and characteristics in patients with recent ischemic stroke. Results Fifteen patients received 18F-THK-5351 positron emission tomography (PET) and diffusion tensor imaging (DTI) approximately 3 months after ischemic stroke. A region of interest (ROI) was placed in the peri-ischemic area and was mirrored on the contralateral side as the control, and a proportional value was derived from the ratio of the peri-ischemic ROI value over the mirrored ROI value. Increased 18F-THK-5351 retention was observed in the areas around and remote from the stroke location. The proportional 18F-THK-5351 values were negatively correlated with the proportional fractional anisotropy values (r = − 0.39, P = 0.04). Conclusion 18F-THK-5351 PET imaging provides a potential tool for in vivo visualization of the widespread ischemia-related changes associated with a microstructural disruption in recent ischemic stroke patients. Electronic supplementary material The online version of this article (10.1186/s13550-018-0417-1) contains supplementary material, which is available to authorized users.
Collapse
|
19
|
Osier N, Motamedi V, Edwards K, Puccio A, Diaz-Arrastia R, Kenney K, Gill J. Exosomes in Acquired Neurological Disorders: New Insights into Pathophysiology and Treatment. Mol Neurobiol 2018; 55:9280-9293. [PMID: 29663285 DOI: 10.1007/s12035-018-1054-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 03/29/2018] [Indexed: 01/08/2023]
Abstract
Exosomes are endogenous nanovesicles that play critical roles in intercellular signaling by conveying functional genetic information and proteins between cells. Exosomes readily cross the blood-brain barrier and have promise as therapeutic delivery vehicles that have the potential to specifically deliver molecules to the central nervous system (CNS). This unique feature also makes exosomes attractive as biomarkers in diagnostics, prognostics, and therapeutics in the context of multiple significant public health conditions, including acquired neurological disorders. The purpose of this review is to summarize the state of the science surrounding the relevance of extracellular vesicles (EVs), particularly exosomes, to acquire neurological disorders, specifically traumatic brain injury (TBI), spinal cord injury (SCI), and ischemic stroke. In total, ten research articles were identified that examined exosomes in the context of TBI, SCI, or stroke; these manuscripts were reviewed and synthesized to further understand the current role of exosomes in the context of acquired neurological disorders. Of the ten published studies, four focused exclusively on TBI, one on both TBI and SCI, and five on ischemic stroke; notably, eight of the ten studies were limited to pre-clinical samples. The present review is the first to discuss the current body of knowledge surrounding the role of exosomes in the pathophysiology, diagnosis, and prognosis, as well as promising therapeutic strategies in TBI, SCI, and stroke research.
Collapse
Affiliation(s)
- Nicole Osier
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA. .,University of Texas at Austin, Austin, TX, USA.
| | - Vida Motamedi
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA
| | - Katie Edwards
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA.,Healthcare Genetics Doctoral Program, Clemson University School of Nursing, 508 Edwards, Clemson, SC, 29631, USA
| | - Ava Puccio
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite B-400, Pittsburgh, PA, 15213, USA
| | - Ramon Diaz-Arrastia
- University of Pennsylvania School of Medicine, Suite 205 Medical Office Building, 51 N 39TH ST, Philadelphia, PA, 19104, USA
| | - Kimbra Kenney
- National Intrepid Center of Excellence, Walter Reed National Military Medical Center, Building 51, Room 2306, 4860 South Palmer Road, Bethesda, MD, 20889-5649, USA
| | - Jessica Gill
- National Institutes of Health, National Institute of Nursing Research, 1 Cloister Ct, Bethesda, MD, 20814, USA
| |
Collapse
|
20
|
Abstract
Diseases of the central nervous system that are caused by an underlying vascular pathology typically result in either hemorrhage or ischemia. Most prominent entities include spontaneous subarachnoid hemorrhage, spontaneous intracerebral hemorrhage, and ischemic stroke. For anatomic reasons, cerebrospinal fluid (CSF) qualifies as body fluid for the exploration of biomarkers in these disorders. Even though in subarachnoid hemorrhage a few CSF parameters have been established for routine diagnostic purposes, there is still an unmet need and broad interest in the identification of molecules that would allow further insight into disease mechanisms and supplement patients' medical care. This chapter provides an overview on what is presently known about CSF biomarkers in spontaneous subarachnoid hemorrhage, spontaneous intracerebral hemorrhage, and ischemic stroke. We recapitulate current evidence on established diagnostic tests, discuss the role of various CSF molecules in the pathophysiology of these diseases, and illuminate their potential use in future clinical practice. Furthermore, we address methodologic aspects as well as shortcomings of research in this field.
Collapse
Affiliation(s)
- Harald Hegen
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Michael Auer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
21
|
Wallin A, Román GC, Esiri M, Kettunen P, Svensson J, Paraskevas GP, Kapaki E. Update on Vascular Cognitive Impairment Associated with Subcortical Small-Vessel Disease. J Alzheimers Dis 2018; 62:1417-1441. [PMID: 29562536 PMCID: PMC5870030 DOI: 10.3233/jad-170803] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Subcortical small-vessel disease (SSVD) is a disorder well characterized from the clinical, imaging, and neuropathological viewpoints. SSVD is considered the most prevalent ischemic brain disorder, increasing in frequency with age. Vascular risk factors include hypertension, diabetes, hyperlipidemia, elevated homocysteine, and obstructive sleep apnea. Ischemic white matter lesions are the hallmark of SSVD; other pathological lesions include arteriolosclerosis, dilatation of perivascular spaces, venous collagenosis, cerebral amyloid angiopathy, microbleeds, microinfarcts, lacunes, and large infarcts. The pathogenesis of SSVD is incompletely understood but includes endothelial changes and blood-brain barrier alterations involving metalloproteinases, vascular endothelial growth factors, angiotensin II, mindin/spondin, and the mammalian target of rapamycin pathway. Metabolic and genetic conditions may also play a role but hitherto there are few conclusive studies. Clinical diagnosis of SSVD includes early executive dysfunction manifested by impaired capacity to use complex information, to formulate strategies, and to exercise self-control. In comparison with Alzheimer's disease (AD), patients with SSVD show less pronounced episodic memory deficits. Brain imaging has advanced substantially the diagnostic tools for SSVD. With the exception of cortical microinfarcts, all other lesions are well visualized with MRI. Diagnostic biomarkers that separate AD from SSVD include reduction of cerebrospinal fluid amyloid-β (Aβ)42 and of the ratio Aβ42/Aβ40 often with increased total tau levels. However, better markers of small-vessel function of intracerebral blood vessels are needed. The treatment of SSVD remains unsatisfactory other than control of vascular risk factors. There is an urgent need of finding targets to slow down and potentially halt the progression of this prevalent, but often unrecognized, disorder.
Collapse
Affiliation(s)
- Anders Wallin
- Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg Sweden and Memory Clinic at Department of Neuropsychiatry, Sahlgrenska University, Hospital, Gothenburg, Sweden
| | - Gustavo C. Román
- Department of Neurology, Methodist Neurological Institute, Houston, TX, USA
- Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Margaret Esiri
- Neuropathology Department, West Wing, John Radcliffe Hospital, Oxford, UK
| | - Petronella Kettunen
- Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg Sweden and Memory Clinic at Department of Neuropsychiatry, Sahlgrenska University, Hospital, Gothenburg, Sweden
- Nuffield Department of Clinical Neurosciences, University of Oxford, West Wing, John Radcliffe Hospital, Oxford, UK
| | - Johan Svensson
- Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - George P. Paraskevas
- 1st Department of Neurology, Neurochemistry Unit, National and Kapodistrian University of Athens, Athens, Greece
| | - Elisabeth Kapaki
- 1st Department of Neurology, Neurochemistry Unit, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
22
|
Radanovic M, Stella F, Silva LG, Talib LL, Forlenza OV. Increased CSF levels of total Tau in patients with subcortical cerebrovascular pathology and cognitive impairment. Dement Neuropsychol 2017; 11:419-425. [PMID: 29354223 PMCID: PMC5770001 DOI: 10.1590/1980-57642016dn11-040012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/07/2017] [Indexed: 11/22/2022] Open
Abstract
Cognitive impairment includes mild cognitive decline and dementia, such as Alzheimer's disease (AD) and cerebrovascular-related pathologies. OBJECTIVE To investigate the profile of AD-related CSF biomarkers in a sample of cognitively impaired and unimpaired older adults with concomitant subcortical cerebrovascular burden. METHODS Seventy-eight older adults attending an outpatient psychogeriatric clinic were enrolled. Diagnoses were based on clinical, neuropsychological, laboratory, and neuroimaging data. Participants were classified into: cognitively normal (controls, n = 30), mild cognitive impairment (MCI, n = 34), and dementia (AD, n = 14). All subjects were submitted to CSF analyses for determination of amyloid-beta (Aβ1-42), total tau (t-tau), phosphorylated tau (p-tau) and Aβ1-42/p-tau ratio according to the Luminex method. MRI was performed in all individuals, and was scored independently by two experts according to Fazekas scale. Statistical analyses were conducted with the aid of general linear model procedures, and the Chi-squared test. RESULTS T-tau levels were significantly associated with subcortical lesion pattern when Fazekas was considered as a group factor. CSF biomarkers were not associated with MCI, AD, or controls when considered separately. There was a tendency for reduction in CSF Aβ1-42 together with increasing Fazekas scores, but without statistical significance. Comparisons of Aβ1-42 and t-tau with each clinical group or with each neuroimaging pattern did not reach statistical differences. Likewise, Fazekas scores had no impact on CAMCOG scores. CONCLUSION We found a significant association between t-tau levels and subcortical lesions when all Fazekas classifications were considered as a single group; comparisons of Fazekas subgroups and CSF biomarkers did not reach significance.
Collapse
Affiliation(s)
- Márcia Radanovic
- Laboratório de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Florindo Stella
- Laboratório de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
- Biosciences Institute, Universidade Estadual Paulista (UNESP), Campus of Rio Claro, SP, Brazil
| | - Lis Gomes Silva
- Laboratório de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Leda L. Talib
- Laboratório de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Orestes V. Forlenza
- Laboratório de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
23
|
Llorens F, Schmitz M, Knipper T, Schmidt C, Lange P, Fischer A, Hermann P, Zerr I. Cerebrospinal Fluid Biomarkers of Alzheimer's Disease Show Different but Partially Overlapping Profile Compared to Vascular Dementia. Front Aging Neurosci 2017; 9:289. [PMID: 28955218 PMCID: PMC5601075 DOI: 10.3389/fnagi.2017.00289] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/18/2017] [Indexed: 11/13/2022] Open
Abstract
Vascular factors increase the risks of developing Alzheimer's disease (AD) and they contribute to AD pathology. Since amyloid beta (Aβ) deposits can be observed in both diseases, there is an overlap which impedes a clear discrimination and difficult clinical diagnosis. In the present study, we compared cerebrospinal fluid (CSF) profiles of neurodegenerative and inflammatory biomarkers in a patient cohort of controls (n = 50), AD (n = 65) and vascular dementia (VaD) (n = 31) cases. Main results were validated in a second cohort composed of AD (n = 26), rapidly progressive AD (rpAD) (n = 15), VaD (n = 21), and cognitively unimpaired patients with vascular encephalopathy (VE) (n = 25) cases. In the study, cohort significant differences were detected in tau, p-tau, and Aβ1-42 (Aβ42) levels between AD and VaD patients, but not for the neuron-specific enolase (NSE), S100B protein, 14-3-3 and YKL-40. Differential tau, p-tau, and Aβ42 levels between AD and VaD were confirmed in the validation cohort, which additionally showed no differences between AD and rpAD, nor between VaD and VE. The evaluation of the biomarker performance in discrimination between AD and VaD patients revealed that the best diagnostic accuracy could be obtained when tau, p-tau, and Aβ42 were combined in form of Aβ42/p-tau (AUC 0.84-0.90, sensitivity 77-81%, specificity 80-93%) and (tau × p-tau)/Aβ42 ratio (AUC 0.83-0.87, sensitivity 73-81%, specificity 78-87%). Altogether, our studies provided neurodegenerative biomarker profiles in two cohorts of AD and VaD patients favoring the combination of CSF biomarker to differentiate between diseases.
Collapse
Affiliation(s)
- Franc Llorens
- Department of Neurology, Universitätsmedizin GöttingenGöttingen, Germany
- Center for Networked Biomedical Research on Neurodegenerative DiseasesBarcelona, Spain
| | - Matthias Schmitz
- Department of Neurology, Universitätsmedizin GöttingenGöttingen, Germany
- German Center for Neurodegenerative Diseases–DZNE Site GöttingenBonn, Germany
| | - Tobias Knipper
- Department of Neurology, Universitätsmedizin GöttingenGöttingen, Germany
| | - Christian Schmidt
- Department of Neurology, Universitätsmedizin GöttingenGöttingen, Germany
| | - Peter Lange
- Department of Neurology, Universitätsmedizin GöttingenGöttingen, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases–DZNE Site GöttingenBonn, Germany
| | - Peter Hermann
- Department of Neurology, Universitätsmedizin GöttingenGöttingen, Germany
- German Center for Neurodegenerative Diseases–DZNE Site GöttingenBonn, Germany
| | - Inga Zerr
- Department of Neurology, Universitätsmedizin GöttingenGöttingen, Germany
- German Center for Neurodegenerative Diseases–DZNE Site GöttingenBonn, Germany
| |
Collapse
|
24
|
Tariq S, Barber PA. Dementia risk and prevention by targeting modifiable vascular risk factors. J Neurochem 2017; 144:565-581. [PMID: 28734089 DOI: 10.1111/jnc.14132] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/23/2017] [Accepted: 07/15/2017] [Indexed: 01/04/2023]
Abstract
The incidence of dementia is expected to double in the next 20 years and will contribute to heavy social and economic burden. Dementia is caused by neuronal loss that leads to brain atrophy years before symptoms manifest. Currently, no cure exists and extensive efforts are being made to mitigate cognitive impairment in late life in order to reduce the burden on patients, caregivers, and society. The most common type of dementia, Alzheimer's disease (AD), and vascular dementia (VaD) often co-exists in the brain and shares common, modifiable risk factors, which are targeted in numerous secondary prevention trials. There is a growing need for non-pharmacological interventions and infrastructural support from governments to encourage psychosocial and behavioral interventions. Secondary prevention trials need to be redesigned based on the risk profile of individual subjects, which require the use of validated and standardized clinical, biological, and neuroimaging biomarkers. Multi-domain approaches have been proposed in high-risk populations that target optimal treatment; clinical trials need to recruit individuals at the highest risk of dementia before symptoms develop, thereby identifying an enriched disease group to test preventative and disease modifying strategies. The underlying aim should be to reduce microscopic brain tissue loss by modifying vascular and lifestyle risk factors over a relatively short period of time, thus optimizing the opportunity for preventing dementia in the future. Collaboration between international research groups is of key importance to the optimal use and allocation of existing resources, and the development of new techniques in preventing dementia. This article is part of the Special Issue "Vascular Dementia".
Collapse
Affiliation(s)
- Sana Tariq
- Seaman Family MR Center, Foothills Medical Centre, Calgary, AB, Canada.,Hotchkiss Brain Institute, Foothills Medical Center, Room 1A10 Health Research Innovation Center, Calgary, AB, Canada
| | - Philip A Barber
- Hotchkiss Brain Institute, Foothills Medical Center, Room 1A10 Health Research Innovation Center, Calgary, AB, Canada.,Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada
| |
Collapse
|
25
|
De Vos A, Bjerke M, Brouns R, De Roeck N, Jacobs D, Van den Abbeele L, Guldolf K, Zetterberg H, Blennow K, Engelborghs S, Vanmechelen E. Neurogranin and tau in cerebrospinal fluid and plasma of patients with acute ischemic stroke. BMC Neurol 2017; 17:170. [PMID: 28854881 PMCID: PMC5577791 DOI: 10.1186/s12883-017-0945-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 08/21/2017] [Indexed: 12/19/2022] Open
Abstract
Background While neurogranin has no value as plasma biomarker for Alzheimer’s disease, it may be a potential blood biomarker for traumatic brain injury. This evokes the question whether there are changes in neurogranin levels in blood in other conditions of brain injury, such as acute ischemic stroke (AIS). Methods We therefore explored neurogranin in paired cerebrospinal fluid (CSF)/plasma samples of AIS patients (n = 50) from a well-described prospective study. In parallel, we investigated another neuronal protein, i.e. tau, which has already been suggested as potential AIS biomarker in CSF and blood. ELISA as well as Single Molecule Array (Simoa) technology were used for the biochemical analyses. Statistical analyses included Shapiro-Wilk testing, Mann-Whitney analyses and Pearson’s correlation analysis. Results In contrast to tau, of which high levels in both CSF and plasma were related to stroke characteristics like severity and long-term outcome, plasma neurogranin levels were only correlated with infarct volume. Likewise, CSF neurogranin levels were significantly higher in patients with an infarct volume > 5 mL than in patients with smaller infarct volumes. Finally, neurogranin and tau were significantly correlated in CSF, whereas a weaker relationship was observed in plasma. Conclusions These findings indicate that although plasma and CSF neurogranin may reflect the volume of acute cerebral ischemia, this synaptic protein is less likely to be a potential AIS biomarker. Levels of tau correlated with severity and outcome of stroke in both plasma and CSF, in the present study as well as previous reports, confirming the potential of tau as an AIS biomarker. Electronic supplementary material The online version of this article (10.1186/s12883-017-0945-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ann De Vos
- ADx NeuroSciences NV, Technologiepark 4, 9052, Ghent, Belgium
| | - Maria Bjerke
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Raf Brouns
- Department of Neurology, Hospital ZorSaam, Terneuzen, The Netherlands
| | - Naomi De Roeck
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Dirk Jacobs
- ADx NeuroSciences NV, Technologiepark 4, 9052, Ghent, Belgium
| | | | - Kaat Guldolf
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | | |
Collapse
|
26
|
Yang SY, Chiu MJ, Chen TF, Lin CH, Jeng JS, Tang SC, Lee YF, Yang CC, Liu BH, Chen HH, Wu CC. Analytical performance of reagent for assaying tau protein in human plasma and feasibility study screening neurodegenerative diseases. Sci Rep 2017; 7:9304. [PMID: 28839167 PMCID: PMC5571227 DOI: 10.1038/s41598-017-09009-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/19/2017] [Indexed: 12/22/2022] Open
Abstract
Immunomagnetic reduction (IMR), which involves the use of antibody-functionalized magnetic nanoparticles to specifically label target biomarkers, was utilized to develop an assay for total tau protein in human plasma. The analytic properties of the IMR assay on tau protein were investigated. The limit of detection was found to be 0.026 pg/ml. Other properties such as Hook effect, assay linearity, dilution recovery range, reagent stability, interference test, and spiked recovery were also characterized. The ultra-sensitive IMR assay was applied to detect the plasma tau protein levels of subjects with prevalent neurodegenerative diseases, such as Alzheimer's disease (AD), mild cognitive impairment (MCI) due to AD, Parkinson's disease (PD), frontotemporal dementia (FTD) and vascular dementia (VD). The concentrations of plasma tau protein in patients with VD, PD, MCI due to AD, FTD, and AD patients were higher than that of healthy controls. Using an ROC curve analysis, the cutoff value for discriminating dementia patients from healthy controls was 17.43 pg/ml, resulting in 0.856 and 0.727 for clinical sensitivity and specificity, respectively. The area under the ROC curve was 0.908. These results imply that the IMR plasma tau assay would be useful to screen for prevalent neurodegenerative diseases.
Collapse
Affiliation(s)
- Shieh-Yueh Yang
- MagQu Co., Ltd., Xindian District, New Taipei City, 231, Taiwan.
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
- Department of Psychology, National Taiwan University, Taipei, 100, Taiwan
- Graduate Institute of Biomedical Engineering and Bioinformatics, National Taiwan University, Taipei, 116, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Jiann-Shing Jeng
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Sung-Chun Tang
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Yen-Fu Lee
- MagQu Co., Ltd., Xindian District, New Taipei City, 231, Taiwan
| | - Che-Chuan Yang
- MagQu Co., Ltd., Xindian District, New Taipei City, 231, Taiwan
| | - Bing-Hsien Liu
- MagQu Co., Ltd., Xindian District, New Taipei City, 231, Taiwan
| | - Hsin-Hsien Chen
- MagQu Co., Ltd., Xindian District, New Taipei City, 231, Taiwan
| | - Chau-Chung Wu
- Departments of Internal Medicine and Primary Care Medicine, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| |
Collapse
|
27
|
Yang SY, Chiu MJ, Chen TF, Horng HE. Detection of Plasma Biomarkers Using Immunomagnetic Reduction: A Promising Method for the Early Diagnosis of Alzheimer's Disease. Neurol Ther 2017; 6:37-56. [PMID: 28733955 PMCID: PMC5520821 DOI: 10.1007/s40120-017-0075-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. The development of assay technologies able to diagnose early-stage AD is important. Blood tests to detect biomarkers, such as amyloid and total Tau protein, are among the most promising diagnostic methods due to their low cost, low risk, and ease of operation. However, such biomarkers in blood occur at extremely low levels and are difficult to detect precisely. In the early 2000s, a highly sensitive assay technology, immunomagnetic reduction (IMR), was developed. IMR involves the use of antibody-functionalized magnetic nanoparticles dispersed in aqueous solution. The concentrations of detected molecules are converted to reductions in the ac magnetic susceptibility of this reagent due to the association between the magnetic nanoparticles and molecules. To achieve ultra-high sensitivity, a high-Tc superconducting-quantum-interference-device (SQUID) ac magnetosusceptometer was designed and applied to detect the tiny reduction in the ac magnetic susceptibility of the reagent. Currently, a 36-channeled high-Tc SQUID-based ac magnetosusceptometer is available. Using the reagent and this analyzer, extremely low concentrations of amyloid and total Tau protein in human plasma could be detected. Further, the feasibility of identifying subjects in early-stage AD via assaying plasma amyloid and total Tau protein is demonstrated. The results show a diagnostic accuracy for prodromal AD higher than 80% and reveal the possibility of screening for early-stage AD using SQUID-based IMR.
Collapse
Affiliation(s)
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 110, Taiwan.,Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 110, Taiwan.,Department of Psychology, National Taiwan University, Taipei, 110, Taiwan.,Graduate Institute of Biomedical Engineering and Bioinformatics, National Taiwan University, Taipei, 116, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 110, Taiwan
| | - Herng-Er Horng
- Institute of Electro-Optical Science and Technology, National Taiwan Normal University, Taipei, 116, Taiwan
| |
Collapse
|
28
|
Wallin A, Kapaki E, Boban M, Engelborghs S, Hermann DM, Huisa B, Jonsson M, Kramberger MG, Lossi L, Malojcic B, Mehrabian S, Merighi A, Mukaetova-Ladinska EB, Paraskevas GP, Popescu BO, Ravid R, Traykov L, Tsivgoulis G, Weinstein G, Korczyn A, Bjerke M, Rosenberg G. Biochemical markers in vascular cognitive impairment associated with subcortical small vessel disease - A consensus report. BMC Neurol 2017; 17:102. [PMID: 28535786 PMCID: PMC5442599 DOI: 10.1186/s12883-017-0877-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 05/09/2017] [Indexed: 12/17/2022] Open
Abstract
Background Vascular cognitive impairment (VCI) is a heterogeneous entity with multiple aetiologies, all linked to underlying vascular disease. Among these, VCI related to subcortical small vessel disease (SSVD) is emerging as a major homogeneous subtype. Its progressive course raises the need for biomarker identification and/or development for adequate therapeutic interventions to be tested. In order to shed light in the current status on biochemical markers for VCI-SSVD, experts in field reviewed the recent evidence and literature data. Method The group conducted a comprehensive search on Medline, PubMed and Embase databases for studies published until 15.01.2017. The proposal on current status of biochemical markers in VCI-SSVD was reviewed by all co-authors and the draft was repeatedly circulated and discussed before it was finalized. Results This review identifies a large number of biochemical markers derived from CSF and blood. There is a considerable overlap of VCI-SSVD clinical symptoms with those of Alzheimer’s disease (AD). Although most of the published studies are small and their findings remain to be replicated in larger cohorts, several biomarkers have shown promise in separating VCI-SSVD from AD. These promising biomarkers are closely linked to underlying SSVD pathophysiology, namely disruption of blood-CSF and blood–brain barriers (BCB-BBB) and breakdown of white matter myelinated fibres and extracellular matrix, as well as blood and brain inflammation. The leading biomarker candidates are: elevated CSF/blood albumin ratio, which reflects BCB/BBB disruption; altered CSF matrix metalloproteinases, reflecting extracellular matrix breakdown; CSF neurofilment as a marker of axonal damage, and possibly blood inflammatory cytokines and adhesion molecules. The suggested SSVD biomarker deviations contrasts the characteristic CSF profile in AD, i.e. depletion of amyloid beta peptide and increased phosphorylated and total tau. Conclusions Combining SSVD and AD biomarkers may provide a powerful tool to identify with greater precision appropriate patients for clinical trials of more homogeneous dementia populations. Thereby, biomarkers might promote therapeutic progress not only in VCI-SSVD, but also in AD.
Collapse
Affiliation(s)
- A Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden. .,Memory Clinic at Department of Neuropsychiatry, Sahlgrenska University Hospital, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Wallinsgatan 6, SE-431 41, Mölndal, Sweden.
| | - E Kapaki
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - M Boban
- Department of Neurology, University Hospital Centre Zagreb, Medical School, University of Zagreb, Zagreb, Croatia
| | - S Engelborghs
- Memory Clinic and Department of Neurology, Hospital Network Antwerp (ZNA) Middelheim and HogeBeuken, Antwerp, Belgium.,Reference Center for Biological Markers of Dementia, Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - D M Hermann
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - B Huisa
- Department of Neurology, University of California, Irvine, California, USA
| | - M Jonsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - M G Kramberger
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - L Lossi
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - B Malojcic
- Department of Neurology, University Hospital Centre Zagreb, Medical School, University of Zagreb, Zagreb, Croatia
| | - S Mehrabian
- Department of Neurology, University Hospital "Alexandrovska", Medical University, Sofia, Bulgaria
| | - A Merighi
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - E B Mukaetova-Ladinska
- Institute of Neuroscience, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - G P Paraskevas
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - B O Popescu
- Department of Neurology, Colentina Clinical Hospital, School of Medicine, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania
| | - R Ravid
- Brain Bank Consultants, Amsterdam, The Netherlands
| | - L Traykov
- Department of Neurology, University Hospital "Alexandrovska", Medical University, Sofia, Bulgaria
| | - G Tsivgoulis
- 2nd Department of Neurology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - G Weinstein
- School of Public Health, University of Haifa, Haifa, Israel
| | - A Korczyn
- Department of Neurology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - M Bjerke
- Reference Center for Biological Markers of Dementia, Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - G Rosenberg
- University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| |
Collapse
|
29
|
Llorens F, Kruse N, Karch A, Schmitz M, Zafar S, Gotzmann N, Sun T, Köchy S, Knipper T, Cramm M, Golanska E, Sikorska B, Liberski PP, Sánchez-Valle R, Fischer A, Mollenhauer B, Zerr I. Validation of α-Synuclein as a CSF Biomarker for Sporadic Creutzfeldt-Jakob Disease. Mol Neurobiol 2017; 55:2249-2257. [PMID: 28321768 PMCID: PMC5840235 DOI: 10.1007/s12035-017-0479-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/02/2017] [Indexed: 01/15/2023]
Abstract
The analysis of cerebrospinal fluid (CSF) biomarkers gains importance in the differential diagnosis of prion diseases. However, no single diagnostic tool or combination of them can unequivocally confirm prion disease diagnosis. Electrochemiluminescence (ECL)-based immunoassays have demonstrated to achieve high diagnostic accuracy in a variety of sample types due to their high sensitivity and dynamic range. Quantification of CSF α-synuclein (a-syn) by an in-house ECL-based ELISA assay has been recently reported as an excellent approach for the diagnosis of sporadic Creutzfeldt-Jakob disease (sCJD), the most prevalent form of human prion disease. In the present study, we validated a commercially available ECL-based a-syn ELISA platform as a diagnostic test for correct classification of sCJD cases. CSF a-syn was analysed in 203 sCJD cases with definite diagnosis and in 445 non-CJD cases. We investigated reproducibility and stability of CSF a-syn and made recommendations for its analysis in the sCJD diagnostic workup. A sensitivity of 98% and a specificity of 97% were achieved when using an optimal cut-off of 820 pg/mL a-syn. Moreover, we were able to show a negative correlation between a-syn levels and disease duration suggesting that CSF a-syn may be a good prognostic marker for sCJD patients. The present study validates the use of a-syn as a CSF biomarker of sCJD and establishes the clinical and pre-analytical parameters for its use in differential diagnosis in clinical routine. Additionally, the current test presents some advantages compared to other diagnostic approaches: it is fast, economic, requires minimal amount of CSF and a-syn levels are stable along disease progression.
Collapse
Affiliation(s)
- Franc Llorens
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany. .,German Center for Neurodegenerative Diseases (DZNE), Site Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany.
| | - Niels Kruse
- Institute for Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - André Karch
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Matthias Schmitz
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany
| | - Saima Zafar
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany
| | - Nadine Gotzmann
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany
| | - Ting Sun
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany
| | - Silja Köchy
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany
| | - Tobias Knipper
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany
| | - Maria Cramm
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany
| | - Ewa Golanska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Lodz, Poland
| | - Beata Sikorska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Lodz, Poland
| | - Pawel P Liberski
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Lodz, Poland
| | - Raquel Sánchez-Valle
- Creutzfeldt-Jakob disease unit. Alzheimer's disease and other cognitive disorders unit. Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Andre Fischer
- German Center for Neurodegenerative Diseases (DZNE), Site Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany
| | - Brit Mollenhauer
- Institute for Neuropathology, University Medical Center Göttingen, Göttingen, Germany.,Paracelsus-Elena Klinik, Center for Parkinsonism and Movement Disorders, Kassel, Germany.,Department of Neurosurgery, University Medical Center Göttingen, Göttingen, Germany
| | - Inga Zerr
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Göttingen, Robert Koch Stasse 40, 37075, Göttingen, Germany
| |
Collapse
|
30
|
Somers C, Goossens J, Engelborghs S, Bjerke M. Selecting Aβ isoforms for an Alzheimer's disease cerebrospinal fluid biomarker panel. Biomark Med 2017; 11:169-178. [PMID: 28111962 DOI: 10.2217/bmm-2016-0276] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although the core cerebrospinal fluid Alzheimer's disease (AD) biomarkers amyloid-β (Aβ1-42) and tau show a high diagnostic accuracy, there are still limitations due to overlap in the biomarker levels with other neurodegenerative and dementia disorders. During Aβ1-42 production and clearance in the brain, several other Aβ peptides and amyloid precursor protein fragments are formed that could potentially serve as biomarkers for this ongoing disease process. Therefore, this review will present the current status of the findings for amyloid precursor protein and Aβ peptide isoforms in AD and clinically related disorders. In conclusion, adding new Aβ isoforms to the AD biomarker panel may improve early differential diagnostic accuracy and increase the cerebrospinal fluid biomarker concordance with AD neuropathological findings in the brain.
Collapse
Affiliation(s)
- Charisse Somers
- Department of Biomedical Sciences, Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Joery Goossens
- Department of Biomedical Sciences, Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Sebastiaan Engelborghs
- Department of Biomedical Sciences, Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology & Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim & Hoge Beuken, Antwerp, Belgium
| | - Maria Bjerke
- Department of Biomedical Sciences, Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
31
|
Llorens F, Kruse N, Schmitz M, Gotzmann N, Golanska E, Thüne K, Zejneli O, Kanata E, Knipper T, Cramm M, Lange P, Zafar S, Sikorska B, Liberski PP, Mitrova E, Varges D, Schmidt C, Sklaviadis T, Mollenhauer B, Zerr I. Evaluation of α‐synuclein as a novel cerebrospinal fluid biomarker in different forms of prion diseases. Alzheimers Dement 2016; 13:710-719. [DOI: 10.1016/j.jalz.2016.09.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/15/2016] [Accepted: 09/29/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Franc Llorens
- Department of Neurology, Clinical Dementia Center University Medical Center Göttingen Göttingen Germany
- German Center for Neurodegenerative Diseases (DZNE) Site Göttingen Germany
| | - Niels Kruse
- Institute for Neuropathology University Medical Center Göttingen Göttingen Germany
| | - Matthias Schmitz
- Department of Neurology, Clinical Dementia Center University Medical Center Göttingen Göttingen Germany
- German Center for Neurodegenerative Diseases (DZNE) Site Göttingen Germany
| | - Nadine Gotzmann
- Department of Neurology, Clinical Dementia Center University Medical Center Göttingen Göttingen Germany
- German Center for Neurodegenerative Diseases (DZNE) Site Göttingen Germany
| | - Ewa Golanska
- Department of Molecular Pathology and Neuropathology Medical University of Lodz Lodz Poland
| | - Katrin Thüne
- Department of Neurology, Clinical Dementia Center University Medical Center Göttingen Göttingen Germany
- German Center for Neurodegenerative Diseases (DZNE) Site Göttingen Germany
| | - Orgeta Zejneli
- Laboratory of Pharmacology, School of Health Sciences, Department of Pharmacy Aristotle University of Thessaloniki Thessaloniki Greece
| | - Eirini Kanata
- Laboratory of Pharmacology, School of Health Sciences, Department of Pharmacy Aristotle University of Thessaloniki Thessaloniki Greece
| | - Tobias Knipper
- Department of Neurology, Clinical Dementia Center University Medical Center Göttingen Göttingen Germany
| | - Maria Cramm
- Department of Neurology, Clinical Dementia Center University Medical Center Göttingen Göttingen Germany
- German Center for Neurodegenerative Diseases (DZNE) Site Göttingen Germany
| | - Peter Lange
- Department of Neurology, Clinical Dementia Center University Medical Center Göttingen Göttingen Germany
| | - Saima Zafar
- Department of Neurology, Clinical Dementia Center University Medical Center Göttingen Göttingen Germany
- German Center for Neurodegenerative Diseases (DZNE) Site Göttingen Germany
| | - Beata Sikorska
- Department of Molecular Pathology and Neuropathology Medical University of Lodz Lodz Poland
| | - Pawel P. Liberski
- Department of Molecular Pathology and Neuropathology Medical University of Lodz Lodz Poland
| | - Eva Mitrova
- Department of Prion Diseases Slovak Medical University Bratislava Bratislava Slovakia
| | - Daniela Varges
- Department of Neurology, Clinical Dementia Center University Medical Center Göttingen Göttingen Germany
| | - Christian Schmidt
- Department of Neurology, Clinical Dementia Center University Medical Center Göttingen Göttingen Germany
| | - Theodoros Sklaviadis
- Laboratory of Pharmacology, School of Health Sciences, Department of Pharmacy Aristotle University of Thessaloniki Thessaloniki Greece
| | - Brit Mollenhauer
- Institute for Neuropathology University Medical Center Göttingen Göttingen Germany
- Paracelsus‐Elena Klinik Center for Parkinsonism and Movement Disorders Kassel Germany
- Department of Neurosurgery University Medical Center Göttingen Göttingen Germany
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Center University Medical Center Göttingen Göttingen Germany
- German Center for Neurodegenerative Diseases (DZNE) Site Göttingen Germany
| |
Collapse
|
32
|
Pietroboni AM, Schiano di Cola F, Scarioni M, Fenoglio C, Spanò B, Arighi A, Cioffi SM, Oldoni E, De Riz MA, Basilico P, Calvi A, Fumagalli GG, Triulzi F, Galimberti D, Bozzali M, Scarpini E. CSF β-amyloid as a putative biomarker of disease progression in multiple sclerosis. Mult Scler 2016; 23:1085-1091. [PMID: 27754941 DOI: 10.1177/1352458516674566] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Neurodegeneration plays a major role in determining disability in multiple sclerosis (MS) patients. Hence, there is increasing need to identify reliable biomarkers, which could serve as prognostic measure of disease progression. OBJECTIVES To assess whether cerebrospinal fluid (CSF) tau and β-amyloid (Aβ) levels were altered in newly diagnosed MS patients and correlated with disability. Moreover, we investigated whether these CSF biomarkers associate with macroscopic brain tissue damage measures. METHODS CSF Aβ and tau levels were determined by enzyme-linked immunosorbent assay in CSF samples from 48 newly diagnosed MS patients, followed-up clinically for 3 years by recording their Expanded Disability Status Scale score at 6-month intervals, and 45 controls. All patients underwent magnetic resonance imaging at baseline and at the end of follow-up to quantify their lesion load (LL). RESULTS CSF Aβ levels were significantly reduced in patients compared to controls ( p < 0.001). Lower CSF Aβ levels at baseline were a disability predictor at 3-year follow-up ( p = 0.009). CSF tau levels correlated with T2- and T1-LL ( p < 0.001). CONCLUSION CSF Aβ reduction is a promising biomarker of neurodegeneration and may predict patients' clinical outcome. Therefore, CSF Aβ should be considered as a potential biomarker of prognostic value.
Collapse
Affiliation(s)
- Anna M Pietroboni
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Schiano di Cola
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marta Scarioni
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Barbara Spanò
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Andrea Arighi
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sara Mg Cioffi
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Emanuela Oldoni
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Milena A De Riz
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Basilico
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Calvi
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio G Fumagalli
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio Triulzi
- Neuroradiology Unit, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Galimberti
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Bozzali
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Elio Scarpini
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
33
|
Bexarotene targets autophagy and is protective against thromboembolic stroke in aged mice with tauopathy. Sci Rep 2016; 6:33176. [PMID: 27624652 PMCID: PMC5021977 DOI: 10.1038/srep33176] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/23/2016] [Indexed: 12/22/2022] Open
Abstract
Stroke is a highly debilitating, often fatal disorder for which current therapies are suitable for only a minor fraction of patients. Discovery of novel, effective therapies is hampered by the fact that advanced age, primary age-related tauopathy or comorbidities typical to several types of dementing diseases are usually not taken into account in preclinical studies, which predominantly use young, healthy rodents. Here we investigated for the first time the neuroprotective potential of bexarotene, an FDA-approved agent, in a co-morbidity model of stroke that combines high age and tauopathy with thromboembolic cerebral ischemia. Following thromboembolic stroke bexarotene enhanced autophagy in the ischemic brain concomitantly with a reduction in lesion volume and amelioration of behavioral deficits in aged transgenic mice expressing the human P301L-Tau mutation. In in vitro studies bexarotene increased the expression of autophagy markers and reduced autophagic flux in neuronal cells expressing P301L-Tau. Bexarotene also restored mitochondrial respiration deficits in P301L-Tau neurons. These newly described actions of bexarotene add to the growing amount of compelling data showing that bexarotene is a potent neuroprotective agent, and identify a novel autophagy-modulating effect of bexarotene.
Collapse
|
34
|
Khan TK, Alkon DL. Alzheimer's Disease Cerebrospinal Fluid and Neuroimaging Biomarkers: Diagnostic Accuracy and Relationship to Drug Efficacy. J Alzheimers Dis 2016; 46:817-36. [PMID: 26402622 DOI: 10.3233/jad-150238] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Widely researched Alzheimer's disease (AD) biomarkers include in vivo brain imaging with PET and MRI, imaging of amyloid plaques, and biochemical assays of Aβ 1 - 42, total tau, and phosphorylated tau (p-tau-181) in cerebrospinal fluid (CSF). In this review, we critically evaluate these biomarkers and discuss their clinical utility for the differential diagnosis of AD. Current AD biomarker tests are either highly invasive (requiring CSF collection) or expensive and labor-intensive (neuroimaging), making them unsuitable for use in the primary care, clinical office-based setting, or to assess drug efficacy in clinical trials. In addition, CSF and neuroimaging biomarkers continue to face challenges in achieving required sensitivity and specificity and minimizing center-to-center variability (for CSF-Aβ 1 - 42 biomarkers CV = 26.5% ; http://www.alzforum.org/news/conference-coverage/paris-standardization-hurdle-spinal-fluid-imaging-markers). Although potentially useful for selecting patient populations for inclusion in AD clinical trials, the utility of CSF biomarkers and neuroimaging techniques as surrogate endpoints of drug efficacy needs to be validated. Recent trials of β- and γ-secretase inhibitors and Aβ immunization-based therapies in AD showed no significant cognitive improvements, despite changes in CSF and neuroimaging biomarkers. As we learn more about the dysfunctional cellular and molecular signaling processes that occur in AD, and how these processes are manifested in tissues outside of the brain, new peripheral biomarkers may also be validated as non-invasive tests to diagnose preclinical and clinical AD.
Collapse
|
35
|
Weise D, Tiepolt S, Awissus C, Hoffmann KT, Lobsien D, Kaiser T, Barthel H, Sabri O, Gertz HJ. Critical Comparison of Different Biomarkers for Alzheimer's Disease in a Clinical Setting. J Alzheimers Dis 2016; 48:425-32. [PMID: 26402006 DOI: 10.3233/jad-150229] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Biomarkers of neuronal injury and amyloid pathology play a pivotal role in the diagnosis of Alzheimer's disease (AD). The degree of AD biomarker congruence is still unclear in clinical practice. OBJECTIVE Diagnosis of AD with regard to the congruence of the clinical diagnosis and different biomarkers. METHODS In this prospective cross-sectional observational study, 54 patients with mild cognitive impairment or dementia due to AD or not due to AD were investigated. Biomarkers of neuronal injury were medial temporal lobe atrophy (MTA) on magnetic resonance imaging (MRI) and tau concentration in the cerebrospinal fluid (CSF). CSF Aβ(1-42) and amyloid-targeting positron emission tomography (PET) were considered as biomarkers of amyloid pathology. RESULTS Forty cases were diagnosed as AD and 14 cases were diagnosed as non-AD based on clinical and routine MRI assessment. AD cases had higher MTA scores, higher levels of CSF tau and lower levels of CSF Aβ(1- 42), and higher amyloid load on PET compared to the non-AD group. In the AD group, completely consistently pathological biomarkers were found in 32.5% , non-pathological in 5% . In 62.5% the findings were inconsistent. Congruence of biomarkers was 67.5% for neuronal injury and for amyloid dysfunction, respectively. In two patients, clinical diagnosis switched to non-AD due to completely consistent non-pathological biomarker findings. The criteria of the international working group were met in 75.0% . CONCLUSION Surprisingly, the number of completely congruent biomarkers was relatively low. Interpretation of AD biomarkers is complicated by multiple biomarker constellations. However, the level of biomarker consistency required to reliably diagnose AD remains uncertain.
Collapse
Affiliation(s)
- David Weise
- Department of Psychiatry, University of Leipzig, Leipzig, Germany.,Department of Neurology, University of Leipzig, Leipzig, Germany
| | - Solveig Tiepolt
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Carolin Awissus
- Department of Psychiatry, University of Leipzig, Leipzig, Germany
| | | | - Donald Lobsien
- Department of Neuroradiology, University of Leipzig, Leipzig, Germany
| | - Thorsten Kaiser
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | | |
Collapse
|
36
|
Riedel BC, Thompson PM, Brinton RD. Age, APOE and sex: Triad of risk of Alzheimer's disease. J Steroid Biochem Mol Biol 2016; 160:134-47. [PMID: 26969397 PMCID: PMC4905558 DOI: 10.1016/j.jsbmb.2016.03.012] [Citation(s) in RCA: 419] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 03/02/2016] [Accepted: 03/06/2016] [Indexed: 02/06/2023]
Abstract
Age, apolipoprotein E ε4 (APOE) and chromosomal sex are well-established risk factors for late-onset Alzheimer's disease (LOAD; AD). Over 60% of persons with AD harbor at least one APOE-ε4 allele. The sex-based prevalence of AD is well documented with over 60% of persons with AD being female. Evidence indicates that the APOE-ε4 risk for AD is greater in women than men, which is particularly evident in heterozygous women carrying one APOE-ε4 allele. Paradoxically, men homozygous for APOE-ε4 are reported to be at greater risk for mild cognitive impairment and AD. Herein, we discuss the complex interplay between the three greatest risk factors for Alzheimer's disease, age, APOE-ε4 genotype and chromosomal sex. We propose that the convergence of these three risk factors, and specifically the bioenergetic aging perimenopause to menopause transition unique to the female, creates a risk profile for AD unique to the female. Further, we discuss the specific risk of the APOE-ε4 positive male which appears to emerge early in the aging process. Evidence for impact of the triad of AD risk factors is most evident in the temporal trajectory of AD progression and burden of pathology in relation to APOE genotype, age and sex. Collectively, the data indicate complex interactions between age, APOE genotype and gender that belies a one size fits all approach and argues for a precision medicine approach that integrates across the three main risk factors for Alzheimer's disease.
Collapse
Affiliation(s)
- Brandalyn C Riedel
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Paul M Thompson
- USC Institute for Neuroimaging and Informatics, University of Southern California, Marina del Rey, CA 90292, USA
| | - Roberta Diaz Brinton
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
37
|
Shams S, Granberg T, Martola J, Li X, Shams M, Fereshtehnejad SM, Cavallin L, Aspelin P, Kristoffersen-Wiberg M, Wahlund LO. Cerebrospinal fluid profiles with increasing number of cerebral microbleeds in a continuum of cognitive impairment. J Cereb Blood Flow Metab 2016; 36:621-8. [PMID: 26661151 PMCID: PMC4794093 DOI: 10.1177/0271678x15606141] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/22/2015] [Accepted: 07/28/2015] [Indexed: 11/15/2022]
Abstract
Cerebral microbleeds (CMBs) are hypothesised to have an important yet unknown role in the dementia disease pathology. In this study we analysed increasing number of CMBs and their independent associations with routine cerebrospinal fluid (CSF) biomarkers in a continuum of cognitive impairment. A total of 1039 patients undergoing dementia investigation were analysed and underwent lumbar puncture, and an MRI scan. CSF samples were analysed for amyloid β (Aβ) 42, total tau (T-tau), tau phosphorylated at threonine 18 (P-tau) and CSF/serum albumin ratios. Increasing number of CMBs were independently associated with low Aβ42 levels, in the whole cohort, Alzheimer's disease and mild cognitive impairment (p < 0.05). CSF/serum albumin ratios were high with multiple CMBs (p < 0.001), reflecting accompanying blood-brain barrier dysfunction. T-tau and P-tau levels were lower in Alzheimer's patients with multiple CMBs when compared to zero CMBs, but did not change in the rest of the cohort. White matter hyperintensities were associated with low Aβ42 in the whole cohort and Alzheimer's disease (p < 0.05). Aβ42 is the routine CSF-biomarker mainly associated with CMBs in cognitive impairment, and there is an accumulative effect with increasing number of CMBs.
Collapse
Affiliation(s)
- Sara Shams
- Department of Clinical Science, Intervention, and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Tobias Granberg
- Department of Clinical Science, Intervention, and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Juha Martola
- Department of Clinical Science, Intervention, and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Xiaozhen Li
- Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden Division of Clinical Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Mana Shams
- Department of Clinical Science, Intervention, and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Seyed-Mohammad Fereshtehnejad
- Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden Division of Clinical Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Lena Cavallin
- Department of Clinical Science, Intervention, and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Aspelin
- Department of Clinical Science, Intervention, and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Kristoffersen-Wiberg
- Department of Clinical Science, Intervention, and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Lars-Olof Wahlund
- Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden Division of Clinical Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
38
|
CSF biomarkers in neurodegenerative and vascular dementias. Prog Neurobiol 2016; 138-140:36-53. [DOI: 10.1016/j.pneurobio.2016.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/12/2016] [Accepted: 03/14/2016] [Indexed: 12/14/2022]
|
39
|
Mitochondrial DNA differentiates Alzheimer's disease from Creutzfeldt-Jakob disease. Alzheimers Dement 2016; 12:546-55. [PMID: 26806388 DOI: 10.1016/j.jalz.2015.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/25/2015] [Accepted: 12/16/2015] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Low content of cell-free mitochondrial DNA (mtDNA) in cerebrospinal fluid (CSF) is a biomarker of early stage Alzheimer's disease (AD), but whether mtDNA is altered in a rapid neurodegenerative dementia such as Creutzfeldt-Jakob disease is unknown. METHODS CSF mtDNA was measured using digital polymerase chain reaction (dPCR) in two independent cohorts comprising a total of 112 patients diagnosed with sporadic Creutzfeldt-Jakob disease (sCJD), probable AD, or non-Alzheimer's type dementia. RESULTS Patients with AD exhibit low mtDNA content in CSF compared with patients diagnosed with sCJD or with non-Alzheimer's type dementias. The CSF concentration of mtDNA does not correlate with Aβ, t-tau, p-tau, and 14-3-3 protein levels in CSF. DISCUSSION Low-CSF mtDNA is not a consequence of brain damage and allows the differential diagnosis of AD from sCJD and other dementias. These results support the hypothesis that mtDNA in CSF is a pathophysiological biomarker of AD.
Collapse
|
40
|
Pan C, Korff A, Galasko D, Ginghina C, Peskind E, Li G, Quinn J, Montine TJ, Cain K, Shi M, Zhang J. Diagnostic Values of Cerebrospinal Fluid T-Tau and Aβ₄₂ using Meso Scale Discovery Assays for Alzheimer's Disease. J Alzheimers Dis 2016; 45:709-19. [PMID: 25613100 DOI: 10.3233/jad-143099] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Meso Scale Discovery (MSD) recently established electrochemiluminescence-based assays to measure cerebrospinal fluid (CSF) levels of total tau (t-tau) and amyloid-β 1-42 peptide (Aβ42) that can aid in the diagnosis of Alzheimer's disease (AD). The goal of this investigation is to independently evaluate this platform and establish cut-off values of these biomarkers for AD diagnosis. OBJECTIVE To validate the analytical and clinical performance of the MSD t-tau and Aβ42 kits and propose diagnostic cut-off values for the field. METHODS The analytical performance of the CSF t-tau and Aβ42 assays was determined, followed by assessment of diagnostic performance of CSF t-tau, Aβ42, and t-tau/Aβ42 in three clinically characterized cohorts. RESULTS Both MSD assays demonstrated consistent and stable analytical performance, as well as resistance to several important pre-analytic variables. Diagnostically, t-tau/Aβ42 performed the best. CONCLUSIONS Our results independently confirm the analytical and clinical performance of the MSD CSF t-tau and Aβ42 assays. Based on a large, multi-center, clinically-diagnosed cohort, we propose for the first time candidate diagnostic cut-offs for MSD measured CSF t-tau, Aβ42, and t-tau/Aβ42. However, these values needs to be refined as more subjects are included and the assays are tested by other laboratories.
Collapse
Affiliation(s)
- Catherine Pan
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Ané Korff
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Douglas Galasko
- Department of Neurosciences, University of California at San Diego, San Diego, CA, USA
| | - Carmen Ginghina
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Elaine Peskind
- Northwest Network VISN-20 Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Ge Li
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Joseph Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA Portland VA Medical Center, Portland, OR, USA
| | - Thomas J Montine
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Kevin Cain
- Department of Biostatistics, University of Washington School of Medicine, Seattle, WA, USA
| | - Min Shi
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Jing Zhang
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
41
|
Sun LH, Ban T, Liu CD, Chen QX, Wang X, Yan ML, Hu XL, Su XL, Bao YN, Sun LL, Zhao LJ, Pei SC, Jiang XM, Zong DK, Ai J. Activation of Cdk5/p25 and tau phosphorylation following chronic brain hypoperfusion in rats involves microRNA-195 down-regulation. J Neurochem 2015; 134:1139-51. [PMID: 26118667 DOI: 10.1111/jnc.13212] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/02/2015] [Accepted: 06/03/2015] [Indexed: 11/27/2022]
Abstract
Chronic brain hypoperfusion (CBH) is a common clinical feature of Alzheimer's disease and vascular dementia, but the underlying molecular mechanism is unclear. Our previous study reported that the down-regulation of microRNA-195 (miR-195) promotes amyloidogenesis via regulation of amyloid precursor protein and β-site amyloid precursor protein cleaving enzyme 1 (BACE1) expression at the post-transcriptional level in CBH rats with bilateral common carotid artery occlusion (2VO). CBH owing to unilateral common carotid artery occlusion (UCCAO) increases tau phosphorylation levels at multiple phosphorylation sites in the brain, but the molecular mechanism is poorly understood. The purpose of this study was to investigate whether miR-195 could both deregulate amyloid metabolism and indirectly deregulate tau phosphorylation in CBH. We observed that 2VO leads to tau hyperphosphorylation at Ser202/Thr205, Ser262, Thr231, and Ser422 and to the conversion from cyclin-dependent kinase 5 (Cdk5)/p35 to Cdk5/p25 in rat hippocampi. Endogenous miR-195 was knocked down using over-expression of its antisense molecule (pre-AMO-miR-195) via a lentivirus (lenti-pre-AMO-miR-195); this knockdown increased the tau phosphorylation at Ser202/Thr205, Ser262, Thr231, Ser422, and the Cdk5/p25 activation, but over-expression of miR-195 using lenti-pre-miR-195 decreased the tau phosphorylation and Cdk5/p25 activation. Further in vitro studies demonstrated that miR-195 over-expression prevented tau hyperphosphorylation and Cdk5/p35 activity, which were increased by miR-195 inhibition. A dual luciferase reporter assay showed that miR-195 bound to the Cdk5r1 gene, which encodes p35 protein, in the 3'UTR and inhibited p35 expression. We concluded that tau hyperphosphorylation involves the down-regulation of miR-195, which is mediated by Cdk5/p25 activation in 2VO rats. Our findings demonstrated that down-regulation of miR-195 led to increased vulnerability via the regulation of multiple targets. Schematic diagram of miR-195 mediated Aβ aggregation and tau hyperphosphorylation in chronic brain hypoperfusion (CBH). First, CBH results in the elevation of nuclear factor-κB (NF-κB), which binds with the promoter sequences of miR-195 and negatively regulates the expression of miR-195. Second, down-regulated miR-195 induces up-regulation of APP and BACE1 and leads to an increase in Aβ levels. Third, some of the elevated Aβ then enter the intracellular space and activate calpain, which promotes the conversion of Cdk5/p35 to Cdk5/p25 and catalyzes the degradation of IκB; IκB is an inhibitor of NF-κB, which activates NF-κB. Cdk5/p25 directly phosphorylates Tau. Fourth, down-regulated miR-195 induces an up-regulation of p35, which provides the active substrates of p25. Our findings demonstrated that the down-regulation of miR-195 plays a key role in the increased vulnerability to dementia via the regulation of multiple targets following CBH.
Collapse
Affiliation(s)
- Li-Hua Sun
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Tao Ban
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Cheng-Di Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Qing-Xin Chen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Xu Wang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Mei-Ling Yan
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Xue-Ling Hu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiao-Lin Su
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Ya-Nan Bao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Lin-Lin Sun
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Lin-Jing Zhao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuang-Chao Pei
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Xue-Mei Jiang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - De-Kang Zong
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| | - Jing Ai
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
42
|
Mukaetova-Ladinska EB, Li M, Kalaria RN. tau protein, ischemic injury and vascular dementia. FUTURE NEUROLOGY 2015. [DOI: 10.2217/fnl.15.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Clinical, neuroimaging and neuropathological studies have confirmed overlap between Alzheimer's disease (AD) and vascular dementia (VaD). Classical neuropathological changes of AD (plaques and tangles) can be present in VaD. We review neuroimaging, biochemical and animal studies to consider the role of tau protein in ischemic injury and VaD pathogenesis. The evidence comes largely from transgenic animal studies that confirm that tau transgenes influence cerebral vasculature. Clinicobiochemical studies in the cerebrospinal fluid (CSF) have, similarly, confirmed alterations in both total and phosphorylated tau protein in VaD. These data suggest that tau protein not only serves as a potential diagnostic tool for differential diagnosis of VaD from other types of dementia, but may also be a therapeutic target in ischemic stroke.
Collapse
Affiliation(s)
| | - Mosi Li
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Raj N Kalaria
- Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, NE4 5PL, UK
| |
Collapse
|
43
|
Forlenza OV, Radanovic M, Talib LL, Aprahamian I, Diniz BS, Zetterberg H, Gattaz WF. Cerebrospinal fluid biomarkers in Alzheimer's disease: Diagnostic accuracy and prediction of dementia. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2015; 1:455-63. [PMID: 27239524 PMCID: PMC4879480 DOI: 10.1016/j.dadm.2015.09.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Guidelines for the use of cerebrospinal fluid (CSF) biomarkers in the diagnosis of Alzheimer's disease (AD) establish that each laboratory must use internally qualified cutoff values. We determined the concentrations of biomarkers that discriminate cases from controls and combinations that predict the progression to dementia in a Brazilian cohort. METHODS Concentrations of amyloid-beta peptide (Aβ1-42), total tau (T-tau), and (181)Thr-phosphorylated-tau (P-tau) were determined in CSF samples from 184 older adults (68 mild cognitive impairment, 41 AD, 34 non-AD cognitive impairment, and 41 controls) by the INNO-BIA AlzBio3 assay. RESULTS Cutoff values discriminating AD from controls are as follows: Aβ1-42: 416.0 pg/mL (sensitivity [SE]: 83%, specificity (SP): 70%); T-tau: 76.7 pg/mL (SE: 82%, SP: 67%); P-tau: 36.1 pg/mL (SE: 83%, SP: 49%); Aβ1-42/P-tau <9.53 (SE: 88%, SP: 78%); and Aβ1-42/T-tau <4.13 (SE: 80%; SP: 80%). Combining values Aβ1-42 <416.5 pg/mL and Aβ1-42/P-tau <9.5 best predicted the conversion in 2 years (Cox regression: hazard ratio 7.24 [2.09-25.06], P = .002, SE: 74%, Sp: 73%). DISCUSSION Our findings are in line with most of the available evidence in this field; yet, our cutoff values are different from those derived from other laboratories.
Collapse
Affiliation(s)
- Orestes V. Forlenza
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Marcia Radanovic
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Leda L. Talib
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Ivan Aprahamian
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Breno S. Diniz
- Department of Mental Health, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- UCL Institute of Neurology, London, UK
| | - Wagner F. Gattaz
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
44
|
Llorens F, Schmitz M, Gloeckner SF, Kaerst L, Hermann P, Schmidt C, Varges D, Zerr I. Increased albumin CSF/serum ratio in dementia with Lewy bodies. J Neurol Sci 2015; 358:398-403. [PMID: 26476775 DOI: 10.1016/j.jns.2015.10.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/02/2015] [Accepted: 10/06/2015] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alterations in the CSF/serum albumin ratio (Qalb) is currently recognized as one of the most reliable markers of blood-brain barrier impairment and blood-CSF barrier permeability, but its potential role as a biomarker in the differential diagnosis of neurological diseases has been poorly analysed. METHODS We evaluated Qalb and core CSF biomarkers (Tau, p-Tau and Aβ42) in a large patient population of neurological and neurodegenerative cases. Diagnostic test evaluation was assessed by ROC-AUC analysis. RESULTS In the differential diagnostic analysis, increased Qalb was found in dementia with Lewy bodies (DLB) patients compared to other diseases, either individually or stratified in non-dementia and dementia groups. When clinical groups were analysed individually and compared to controls, Qalb was also increased in stroke and Parkinson's disease dementia (PDD) cases, but not in Parkinson's disease (PD). Qalb in DLB cases correlate with CSF Aβ42 levels but not with Tau and p-Tau levels. Due to the lower CSF Aβ42 levels in DLB compared to PD and PDD, the potential clinical applicability of Qalb with respect to the DLB diagnosis is increased in combination with CSF Aβ42 analysis. CONCLUSIONS The present study demonstrates increased Qalb in synucleinopathies associated with dementia revealing a potential new clinical approach for the differential diagnosis of DLB.
Collapse
Affiliation(s)
- Franc Llorens
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.
| | - Matthias Schmitz
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | | | - Lisa Kaerst
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Germany
| | - Peter Hermann
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Germany
| | - Christian Schmidt
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Germany
| | - Daniela Varges
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Germany
| | - Inga Zerr
- Clinical Dementia Center, Department of Neurology, University Medical Center Göttingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| |
Collapse
|
45
|
Sancesario GM, Bernardini S. How many biomarkers to discriminate neurodegenerative dementia? Crit Rev Clin Lab Sci 2015; 52:314-26. [PMID: 26292074 DOI: 10.3109/10408363.2015.1051658] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A number of cerebrospinal fluid (CSF) biomarkers are currently used for the diagnosis of dementia. Opposite changes in the level of amyloid-β(1-42) versus total tau and phosphorylated-tau181 in the CSF reflect the specific pathology of Alzheimer's disease (AD) in the brain. This panel of biomarkers has proven to be effective to differentiate AD from controls and from the major types of neurodegenerative dementia, and to evaluate the progression from mild cognitive impairment to AD. In the absence of specific biomarkers reflecting the pathologies of the other most common forms of dementia, such as Lewy Body disease, Frontotemporal lobar degeneration, Creutzfeldt-Jakob disease, etc., the evaluation of biomarkers of AD pathology is used, attempting to exclude rather than to confirm AD. Other biomarkers included in the common clinical practice do not clearly relate to the underlying pathology: progranulin (PGRN) is a selective marker of frontotemporal dementia with mutations in the PGRN gene; the 14-3-3 protein is a highly sensitive and specific marker for Creutzfeldt-Jakob disease, but has to be used carefully in differentiating rapid progressive dementia; and α-synuclein is an emerging candidate biomarker of the different forms of synucleinopathy. This review summarizes several biomarkers of neurodegenerative dementia validated based on the neuropathological processes occurring in brain tissue. Notwithstanding the paucity of pathologically validated biomarkers and their high analytical variability, the combinations of these biomarkers may well represent a key and more precise analytical and diagnostic tool in the complex plethora of degenerative dementia.
Collapse
Affiliation(s)
- Giulia M Sancesario
- a Department of Clinical and Behavioural Neurology , Santa Lucia Foundation, IRCCS , Rome , Italy and
| | - Sergio Bernardini
- b Department of Experimental Medicine and Surgery , Tor Vergata University of Rome , Rome , Italy
| |
Collapse
|
46
|
Brock AJ, Kasus-Jacobi A, Lerner M, Logan S, Adesina AM, Anne Pereira H. The antimicrobial protein, CAP37, is upregulated in pyramidal neurons during Alzheimer's disease. Histochem Cell Biol 2015; 144:293-308. [PMID: 26170148 PMCID: PMC4575391 DOI: 10.1007/s00418-015-1347-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2015] [Indexed: 01/02/2023]
Abstract
Inflammation is a well-defined factor in Alzheimer's disease (AD). There is a strong need to identify the molecules contributing to neuroinflammation so that therapies can be designed to prevent immune-mediated neurotoxicity. The cationic antimicrobial protein of 37 kDa (CAP37) is an inflammatory mediator constitutively expressed in neutrophils (PMNs). In addition to antibiotic activity, CAP37 exerts immunomodulatory effects on microglia. We hypothesize that CAP37 mediates the neuroinflammation associated with AD. However, PMNs are not customarily associated with the pathology of AD. This study was therefore designed to identify non-neutrophilic source(s) of CAP37 in brains of AD patients. Brain tissues from patients and age-matched controls were analyzed for CAP37 expression using immunohistochemistry (IHC). To determine factors that induce CAP37 in AD, HCN-1A primary human neurons were treated with tumor necrosis factor-alpha (TNF-α) or amyloid β1-40 (Aβ) and analyzed by IHC. Western blotting and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to confirm CAP37 expression in neurons and brain tissues. IHC revealed CAP37 in cortical neurons in temporal and parietal lobes as well as CA3 and CA4 hippocampal neurons in patients with AD. CAP37 was found in more neurons in AD patients compared with age-matched controls. qRT-PCR and Western blotting showed an increase in CAP37 transcript and protein in the AD temporal lobe, a brain region that is highly impacted in AD. qRT-PCR observations confirmed CAP37 expression in neurons. TNF-α and Aβ increased neuronal expression of CAP37. These findings support our hypothesis that neuronal CAP37 may modulate the neuroinflammatory response in AD.
Collapse
Affiliation(s)
- Amanda J Brock
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA
| | - Anne Kasus-Jacobi
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA.,Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA
| | - Megan Lerner
- Department of Surgery, University of Oklahoma Health Sciences Center, 1122 NE 13th St., ORB 350, Oklahoma City, OK, 73117, USA
| | - Sreemathi Logan
- Department of Geriatrics, University of Oklahoma Health Sciences Center, 975 NE 10th St., BRC 1303, Oklahoma City, OK, 73104, USA
| | - Adekunle M Adesina
- Department of Pathology, Baylor College of Medicine, One Baylor Plaza, Rm 286A, Houston, TX, 77030, USA
| | - H Anne Pereira
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA. .,Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA. .,Department of Cell Biology, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 329, Oklahoma City, OK, USA. .,Department of Pathology, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 329, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
47
|
Llorens F, Kruse N, Schmitz M, Shafiq M, da Cunha JEG, Gotzman N, Zafar S, Thune K, de Oliveira JRM, Mollenhauer B, Zerr I. Quantification of CSF biomarkers using an electrochemiluminescence-based detection system in the differential diagnosis of AD and sCJD. J Neurol 2015; 262:2305-11. [DOI: 10.1007/s00415-015-7837-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 06/24/2015] [Accepted: 06/24/2015] [Indexed: 01/26/2023]
|
48
|
Skillbäck T, Farahmand BY, Rosén C, Mattsson N, Nägga K, Kilander L, Religa D, Wimo A, Winblad B, Schott JM, Blennow K, Eriksdotter M, Zetterberg H. Cerebrospinal fluid tau and amyloid-β1-42 in patients with dementia. Brain 2015; 138:2716-31. [PMID: 26133663 DOI: 10.1093/brain/awv181] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 04/30/2015] [Indexed: 12/12/2022] Open
Abstract
Progressive cognitive decline in combination with a cerebrospinal fluid biomarker pattern of low levels of amyloid-β1-42 and high levels of total tau and phosphorylated tau is typical of Alzheimer's disease. However, several neurodegenerative disorders may overlap with Alzheimer's disease both in regards to clinical symptoms and neuropathology. In a uniquely large cohort of dementia patients, we examined the associations of cerebrospinal fluid biomarkers for Alzheimer's disease molecular pathology with clinical dementia diagnoses and disease severity. We cross-referenced the Swedish Dementia Registry with the clinical laboratory database at the Sahlgrenska University Hospital. The final data set consisted of 5676 unique subjects with a clinical dementia diagnosis and a complete set of measurements for cerebrospinal fluid amyloid-β1-42, total tau and phosphorylated tau. In cluster analysis, disregarding clinical diagnosis, the optimal natural separation of this data set was into two clusters, with the majority of patients with early onset Alzheimer's disease (75%) and late onset Alzheimer's disease (73%) assigned to one cluster and the patients with vascular dementia (91%), frontotemporal dementia (94%), Parkinson's disease dementia (94%) and dementia with Lewy bodies (87%) to the other cluster. Frontotemporal dementia had the highest cerebrospinal fluid levels of amyloid-β1-42 and the lowest levels of total tau and phosphorylated tau. The highest levels of total tau and phosphorylated tau and the lowest levels of amyloid-β1-42 and amyloid-β1-42:phosphorylated tau ratios were found in Alzheimer's disease. Low amyloid-β1-42, high total tau and high phosphorylated tau correlated with low Mini-Mental State Examination scores in Alzheimer's disease. In Parkinson's disease dementia and vascular dementia low cerebrospinal fluid amyloid-β1-42 was associated with low Mini-Mental State Examination score. In the vascular dementia, frontotemporal dementia, dementia with Lewy bodies and Parkinson's disease dementia groups 53%, 34%, 67% and 53% of the subjects, respectively had abnormal amyloid-β1-42 levels, 41%, 41%, 28% and 28% had abnormal total tau levels, and 29%, 28%, 25% and 19% had abnormal phosphorylated tau levels. Cerebrospinal fluid biomarkers were strongly associated with specific clinical dementia diagnoses with Alzheimer's disease and frontotemporal dementia showing the greatest difference in biomarker levels. In addition, cerebrospinal fluid amyloid-β1-42, total tau, phosphorylated tau and the amyloid-β1-42:phosphorylated tau ratio all correlated with poor cognitive performance in Alzheimer's disease, as did cerebrospinal fluid amyloid-β1-42 in Parkinson's disease dementia and vascular dementia. The results support the use of cerebrospinal fluid biomarkers to differentiate between dementias in clinical practice, and to estimate disease severity.
Collapse
Affiliation(s)
- Tobias Skillbäck
- 1 Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Bahman Y Farahmand
- 2 Department of Neurobiology, Care Sciences, and Society (NVS), Centre for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden
| | - Christoffer Rosén
- 1 Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Niklas Mattsson
- 1 Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden 3 Department of Veterans Affairs Medical Centre, Centre for Imaging of Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Katarina Nägga
- 4 Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Lena Kilander
- 5 Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Dorota Religa
- 6 Department of Neurobiology, Care Sciences, and Society (NVS), Centre for Alzheimer Research, Division for Neurogeriatrtics, Karolinska Institutet, Huddinge, Sweden 7 Department Geriatric Medicine, Karolinska University Hospital, Huddinge, Sweden
| | - Anders Wimo
- 6 Department of Neurobiology, Care Sciences, and Society (NVS), Centre for Alzheimer Research, Division for Neurogeriatrtics, Karolinska Institutet, Huddinge, Sweden
| | - Bengt Winblad
- 6 Department of Neurobiology, Care Sciences, and Society (NVS), Centre for Alzheimer Research, Division for Neurogeriatrtics, Karolinska Institutet, Huddinge, Sweden 7 Department Geriatric Medicine, Karolinska University Hospital, Huddinge, Sweden
| | - Jonathan M Schott
- 1 Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- 1 Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Maria Eriksdotter
- 2 Department of Neurobiology, Care Sciences, and Society (NVS), Centre for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden 7 Department Geriatric Medicine, Karolinska University Hospital, Huddinge, Sweden
| | - Henrik Zetterberg
- 1 Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden 8 UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| |
Collapse
|
49
|
|
50
|
Inekci D, Jonesco DS, Kennard S, Karsdal MA, Henriksen K. The potential of pathological protein fragmentation in blood-based biomarker development for dementia - with emphasis on Alzheimer's disease. Front Neurol 2015; 6:90. [PMID: 26029153 PMCID: PMC4426721 DOI: 10.3389/fneur.2015.00090] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 04/10/2015] [Indexed: 12/12/2022] Open
Abstract
The diagnosis of dementia is challenging and early stages are rarely detected limiting the possibilities for early intervention. Another challenge is the overlap in the clinical features across the different dementia types leading to difficulties in the differential diagnosis. Identifying biomarkers that can detect the pre-dementia stage and allow differential diagnosis could provide an opportunity for timely and optimal intervention strategies. Also, such biomarkers could help in selection and inclusion of the right patients in clinical trials of both Alzheimer’s disease and other dementia treatment candidates. The cerebrospinal fluid (CSF) has been the most investigated source of biomarkers and several candidate proteins have been identified. However, looking solely at protein levels is too simplistic to provide enough detailed information to differentiate between dementias, as there is a significant crossover between the proteins involved in the different types of dementia. Additionally, CSF sampling makes these biomarkers challenging for presymptomatic identification. We need to focus on disease-specific protein fragmentation to find a fragment pattern unique for each separate dementia type – a form of protein fragmentology. Targeting protein fragments generated by disease-specific combinations of proteins and proteases opposed to detecting the intact protein could reduce the overlap between diagnostic groups as the extent of processing as well as which proteins and proteases constitute the major hallmark of each dementia type differ. In addition, the fragments could be detectable in blood as they may be able to cross the blood–brain barrier due to their smaller size. In this review, the potential of the fragment-based biomarker discovery for dementia diagnosis and prognosis is discussed, especially highlighting how the knowledge from CSF protein biomarkers can be used to guide blood-based biomarker development.
Collapse
Affiliation(s)
- Dilek Inekci
- Nordic Bioscience, Biomarkers and Research , Herlev , Denmark ; Systems Biology, Technical University of Denmark , Lyngby , Denmark
| | | | - Sophie Kennard
- Nordic Bioscience, Biomarkers and Research , Herlev , Denmark
| | | | - Kim Henriksen
- Nordic Bioscience, Biomarkers and Research , Herlev , Denmark
| |
Collapse
|