1
|
Rezaee N, Hone E, Sohrabi H, Abdulraheem R, Johnson SK, Gunzburg S, Martins RN, Fernando WMADB. Investigating the Impact of Sorghum on Tau Protein Phosphorylation and Mitochondrial Dysfunction Modulation in Alzheimer's Disease: An In Vitro Study. Nutrients 2025; 17:516. [PMID: 39940374 PMCID: PMC11820761 DOI: 10.3390/nu17030516] [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: 12/23/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder with poorly understood pathology. Elevated tau, phospho-tau and mitochondrial dysfunction are significantly correlated with an increased risk of AD and are therefore targets for disease-modifying therapy. In this study, we examined the effects of polyphenolic extracts from six different varieties of sorghum: Shawaya short black-1 (Black), IS1311C (Brown), QL33/QL36 (Red), B923296 (Red), QL12 (White), and QL33 (Red) on the attenuation of beta amyloid-induced phospho-tau levels, total tau levels, and mitochondrial dysfunction in neuronal cells. METHOD Tau proteins (231 (pT231), Serine- 199 (pS199), and total tau proteins (T-tau)) were detected and quantified using sandwich ELISA kits, while mitochondrial dysfunction was measured in terms of mitochondrial membrane potential (Δψm) and adenosine triphosphate (ATP) levels. RESULTS Almost all varieties of the sorghum extracts reduced the beta amyloid-induced pS199 and pT231 levels (p ≤ 0.05). The optimum concentration of QL33/QL36 (1000 µg/mL), QL12 (2000 µg/mL), and QL33 (2000 µg/mL) strongly attenuated the phospho-tau level. Sorghum IS1311C (750 µg/mL) showed the highest Δψm reduction (39.8%), whereas QL33 (2000 µg/mL) most strongly improved the ATP level (37.7%) (p ≤ 0.01). For both Δψm and ATP assays, the least activity was observed in sorghum B923296 at 21% and 25.5%, respectively (p ≤ 0.01). CONCLUSIONS The polyphenol extracts from sorghum attenuated the tau toxicity and Aβ-induced mitochondrial dysfunction in a variety- and dose-dependent manner and made a promising disease-modifying agent against AD. However, extensive research is needed to validate the efficacy of the sorghum extracts prior to animal and clinical studies.
Collapse
Affiliation(s)
- Nasim Rezaee
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia; (N.R.); (E.H.); (H.S.); (R.A.); (R.N.M.)
| | - Eugene Hone
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia; (N.R.); (E.H.); (H.S.); (R.A.); (R.N.M.)
| | - Hamid Sohrabi
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia; (N.R.); (E.H.); (H.S.); (R.A.); (R.N.M.)
- Centre for Ageing, Health Future Institute, Murdoch University, Murdoch, WA 6150, Australia
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Rasheed Abdulraheem
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia; (N.R.); (E.H.); (H.S.); (R.A.); (R.N.M.)
| | | | | | - Ralph N. Martins
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia; (N.R.); (E.H.); (H.S.); (R.A.); (R.N.M.)
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - W. M. A. D. Binosha Fernando
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia; (N.R.); (E.H.); (H.S.); (R.A.); (R.N.M.)
| |
Collapse
|
2
|
Hüper L, Steinacker P, Polyakova M, Mueller K, Godulla J, Herzig S, Danek A, Engel A, Diehl‐Schmid J, Classen J, Fassbender K, Fliessbach K, Jahn H, Kassubek J, Kornhuber J, Landwehrmeyer B, Lauer M, Obrig H, Oeckl P, Prudlo J, Saur D, Anderl‐Straub S, Synofzik M, Wagner M, Wiltfang J, Winkelmann J, Volk AE, FTLD Consortium Germany, Huppertz H, Otto M, Schroeter ML. Neurofilaments and progranulin are related to atrophy in frontotemporal lobar degeneration - A transdiagnostic study cross-validating atrophy and fluid biomarkers. Alzheimers Dement 2024; 20:4461-4475. [PMID: 38865340 PMCID: PMC11247715 DOI: 10.1002/alz.13863] [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: 12/21/2023] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 06/14/2024]
Abstract
INTRODUCTION Frontotemporal lobar degeneration (FTLD) encompasses behavioral variant frontotemporal dementia (bvFTD), progressive supranuclear palsy, corticobasal syndrome/degeneration, and primary progressive aphasias (PPAs). We cross-validated fluid biomarkers and neuroimaging. METHODS Seven fluid biomarkers from cerebrospinal fluid and serum were related to atrophy in 428 participants including these FTLD subtypes, logopenic variant PPA (lvPPA), Alzheimer's disease (AD), and healthy subjects. Atrophy was assessed by structural magnetic resonance imaging and atlas-based volumetry. RESULTS FTLD subtypes, lvPPA, and AD showed specific profiles for neurofilament light chain, phosphorylated heavy chain, tau, phospho-tau, amyloid beta1-42 from serum/cerebrospinal fluid, and brain atrophy. Neurofilaments related to regional atrophy in bvFTD, whereas progranulin was associated with atrophy in semantic variant PPA. Ubiquitin showed no effects. DISCUSSION Results specify biomarker and atrophy patterns in FTLD and AD supporting differential diagnosis. They identify neurofilaments and progranulin in interaction with structural imaging as promising candidates for monitoring disease progression and therapy. HIGHLIGHTS Study cross-validated neuroimaging and fluid biomarkers in dementia. Five kinds of frontotemporal lobar degeneration and two variants of Alzheimer's disease. Study identifies disease-specific fluid biomarker and atrophy profiles. Fluid biomarkers and atrophy interact in a disease-specific way. Neurofilaments and progranulin are proposed as biomarkers for diagnosis and therapy.
Collapse
|
3
|
Abukuri DN. Novel Biomarkers for Alzheimer's Disease: Plasma Neurofilament Light and Cerebrospinal Fluid. Int J Alzheimers Dis 2024; 2024:6668159. [PMID: 38779175 PMCID: PMC11111307 DOI: 10.1155/2024/6668159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD) represent an increasingly significant public health concern. As clinical diagnosis faces challenges, biomarkers are becoming increasingly important in research, trials, and patient assessments. While biomarkers like amyloid-β peptide, tau proteins, CSF levels (Aβ, tau, and p-tau), and neuroimaging techniques are commonly used in AD diagnosis, they are often limited and invasive in monitoring and diagnosis. For this reason, blood-based biomarkers are the optimal choice for detecting neurodegeneration in brain diseases due to their noninvasiveness, affordability, reliability, and consistency. This literature review focuses on plasma neurofilament light (NfL) and CSF NfL as blood-based biomarkers used in recent AD diagnosis. The findings revealed that the core CSF biomarkers of neurodegeneration (T-tau, P-tau, and Aβ42), CSF NFL, and plasma T-tau were strongly associated with Alzheimer's disease, and the core biomarkers were strongly associated with mild cognitive impairment due to Alzheimer's disease. Elevated levels of plasma and cerebrospinal fluid NfL were linked to decreased [18F]FDG uptake in corresponding brain areas. In participants with Aβ positivity (Aβ+), NfL correlated with reduced metabolism in regions susceptible to Alzheimer's disease. In addition, CSF NfL levels correlate with brain atrophy and predict cognitive changes, while plasma total tau does not. Plasma P-tau, especially in combination with Aβ42/Aβ40, is promising for symptomatic AD stages. Though not AD-exclusive, blood NfL holds promise for neurodegeneration detection and assessing treatment efficacy. Given the consistent levels of T-tau, P-tau, Aβ42, and NFL in CSF, their incorporation into both clinical practice and research is highly recommended.
Collapse
|
4
|
Singh S A, Ansari MN, M. Elossaily G, Vellapandian C, Prajapati B. Investigating the Potential Impact of Air Pollution on Alzheimer's Disease and the Utility of Multidimensional Imaging for Early Detection. ACS OMEGA 2024; 9:8615-8631. [PMID: 38434844 PMCID: PMC10905749 DOI: 10.1021/acsomega.3c06328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/25/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024]
Abstract
Pollution is ubiquitous, and much of it is anthropogenic in nature, which is a severe risk factor not only for respiratory infections or asthma sufferers but also for Alzheimer's disease, which has received a lot of attention recently. This Review aims to investigate the primary environmental risk factors and their profound impact on Alzheimer's disease. It underscores the pivotal role of multidimensional imaging in early disease identification and prevention. Conducting a comprehensive review, we delved into a plethora of literature sources available through esteemed databases, including Science Direct, Google Scholar, Scopus, Cochrane, and PubMed. Our search strategy incorporated keywords such as "Alzheimer Disease", "Alzheimer's", "Dementia", "Oxidative Stress", and "Phytotherapy" in conjunction with "Criteria Pollutants", "Imaging", "Pathology", and "Particulate Matter". Alzheimer's disease is not only a result of complex biological factors but is exacerbated by the infiltration of airborne particles and gases that surreptitiously breach the nasal defenses to traverse the brain, akin to a Trojan horse. Various imaging modalities and noninvasive techniques have been harnessed to identify disease progression in its incipient stages. However, each imaging approach possesses inherent limitations, prompting exploration of a unified technique under a single umbrella. Multidimensional imaging stands as the linchpin for detecting and forestalling the relentless march of Alzheimer's disease. Given the intricate etiology of the condition, identifying a prospective candidate for Alzheimer's disease may take decades, rendering the development of a multimodal imaging technique an imperative. This research underscores the pressing need to recognize the chronic ramifications of invisible particulate matter and to advance our understanding of the insidious environmental factors that contribute to Alzheimer's disease.
Collapse
Affiliation(s)
- Ankul Singh S
- Department
of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology (SRMIST), Kattankulathur, Tamil Nadu 603203, India
| | - Mohd Nazam Ansari
- Department
of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Gehan M. Elossaily
- Department
of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 13713, Saudi Arabia
| | - Chitra Vellapandian
- Department
of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology (SRMIST), Kattankulathur, Tamil Nadu 603203, India
| | - Bhupendra Prajapati
- Department
of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy,
Shree S.K. Patel College of Pharmaceutical Education and Research, Ganpat University, Gozaria Highway, Mehsana, North Gujarat 384012, India
| |
Collapse
|
5
|
Doher N, Davoudi V, Magaki S, Townley RA, Haeri M, Vinters HV. Illustrated Neuropathologic Diagnosis of Alzheimer's Disease. Neurol Int 2023; 15:857-867. [PMID: 37489360 PMCID: PMC10366902 DOI: 10.3390/neurolint15030054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 07/26/2023] Open
Abstract
As of 2022, the prevalence of Alzheimer's disease (AD) among individuals aged 65 and older is estimated to be 6.2 million in the United States. This figure is predicted to grow to 13.8 million by 2060. An accurate assessment of neuropathologic changes represents a critical step in understanding the underlying mechanisms in AD. The current method for assessing postmortem Alzheimer's disease neuropathologic change follows version 11 of the National Alzheimer's Coordinating Center (NACC) coding guidebook. Ambiguity regarding steps in the ABC scoring method can lead to increased time or inaccuracy in staging AD. We present a concise overview of how this postmortem diagnosis is made and relate it to the evolving understanding of antemortem AD biomarkers.
Collapse
Affiliation(s)
- Nicholas Doher
- Department of Neurology, University of Florida, Gainesville, FL 32611, USA
| | - Vahid Davoudi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Shino Magaki
- Department of Pathology and Laboratory Medicine, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - Ryan A Townley
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- The University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway City, KS 66205, USA
| | - Mohammad Haeri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- The University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway City, KS 66205, USA
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
- Department of Neurology, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
- Brain Research Institute, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
6
|
del Campo M, Zetterberg H, Gandy S, Onyike CU, Oliveira F, Udeh‐Momoh C, Lleó A, Teunissen CE, Pijnenburg Y. New developments of biofluid-based biomarkers for routine diagnosis and disease trajectories in frontotemporal dementia. Alzheimers Dement 2022; 18:2292-2307. [PMID: 35235699 PMCID: PMC9790674 DOI: 10.1002/alz.12643] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 01/31/2023]
Abstract
Frontotemporal dementia (FTD) covers a spectrum of neurodegenerative disorders with different phenotypes, genetic backgrounds, and pathological states. Its clinicopathological diversity challenges the diagnostic process and the execution of clinical trials, calling for specific diagnostic biomarkers of pathologic FTD types. There is also a need for biomarkers that facilitate disease staging, quantification of severity, monitoring in clinics and observational studies, and for evaluation of target engagement and treatment response in clinical trials. This review discusses current FTD biofluid-based biomarker knowledge taking into account the differing applications. The limitations, knowledge gaps, and challenges for the development and implementation of such markers are also examined. Strategies to overcome these hurdles are proposed, including the technologies available, patient cohorts, and collaborative research initiatives. Access to robust and reliable biomarkers that define the exact underlying pathophysiological FTD process will meet the needs for specific diagnosis, disease quantitation, clinical monitoring, and treatment development.
Collapse
Affiliation(s)
- Marta del Campo
- Departamento de Ciencias Farmacéuticas y de la SaludFacultad de FarmaciaUniversidad San Pablo‐CEUCEU UniversitiesMadridSpain
| | - Henrik Zetterberg
- Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden,Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden,UK Dementia Research Institute at UCLLondonUK,Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK,Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| | - Sam Gandy
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Chiadi U Onyike
- Division of Geriatric Psychiatry and NeuropsychiatryThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Fabricio Oliveira
- Department of Neurology and NeurosurgeryEscola Paulista de MedicinaFederal University of São Paulo (UNIFESP)São PauloSão PauloBrazil
| | - Chi Udeh‐Momoh
- Ageing Epidemiology Research UnitSchool of Public HealthFaculty of MedicineImperial College LondonLondonUK,Translational Health SciencesFaculty of MedicineUniversity of BristolBristolUK
| | - Alberto Lleó
- Neurology DepartmentHospital de la Santa Creu I Sant PauBarcelonaSpain
| | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Clinical ChemistryAmsterdam NeuroscienceAmsterdam University Medical CentersVrije UniversiteitAmsterdamthe Netherlands
| | - Yolande Pijnenburg
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
| |
Collapse
|
7
|
Zetterberg H. Biofluid-based biomarkers for Alzheimer's disease-related pathologies: An update and synthesis of the literature. Alzheimers Dement 2022; 18:1687-1693. [PMID: 35213777 PMCID: PMC9514308 DOI: 10.1002/alz.12618] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/05/2021] [Accepted: 01/10/2022] [Indexed: 01/24/2023]
Abstract
The past few years have seen an explosion in sensitive and specific assays for cerebrospinal fluid (CSF) and blood biomarkers for Alzheimer's disease (AD) and related disorders, as well as some novel assays based on pathological seed-induced protein misfolding in patient samples. Here, I review this exciting field that promises to transform dementia diagnostics and disease monitoring. I discuss data on biomarkers for amyloid beta (Aβ) and tau pathology, neurodegeneration, and glial activation, mention the most promising biomarkers for α-synuclein and TDP-43 pathology, and highlight the need for further research into common co-pathologies. Finally, I consider practical aspects of blood-based biomarker-supported AD diagnostics and emphasize the importance of biomarker interpretation in a full clinical context.
Collapse
Affiliation(s)
- Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| |
Collapse
|
8
|
Haußmann R, Homeyer P, Brandt MD, Donix M. [Prognostic and diagnostic value of cerebrospinal fluid analysis in neurodegenerative dementia diseases]. DER NERVENARZT 2022; 93:1236-1242. [PMID: 35670835 DOI: 10.1007/s00115-022-01339-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 11/25/2022]
Abstract
Cerebrospinal fluid (CSF) analysis is an important diagnostic tool in the assessment of dementia. For the differentiation of Alzheimer's disease from other etiologies of dementia syndromes, established biological markers could be helpful to confirm a distinctive neuropathology. Whereas negative CSF findings can rule out the majority of primarily neurodegenerative disorders, overlapping biomarker profiles remain a diagnostic challenge. Therefore, it is important to interpret CSF results within a specific clinical context. Furthermore, atypical CSF data can be challenging and require profound knowledge of preanalytics, biomarker profiles and the broad spectrum of diseases associated with cognitive decline. Beyond the Alzheimer's disease clinical spectrum, current studies aim at investigating CSF biomarkers to better differentiate tauopathies, TDP43(Transactive response DNA binding protein 43 kDa)-proteinopathies and synucleinopathies.
Collapse
Affiliation(s)
- R Haußmann
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.
| | - P Homeyer
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland
| | - M D Brandt
- Klinik und Poliklinik für Neurologie, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.,DZNE, Deutsches Zentrum für Neurodegenerative Erkrankungen, Dresden, Deutschland
| | - M Donix
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.,DZNE, Deutsches Zentrum für Neurodegenerative Erkrankungen, Dresden, Deutschland
| |
Collapse
|
9
|
Kocurova G, Ricny J, Ovsepian SV. Autoantibodies targeting neuronal proteins as biomarkers for neurodegenerative diseases. Theranostics 2022; 12:3045-3056. [PMID: 35547759 PMCID: PMC9065204 DOI: 10.7150/thno.72126] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/09/2022] [Indexed: 01/08/2023] Open
Abstract
Neurodegenerative diseases (NDDs) are associated with the accumulation of a range of misfolded proteins across the central nervous system and related autoimmune responses, including the generation of antibodies and the activation of immune cells. Both innate and adaptive immunity become mobilized, leading to cellular and humoral effects. The role of humoral immunity in disease onset and progression remains to be elucidated with rising evidence suggestive of positive (protection, repair) and negative (injury, toxicity) outcomes. In this study, we review advances in research of neuron-targeting autoantibodies in the most prevalent NDDs. We discuss their biological origin, molecular diversity and changes in the course of diseases, consider their relevance to the initiation and progression of pathology as well as diagnostic and prognostic significance. It is suggested that the emerging autoimmune aspects of NDDs not only could facilitate the early detection but also might help to elucidate previously unknown facets of pathobiology with relevance to the development of precision medicine.
Collapse
Affiliation(s)
- Gabriela Kocurova
- Experimental Neurobiology Program, National Institute of Mental Health, Klecany, Czech Republic
| | - Jan Ricny
- Experimental Neurobiology Program, National Institute of Mental Health, Klecany, Czech Republic
| | - Saak V. Ovsepian
- Faculty of Science and Engineering, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, United Kingdom
| |
Collapse
|
10
|
Shimada T, Uehara T, Nagasawa T, Hasegawa M, Maeda Y, Kawasaki Y. A case report of late-onset schizophrenia differentiated from a dementing disorder. Neurocase 2021; 27:467-473. [PMID: 34949153 DOI: 10.1080/13554794.2021.2016858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
We report a case of late-onset schizophrenia that required differentiation from a dementing disorder. The patient was an 83-year-old woman who had experienced auditory hallucinations since she was 67 years old. The patient had slightly elevated total tau and slightly decreased amyloid β1-42, cerebrospinal fluid biomarkers. This case was identified as late-onset schizophrenia. However, the results of cerebrospinal fluid biomarkers indicated that neurofibrillary tangles and neuronal death, which are characteristic of Alzheimer 's disease, may also have been present. Late-onset schizophrenia should be treated based on an appropriate differential diagnosis, including neuropathological consideration of dementing disorders.
Collapse
Affiliation(s)
- Takamitsu Shimada
- Department of Neuropsychiatry, Kanazawa Medical University, Kahoku-county, Ishikawa, Japan.,Department of Psychiatry, Medical Corporation Sekijinkai Okabe Hospital, Kanazawa-City, Ishikawa, Japan
| | - Takashi Uehara
- Department of Neuropsychiatry, Kanazawa Medical University, Kahoku-county, Ishikawa, Japan
| | - Tatsuya Nagasawa
- Department of Neuropsychiatry, Kanazawa Medical University, Kahoku-county, Ishikawa, Japan
| | - Mitsuru Hasegawa
- Department of Psychiatry, Medical Corporation Sekijinkai Okabe Hospital, Kanazawa-City, Ishikawa, Japan
| | - Yoshiki Maeda
- Department of Psychiatry, Medical Corporation Sekijinkai Okabe Hospital, Kanazawa-City, Ishikawa, Japan.,Department of Psychiatry, Medical Corporation Okabe Clinic, Kanazawa-City, Ishikawa, Japan
| | - Yasuhiro Kawasaki
- Department of Neuropsychiatry, Kanazawa Medical University, Kahoku-county, Ishikawa, Japan
| |
Collapse
|
11
|
Zetterberg H, Blennow K. Moving fluid biomarkers for Alzheimer's disease from research tools to routine clinical diagnostics. Mol Neurodegener 2021; 16:10. [PMID: 33608044 PMCID: PMC7893769 DOI: 10.1186/s13024-021-00430-x] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/05/2021] [Indexed: 12/31/2022] Open
Abstract
Four fluid-based biomarkers have been developed into diagnostic tests for Alzheimer’s disease (AD) pathology: the ratio of 42 to 40 amino acid-long amyloid β, a marker of plaque pathology; total-tau and phosphorylated tau, markers of AD-related changes in tau metabolism and secretion; and neurofilament light, a marker of neurodegeneration. When measured in cerebrospinal fluid, these biomarkers can be used in clinical practice to support a diagnosis of mild cognitive impairment or dementia due to AD. Recently, technological breakthroughs have made it possible to measure them in standard blood samples as well. Here, we give an updated account of the current state of the fluid-based AD biomarker research field. We discuss how the new blood tests may be used in research and clinical practice, and what role they may play in relation to more established diagnostic tests, such as CSF biomarkers and amyloid and tau positron emission tomography, to facilitate the effective implementation of future disease-modifying therapies.
Collapse
Affiliation(s)
- Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden. .,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden. .,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK. .,UK Dementia Research Institute at UCL, London, UK.
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden. .,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| |
Collapse
|
12
|
Zetterberg H, Bendlin BB. Biomarkers for Alzheimer's disease-preparing for a new era of disease-modifying therapies. Mol Psychiatry 2021; 26:296-308. [PMID: 32251378 PMCID: PMC8172244 DOI: 10.1038/s41380-020-0721-9] [Citation(s) in RCA: 224] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/17/2020] [Accepted: 03/24/2020] [Indexed: 12/25/2022]
Abstract
Clinical trial results presented in 2019 suggest that antibody-based removal of cerebral amyloid β (Aβ) plaques may possibly clear tau tangles and modestly slow cognitive decline in symptomatic Alzheimer's disease (AD). Although regulatory approval of this approach is still pending, preparing the healthcare system for the advent of disease-modifying therapies against AD is imperative. In particular, it will be necessary to identify the most suitable biomarkers to facilitate appropriate treatment of AD. Here, we give an update on recent developments in fluid and imaging biomarkers for AD-related pathologies and discuss potential approaches that could be adopted to screen for and clarify the underlying pathology in people seeking medical advice because of cognitive symptoms. We succinctly review recent data regarding biomarkers for Aβ and tau pathology, neurodegeneration, synaptic dysfunction, and inflammation, highlight the need for further research into common copathologies, and suggest how different biomarkers could be used (most likely in combination) to facilitate the development and clinical implementation of novel drug candidates against AD.
Collapse
Affiliation(s)
- Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.
- UK Dementia Research Institute at UCL, London, UK.
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
13
|
Di Meco A, Vassar R. Early detection and personalized medicine: Future strategies against Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 177:157-173. [PMID: 33453940 PMCID: PMC8641915 DOI: 10.1016/bs.pmbts.2020.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia and sixth cause of death in elderly adults. AD poses a huge economic burden on society and constitutes an unprecedented challenge for caregivers and families affected. Aging of the population is projected to drastically aggravate the situation in the near future. To date, no therapy is available to prevent or ameliorate the disease. Moreover, several clinical trials for promising therapeutic agents have failed. Lack of supporting biomarker data for pre-symptomatic enrollment and inaccurate stratification of patients based on genetic heterogeneity appear to be contributing factors to this lack of success. Recently, the treatment of cancer has seen enormous advances based on the personalized genetics and biomarkers of the individual patient, forming the foundation of precision medicine for cancer. Likewise, technological progress in AD biomarker research promises the availability of reliable assays for pathology staging on a routine basis relatively soon. Moreover, tremendous achievements in AD genetics and high-throughput genotyping technology allow identification of predisposing risk alleles accurately and on a large scale. Finally, availability of electronic health records (EHR) promises the opportunity to integrate biomarker, genomic and clinical data efficiently. Together, these advances will form the basis of precision medicine for AD.
Collapse
Affiliation(s)
- Antonio Di Meco
- Department of Neurology, Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| | - Robert Vassar
- Department of Neurology, Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| |
Collapse
|
14
|
Arastoo M, Lofthouse R, Penny LK, Harrington CR, Porter A, Wischik CM, Palliyil S. Current Progress and Future Directions for Tau-Based Fluid Biomarker Diagnostics in Alzheimer's Disease. Int J Mol Sci 2020; 21:E8673. [PMID: 33212983 PMCID: PMC7698492 DOI: 10.3390/ijms21228673] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 11/17/2022] Open
Abstract
Despite continued efforts, there remain no disease-modifying drugs approved by the United States Food and Drug Administration (FDA) or European Medicines Agency (EMA) to combat the global epidemic of Alzheimer's disease. Currently approved medicines are unable to delay disease progression and are limited to symptomatic treatment. It is well established that the pathophysiology of this disease remains clinically silent for decades prior to symptomatic clinical decline. Identifying those at risk of disease progression could allow for effective treatment whilst the therapeutic window remains open for preservation of quality of life. This review aims to evaluate critically the current advances in the interpretation of tau-based biomarkers and their use to provide insights into the onset and progression of Alzheimer's disease, whilst highlighting important future directions for the field. This review emphasises the need for a more comprehensive analysis and interrogation of tau within biological fluids, to aid in obtaining a disease specific molecular signature for each stage of Alzheimer's disease. Success in achieving this could provide essential utility for presymptomatic patient selection for clinical trials, monitoring disease progression, and evaluating disease modifying therapies.
Collapse
Affiliation(s)
- Mohammad Arastoo
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZP, UK; (M.A.); (R.L.); (L.K.P.); (C.R.H.); (A.P.); (S.P.)
- Scottish Biologics Facility, University of Aberdeen, Aberdeen AB25 2ZP, UK
| | - Richard Lofthouse
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZP, UK; (M.A.); (R.L.); (L.K.P.); (C.R.H.); (A.P.); (S.P.)
- Scottish Biologics Facility, University of Aberdeen, Aberdeen AB25 2ZP, UK
| | - Lewis K. Penny
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZP, UK; (M.A.); (R.L.); (L.K.P.); (C.R.H.); (A.P.); (S.P.)
- Scottish Biologics Facility, University of Aberdeen, Aberdeen AB25 2ZP, UK
| | - Charles R. Harrington
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZP, UK; (M.A.); (R.L.); (L.K.P.); (C.R.H.); (A.P.); (S.P.)
- Genting TauRx Diagnostic Centre Sdn. Bhd., Aberdeen AB24 5RP, UK
- TauRx Therapeutics Ltd., Aberdeen AB24 5RP, UK
| | - Andy Porter
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZP, UK; (M.A.); (R.L.); (L.K.P.); (C.R.H.); (A.P.); (S.P.)
- Scottish Biologics Facility, University of Aberdeen, Aberdeen AB25 2ZP, UK
| | - Claude M. Wischik
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZP, UK; (M.A.); (R.L.); (L.K.P.); (C.R.H.); (A.P.); (S.P.)
- Genting TauRx Diagnostic Centre Sdn. Bhd., Aberdeen AB24 5RP, UK
- TauRx Therapeutics Ltd., Aberdeen AB24 5RP, UK
| | - Soumya Palliyil
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZP, UK; (M.A.); (R.L.); (L.K.P.); (C.R.H.); (A.P.); (S.P.)
- Scottish Biologics Facility, University of Aberdeen, Aberdeen AB25 2ZP, UK
| |
Collapse
|
15
|
Luchsinger JA, Zetterberg H. Tracking the potential involvement of metabolic disease in Alzheimer's disease-Biomarkers and beyond. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:51-77. [PMID: 32739014 DOI: 10.1016/bs.irn.2020.03.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
There is a vast literature linking systemic metabolic conditions to dementia due to Alzheimer's disease (AD). Advances in in vivo measurements of AD neuropathology using brain imaging, cerebrospinal fluid (CSF), and/or blood biomarkers have led to research in AD that uses in vivo biomarkers as outcomes, focusing primarily on amyloid, tau, and neurodegeneration as constructs. Studies of Type 2 Diabetes Mellitus (T2DM) and AD biomarkers seem to show that T2DM is not related to amyloid deposition, but is related to neurodegeneration and tau deposition. There is a dearth of studies examining adiposity, insulin resistance, and metabolic syndrome in relation to AD biomarkers and the associations in these studies are inconsistent. Metabolomics studies have reported associations of unsaturated fatty acids with AD neuropathology at autopsy, and sphingolipids and glycerophospholipids in relation to neurodegeneration and amyloid and tau. There are other neurodegenerative diseases, such as Lewy body disease that may overlap with AD, and specific biomarkers for these pathologies are being developed and should be integrated into AD biomarker research. More longitudinal studies are needed with concurrent assessment of metabolic factors and AD biomarkers in order to improve the opportunity to assess causality. Ideally, AD biomarkers should be integrated into clinical trials of interventions that affect metabolic factors. Advances in blood-based AD biomarkers, which are less costly and invasive compared with CSF and brain imaging biomarkers, could facilitate widespread implementation of AD biomarkers in studies examining the metabolic contribution to AD.
Collapse
Affiliation(s)
- José A Luchsinger
- Departments of Medicine and Epidemiology, Columbia University Irving Medical Center, New York, NY, United States.
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom; UK Dementia Research Institute at UCL, London, United Kingdom
| |
Collapse
|
16
|
Lewczuk P, Łukaszewicz-Zając M, Mroczko P, Kornhuber J. Clinical significance of fluid biomarkers in Alzheimer's Disease. Pharmacol Rep 2020; 72:528-542. [PMID: 32385624 PMCID: PMC7329803 DOI: 10.1007/s43440-020-00107-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 12/23/2022]
Abstract
The number of patients with Alzheimer's Disease (AD) and other types of dementia disorders has drastically increased over the last decades. AD is a complex progressive neurodegenerative disease affecting about 14 million patients in Europe and the United States. The hallmarks of this disease are neurotic plaques consist of the Amyloid-β peptide (Aβ) and neurofibrillary tangles (NFTs) formed of hyperphosphorylated Tau protein (pTau). Currently, four CSF biomarkers: Amyloid beta 42 (Aβ42), Aβ42/40 ratio, Tau protein, and Tau phosphorylated at threonine 181 (pTau181) have been indicated as core neurochemical AD biomarkers. However, the identification of additional fluid biomarkers, useful in the prognosis, risk stratification, and monitoring of drug response is sorely needed to better understand the complex heterogeneity of AD pathology as well as to improve diagnosis of patients with the disease. Several novel biomarkers have been extensively investigated, and their utility must be proved and eventually integrated into guidelines for use in clinical practice. This paper presents the research and development of CSF and blood biomarkers for AD as well as their potential clinical significance. Upper panel: Aβ peptides are released from transmembrane Amyloid Precursor Protein (APP) under physiological conditions (blue arrow). In AD, however, pathologic accumulation of Aβ monomers leads to their accumulation in plaques (red arrow). This is reflected in decreased concentration of Aβ1-42 and decreased Aβ42/40 concentration ratio in the CSF. Lower panel: Phosphorylated Tau molecules maintain axonal structures; hyperphosphorylation of Tau (red arrow) in AD leads to degeneration of axons, and release of pTau molecules, which then accumulate in neurofibrillary tangles. This process is reflected by increased concentrations of Tau and pTau in the CSF.
Collapse
Affiliation(s)
- Piotr Lewczuk
- Lab for Clinical Neurochemistry and Neurochemical Dementia Diagnostics, Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany.
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, Białystok, Poland.
| | | | - Piotr Mroczko
- Department of Criminal Law and Criminology, Faculty of Law, University of Białystok, Białystok, Poland
| | - Johannes Kornhuber
- Lab for Clinical Neurochemistry and Neurochemical Dementia Diagnostics, Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| |
Collapse
|
17
|
Nguyen TT, Ta QTH, Nguyen TKO, Nguyen TTD, Vo VG. Role of Body-Fluid Biomarkers in Alzheimer's Disease Diagnosis. Diagnostics (Basel) 2020; 10:diagnostics10050326. [PMID: 32443860 PMCID: PMC7277970 DOI: 10.3390/diagnostics10050326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/02/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) is a complex neurodegenerative disease that requires extremely specific biomarkers for its diagnosis. For current diagnostics capable of identifying AD, the development and validation of early stage biomarkers is a top research priority. Body-fluid biomarkers might closely reflect synaptic dysfunction in the brain and, thereby, could contribute to improving diagnostic accuracy and monitoring disease progression, and serve as markers for assessing the response to disease-modifying therapies at early onset. Here, we highlight current advances in the research on the capabilities of body-fluid biomarkers and their role in AD pathology. Then, we describe and discuss current applications of the potential biomarkers in clinical diagnostics in AD.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Faculty of Pharmacy, Ho Chi Minh City University of Technology (HUTECH), Ho Chi Minh City 700000, Vietnam;
| | - Qui Thanh Hoai Ta
- Institute of Research and Development, Duy Tan University, Danang 550000, Vietnam;
| | - Thi Kim Oanh Nguyen
- Faculty of Food Science and Technology, Ho Chi Minh City University of Food Industry, Ho Chi Minh City 700000, Vietnam;
| | - Thi Thuy Dung Nguyen
- Faculty of Environmental and Food Engineering, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
- Correspondence: (T.T.D.N.); (V.G.V.)
| | - Van Giau Vo
- Department of Industrial and Environmental Engineering, Graduate School of Environment, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si, Gyeonggi-do 461-701, Korea
- Department of BionanoTechnology, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si, Gyeonggi-do 461-701, Korea
- Correspondence: (T.T.D.N.); (V.G.V.)
| |
Collapse
|
18
|
Milà-Alomà M, Suárez-Calvet M, Molinuevo JL. Latest advances in cerebrospinal fluid and blood biomarkers of Alzheimer's disease. Ther Adv Neurol Disord 2019; 12:1756286419888819. [PMID: 31897088 PMCID: PMC6920596 DOI: 10.1177/1756286419888819] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and its diagnosis has classically been based on clinical symptoms. Recently, a biological rather than a syndromic definition of the disease has been proposed that is based on biomarkers that reflect neuropathological changes. In AD, there are two main biomarker categories, namely neuroimaging and fluid biomarkers [cerebrospinal fluid (CSF) and blood]. As a complex and multifactorial disease, AD biomarkers are important for an accurate diagnosis and to stage the disease, assess the prognosis, test target engagement, and measure the response to treatment. In addition, biomarkers provide us with information that, even if it does not have a current clinical use, helps us to understand the mechanisms of the disease. In addition to the pathological hallmarks of AD, which include amyloid-β and tau deposition, there are multiple concomitant pathological events that play a key role in the disease. These include, but are not limited to, neurodegeneration, inflammation, vascular dysregulation or synaptic dysfunction. In addition, AD patients often have an accumulation of other proteins including α-synuclein and TDP-43, which may have a pathogenic effect on AD. In combination, there is a need to have biomarkers that reflect different aspects of AD pathogenesis and this will be important in the future to establish what are the most suitable applications for each of these AD-related biomarkers. It is unclear whether sex, gender, or both have an effect on the causes of AD. There may be differences in fluid biomarkers due to sex but this issue has often been neglected and warrants further research. In this review, we summarize the current state of the principal AD fluid biomarkers and discuss the effect of sex on these biomarkers.
Collapse
Affiliation(s)
- Marta Milà-Alomà
- Barcelonaβeta Brain Research Center (BBRC),
Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research
Institute), Barcelona
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC),
Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research
Institute), Barcelona
- Department of Neurology, Hospital del Mar,
Barcelona
| | - José Luís Molinuevo
- Scientific Director, Alzheimer’s Prevention
Program, Barcelonaβeta Brain Research Center, Wellington 30, Barcelona,
08005, Spain
- IMIM (Hospital del Mar Medical Research
Institute), Barcelona
- CIBER Fragilidad y Envejecimiento Saludable,
Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
19
|
Use of the tau protein-to-peptide ratio in CSF to improve diagnostic classification of Alzheimer's disease. CLINICAL MASS SPECTROMETRY 2019; 14 Pt B:74-82. [PMID: 34917763 DOI: 10.1016/j.clinms.2019.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 07/09/2019] [Accepted: 07/09/2019] [Indexed: 11/22/2022]
Abstract
Cerebrospinal fluid (CSF) tau and phospho-tau are well established biomarkers of Alzheimer's disease. While these measures are conventionally referred to as 'total tau' (T-tau) and 'phospho-tau' (P-tau), several truncated and modified tau forms exist that may relay additional diagnostic information. We evaluated the diagnostic performance of an endogenous tau peptide in CSF, tau 175-190, in the phosphorylated and non-phosphorylated state. A liquid chromatography-mass spectrometry (LC-MS) method was established to measure these peptides in CSF and was used to analyze two independent clinical cohorts; the first cohort included patients with Alzheimer's disease (AD, n = 15), Parkinson's disease (PD, n = 15), progressive supranuclear palsy (PSP, n = 15), and healthy controls (n = 15), the second cohort included AD patients (n = 16), and healthy controls (n = 24). In both cohorts T-tau and P-tau concentrations were determined by immunoassay. While tau 175-190 and P-tau 175-190 did not differentiate the study groups, the separation of AD and controls by T-tau (area under the ROC Curve (AUC) = 95%) and P-tau (AUC = 92%) was improved when normalizing the ELISA measurements to the concentrations of the endogenous peptides: T-tau/tau 175-190 (AUC = 100%), P-tau/P-tau 175-190 (AUC = 95%). The separation between patients and controls by T-tau (AUC = 88%) and P-tau (AUC = 82%) was similarly improved in the second cohort by taking the ratios of T-tau/tau 175-190 (AUC = 97%) and P-tau/P-tau 175-190 (AUC = 98%). In conclusion, our results suggest that the performance of the AD biomarkers T-tau and P-tau could be improved by normalizing their measurements to the endogenous peptides tau 175-190 and P-tau 175-190, possibly because these endogenous tau peptides serve to normalize for physiological, and disease-independent, secretion of tau from neurons to the extracellular space and the CSF. Finally, the observations made here add to the general applicability of mass spectrometry as a tool for rapid identification and accurate quantification of biomarker candidates.
Collapse
Key Words
- AD
- AD, Alzheimer’s disease
- AUC, Area under the ROC curve
- Biomarker
- CSF, cerebrospinal fluid
- ELISA, enzyme-linked immunosorbent assay
- Endogenous peptides
- GdnHCl, Guanidinium hydrochloride
- Mass spectrometry
- Microtubule-associated protein tau
- P-tau, phospho-tau protein
- PD, Parkinson’s disease
- PSP, Progressive Supranuclear Palsy
- Peptidomics
- SIL, peptide Synthetic isotope-labelled peptide
- T-tau, total tau protein
Collapse
|
20
|
Cerebrospinal Fluid and Plasma Tau as a Biomarker for Brain Tauopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1184:393-405. [PMID: 32096052 DOI: 10.1007/978-981-32-9358-8_29] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Cerebrospinal fluid (CSF) tau and phosphorylated tau (ptau) are definite biomarkers of Alzheimer's disease (AD). After discovery of presence and increased levels tau in CSF from AD patients using specific ELISA, numerous reports revealed that CSF levels of tau are increased in AD and brain injury, phosphorylated tau are specifically increased in AD. Many large cohort studies also confirmed that natural course of CSF tau and ptau levels initiated from cognitively unimpaired AD stage after longstanding progress of brain Aß amyloidosis. Close correlation with neuroimaging findings of Tau PET and with deterioration of cognitive function domains have been elucidated. CSF tau also increase in neurodegeneration and acute brain injury. Global standardization, assay technology inventions, and research of tau kinetics from brain synthesis and clearance into CSF are developing. Trace amount of plasma p-tau assay are also validated. Development of these studies provide that CSF tau is the biomarker of CNS neurodegeneration and CSF ptau is the specific biomarker of CNS tauopathy. Assays of CSF and plasma tau and ptau are essential tools not only for prediction and diagnosis of AD and but for newly developing disease modified therapies of AD.
Collapse
|
21
|
Koehler D, Shah ZA, Williams FE. The GSK3β inhibitor, TDZD-8, rescues cognition in a zebrafish model of okadaic acid-induced Alzheimer's disease. Neurochem Int 2019; 122:31-37. [PMID: 30392874 PMCID: PMC11956762 DOI: 10.1016/j.neuint.2018.10.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/24/2018] [Accepted: 10/27/2018] [Indexed: 01/01/2023]
Abstract
Currently, no treatments exist that are able to directly treat against Alzheimer's disease (AD), and we are facing an inevitable increase in the near future of the amount of patients who will suffer from AD. Most animal models of AD are limited by not being able to recapitulate the entire pathology of AD. Recently an AD model in zebrafish was established by using the protein phosphatase 2A inhibitor, okadaic acid (OKA). Administering OKA to zebrafish was able to recapitulate most of the neuropathology associated with AD. Therefore, providing a drug discovery model for AD that is also time and cost efficient. This study was designed to investigate the effects of GSK3β inhibition by 4-benzyl-2-methyl-1, 2, 4-thiadiazolidine-3, 5-dione (TDZD-8) on this newly developed AD model. Fish were divided into 4 groups and each group received a different treatment. The fish were divided into a control group, a group treated with 1 μM TDZD-8 only, a group treated with 1 μM TDZD-8 + 100 nM OKA, and a group treated with 100 nM OKA only. Administering the GSK3β inhibitor to zebrafish concomitantly with OKA proved to be protective. TDZD-8 treatment reduced the mortality rate, the ratio of active: inactive GSK3β, pTau (Ser199), and restored PP2A activity. This further corroborates the use of GSKβ inhibitors in the treatment against AD and bolsters the use of the OKA-induced AD-like zebrafish model for drug discovery.
Collapse
Affiliation(s)
- Daniel Koehler
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Frederick E Williams
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
22
|
Chen JA, Fears SC, Jasinska AJ, Huang A, Al‐Sharif NB, Scheibel KE, Dyer TD, Fagan AM, Blangero J, Woods R, Jorgensen MJ, Kaplan JR, Freimer NB, Coppola G. Neurodegenerative disease biomarkers Aβ 1-40, Aβ 1-42, tau, and p-tau 181 in the vervet monkey cerebrospinal fluid: Relation to normal aging, genetic influences, and cerebral amyloid angiopathy. Brain Behav 2018; 8:e00903. [PMID: 29484263 PMCID: PMC5822592 DOI: 10.1002/brb3.903] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 11/19/2017] [Indexed: 01/27/2023] Open
Abstract
Background The Caribbean vervet monkey (Chlorocebus aethiops sabaeus) is a potentially valuable animal model of neurodegenerative disease. However, the trajectory of aging in vervets and its relationship to human disease is incompletely understood. Methods To characterize biomarkers associated with neurodegeneration, we measured cerebrospinal fluid (CSF) concentrations of Aβ1-40, Aβ1-42, total tau, and p-tau181 in 329 members of a multigenerational pedigree. Linkage and genome-wide association were used to elucidate a genetic contribution to these traits. Results Aβ1-40 concentrations were significantly correlated with age, brain total surface area, and gray matter thickness. Levels of p-tau181 were associated with cerebral volume and brain total surface area. Among the measured analytes, only CSF Aβ1-40 was heritable. No significant linkage (LOD > 3.3) was found, though suggestive linkage was highlighted on chromosomes 4 and 12. Genome-wide association identified a suggestive locus near the chromosome 4 linkage peak. Conclusions Overall, these results support the vervet as a non-human primate model of amyloid-related neurodegeneration, such as Alzheimer's disease and cerebral amyloid angiopathy, and highlight Aβ1-40 and p-tau181 as potentially valuable biomarkers of these processes.
Collapse
Affiliation(s)
- Jason A. Chen
- Department of PsychiatryThe Jane and Terry Semel Institute for Neuroscience and Human BehaviorDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Interdepartmental Program in BioinformaticsUniversity of CaliforniaLos AngelesCAUSA
- Verge GenomicsSan FranciscoCAUSA
| | - Scott C. Fears
- Department of PsychiatryThe Jane and Terry Semel Institute for Neuroscience and Human BehaviorDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of PsychiatryGreater Los Angeles Veterans AdministrationLos AngelesCAUSA
| | - Anna J. Jasinska
- Department of PsychiatryThe Jane and Terry Semel Institute for Neuroscience and Human BehaviorDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
| | - Alden Huang
- Department of PsychiatryThe Jane and Terry Semel Institute for Neuroscience and Human BehaviorDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Interdepartmental Program in BioinformaticsUniversity of CaliforniaLos AngelesCAUSA
| | - Noor B. Al‐Sharif
- Department of PsychiatryThe Jane and Terry Semel Institute for Neuroscience and Human BehaviorDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Kevin E. Scheibel
- Department of PsychiatryThe Jane and Terry Semel Institute for Neuroscience and Human BehaviorDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Thomas D. Dyer
- South Texas Diabetes and Obesity InstituteUniversity of Texas Rio Grande Valley School of MedicineBrownsvilleTXUSA
| | - Anne M. Fagan
- Department of NeurologyWashington University in St. LouisSt. LouisMOUSA
| | - John Blangero
- South Texas Diabetes and Obesity InstituteUniversity of Texas Rio Grande Valley School of MedicineBrownsvilleTXUSA
| | - Roger Woods
- Department of PsychiatryThe Jane and Terry Semel Institute for Neuroscience and Human BehaviorDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of NeurologyDavid Geffen School of Medicine at UCLAUniversity of CaliforniaLos AngelesCAUSA
| | - Matthew J. Jorgensen
- Department of PathologySection on Comparative MedicineWake Forest School of MedicineWinston‐SalemNCUSA
| | - Jay R. Kaplan
- Department of PathologySection on Comparative MedicineWake Forest School of MedicineWinston‐SalemNCUSA
| | - Nelson B. Freimer
- Department of PsychiatryThe Jane and Terry Semel Institute for Neuroscience and Human BehaviorDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Giovanni Coppola
- Department of PsychiatryThe Jane and Terry Semel Institute for Neuroscience and Human BehaviorDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of NeurologyDavid Geffen School of Medicine at UCLAUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
23
|
Ba M, Li X, Ng KP, Pascoal TA, Mathotaarachchi S, Rosa-Neto P, Gauthier S, Alzheimer's Disease Neuroimaging Initiative. The prevalence and biomarkers' characteristic of rapidly progressive Alzheimer's disease from the Alzheimer's Disease Neuroimaging Initiative database. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2017; 3:107-113. [PMID: 29067322 PMCID: PMC5651370 DOI: 10.1016/j.trci.2016.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The prevalence and detailed biomarkers' characteristic of rapidly progressive Alzheimer's disease (rpAD) remain incompletely understood. METHODS A total of 312 mild AD patients from the Alzheimer's Disease Neuroimaging Initiative database were chosen and dichotomized into rpAD and non-rpAD groups. We performed the prevalence and comprehensive biomarker evaluation. RESULTS The prevalence of rpAD was 17.6% in mild AD. Compared with non-rpAD, there were no differences in APOE ε4/ε4, APOE ε3/ε4, and APOE ε2/ε4 genotype distribution, cerebrospinal fluid tau, phosphorylated tau (p-tau), amyloid-β, hippocampus volume, and amyloid deposition in rpAD. Yet, a lower p-tau/tau ratio was observed in rpAD (P = .04). rpAD showed region-specific hypometabolism ([18F]fluorodeoxyglucose-positron emission tomography [FDG-PET]) (P = .001). Receiver-operating characteristic analysis of FDG-PET demonstrated that left angular and left temporal cortices were the regions with higher area under the curve and predictive value for identifying clinical at-risk rpAD. DISCUSSION We identified that rpAD commonly existed in mild AD. Cerebral hypometabolism could provide potential clinical differential value for rpAD in the short-term follow-up period.
Collapse
Affiliation(s)
- Maowen Ba
- Department of Neurology, Yantai Yuhuangding Hospital Affiliated to Qingdao Medical University, Shandong, People’s Republic of China
- Alzheimer's Disease Research Unit, McGill Centre for Studies in Aging, McGill University, Douglas Institute, Montreal, Quebec, Canada
| | - Xiaofeng Li
- Alzheimer's Disease Research Unit, McGill Centre for Studies in Aging, McGill University, Douglas Institute, Montreal, Quebec, Canada
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Kok Pin Ng
- Alzheimer's Disease Research Unit, McGill Centre for Studies in Aging, McGill University, Douglas Institute, Montreal, Quebec, Canada
- Department of Neurology, National Neuroscience Institute, Singapore
| | - Tharick A. Pascoal
- Alzheimer's Disease Research Unit, McGill Centre for Studies in Aging, McGill University, Douglas Institute, Montreal, Quebec, Canada
| | - Sulantha Mathotaarachchi
- Alzheimer's Disease Research Unit, McGill Centre for Studies in Aging, McGill University, Douglas Institute, Montreal, Quebec, Canada
| | - Pedro Rosa-Neto
- Alzheimer's Disease Research Unit, McGill Centre for Studies in Aging, McGill University, Douglas Institute, Montreal, Quebec, Canada
| | - Serge Gauthier
- Alzheimer's Disease Research Unit, McGill Centre for Studies in Aging, McGill University, Douglas Institute, Montreal, Quebec, Canada
| | | |
Collapse
|
24
|
Kincses ZT, Király A, Veréb D, Vécsei L. Structural Magnetic Resonance Imaging Markers of Alzheimer's Disease and Its Retranslation to Rodent Models. J Alzheimers Dis 2016; 47:277-90. [PMID: 26401552 DOI: 10.3233/jad-143195] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The importance of imaging biomarkers has been acknowledged in the diagnosis and in the follow-up of Alzheimer's disease (AD), one of the major causes of dementia. Next to the molecular biomarkers and PET imaging investigations, structural MRI approaches provide important information about the disease progression and about the pathomechanism. Furthermore,a growing body of literature retranslates these imaging biomarkers to various rodent models of the disease. The goal of this review is to provide an overview of the macro- and microstructural imaging biomarkers of AD, concentrating on atrophy measures and diffusion MRI alterations. A survey is also given of the imaging approaches used in rodent models of dementias that can promote drug development.
Collapse
Affiliation(s)
- Zsigmond Tamas Kincses
- Department of Neurology, University of Szeged, Szeged, Hungary.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - András Király
- Department of Neurology, University of Szeged, Szeged, Hungary
| | - Dániel Veréb
- Department of Neurology, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, University of Szeged, Szeged, Hungary.,MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| |
Collapse
|
25
|
RNA Interference Silencing of Glycogen Synthase Kinase 3β Inhibites Tau Phosphorylation in Mice with Alzheimer Disease. Neurochem Res 2016; 41:2470-80. [DOI: 10.1007/s11064-016-1960-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 04/27/2016] [Accepted: 05/17/2016] [Indexed: 12/22/2022]
|
26
|
Abnormal tau induces cognitive impairment through two different mechanisms: synaptic dysfunction and neuronal loss. Sci Rep 2016; 6:20833. [PMID: 26888634 PMCID: PMC4757872 DOI: 10.1038/srep20833] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 01/08/2016] [Indexed: 01/26/2023] Open
Abstract
The hyperphosphorylated microtubule-associated protein tau is present in several neurodegenerative diseases, although the causal relationship remains elusive. Few mouse models used to study Alzheimer-like dementia target tau phosphorylation. We created an inducible pseudophosphorylated tau (Pathological Human Tau, PH-Tau) mouse model to study the effect of conformationally modified tau in vivo. Leaky expression resulted in two levels of PH-Tau: low basal level and higher upon induction (4% and 14% of the endogenous tau, respectively). Unexpectedly, low PH-Tau resulted in significant cognitive deficits, decrease in the number of synapses (seen by EM in the CA1 region), reduction of synaptic proteins, and localization to the nucleus. Induction of PH-Tau triggered neuronal death (60% in CA3), astrocytosis, and loss of the processes in CA1. These findings suggest, that phosphorylated tau is sufficient to induce neurodegeneration and that two different mechanisms can induce cognitive impairment depending on the levels of PH-Tau expression.
Collapse
|
27
|
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
|
28
|
Mishima A, Nihashi T, Ando Y, Kawai H, Kato T, Ito K, Terasawa T. Biomarkers Differentiating Dementia with Lewy Bodies from Other Dementias: A Meta-Analysis. J Alzheimers Dis 2015; 50:161-74. [DOI: 10.3233/jad-150675] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Aki Mishima
- Section of General Internal Medicine, Department of Emergency and General Internal Medicine, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Takashi Nihashi
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yoshio Ando
- Department of Radiology, Toyota Memorial Hospital, Toyota, Aichi, Japan
| | - Hisashi Kawai
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Takashi Kato
- Department of Radiology, Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kengo Ito
- Department of Radiology, Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Teruhiko Terasawa
- Section of General Internal Medicine, Department of Emergency and General Internal Medicine, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
- Center for Clinical Evidence Synthesis, Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
29
|
Ishiki A, Okamura N, Furukawa K, Furumoto S, Harada R, Tomita N, Hiraoka K, Watanuki S, Ishikawa Y, Tago T, Funaki Y, Iwata R, Tashiro M, Yanai K, Kudo Y, Arai H. Longitudinal Assessment of Tau Pathology in Patients with Alzheimer's Disease Using [18F]THK-5117 Positron Emission Tomography. PLoS One 2015; 10:e0140311. [PMID: 26461913 PMCID: PMC4604169 DOI: 10.1371/journal.pone.0140311] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 09/24/2015] [Indexed: 01/08/2023] Open
Abstract
The formation of neurofibrillary tangles is believed to contribute to the neurodegeneration observed in Alzheimer’s disease (AD). Postmortem studies have shown strong associations between the neurofibrillary pathology and both neuronal loss and the severity of cognitive impairment. However, the temporal changes in the neurofibrillary pathology and its association with the progression of the disease are not well understood. Tau positron emission tomography (PET) imaging is expected to be useful for the longitudinal assessment of neurofibrillary pathology in the living brain. Here, we performed a longitudinal PET study using the tau-selective PET tracer [18F]THK-5117 in patients with AD and in healthy control subjects. Annual changes in [18F]THK-5117 binding were significantly elevated in the middle and inferior temporal gyri and in the fusiform gyrus of patients with AD. Compared to patients with mild AD, patients with moderate AD showed greater changes in the tau load that were more widely distributed across the cortical regions. Furthermore, a significant correlation was observed between the annual changes in cognitive decline and regional [18F]THK-5117 binding. These results suggest that the cognitive decline observed in patients with AD is attributable to the progression of neurofibrillary pathology. Longitudinal assessment of tau pathology will contribute to the assessment of disease progression and treatment efficacy.
Collapse
Affiliation(s)
- Aiko Ishiki
- Department of Geriatrics and Gerontology, Institute of Development, Aging and Cancer, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Nobuyuki Okamura
- Department of Pharmacology, Tohoku University School of Medicine, Sendai city, Miyagi prefecture, Japan; Division of Neuro-imaging, Institute of Development, Aging and Cancer, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Katsutoshi Furukawa
- Department of Geriatrics and Gerontology, Institute of Development, Aging and Cancer, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Shozo Furumoto
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Ryuichi Harada
- Division of Neuro-imaging, Institute of Development, Aging and Cancer, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Naoki Tomita
- Department of Geriatrics and Gerontology, Institute of Development, Aging and Cancer, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Kotaro Hiraoka
- Division of Cyclotron Nuclear Medicine, Cyclotron and Radioisotope Center, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Shoichi Watanuki
- Division of Cyclotron Nuclear Medicine, Cyclotron and Radioisotope Center, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Yoichi Ishikawa
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Tetsuro Tago
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Yoshihito Funaki
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Ren Iwata
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Manabu Tashiro
- Division of Cyclotron Nuclear Medicine, Cyclotron and Radioisotope Center, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University School of Medicine, Sendai city, Miyagi prefecture, Japan
| | - Yukitsuka Kudo
- Division of Neuro-imaging, Institute of Development, Aging and Cancer, Tohoku University, Sendai city, Miyagi prefecture, Japan; Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai city, Miyagi prefecture, Japan
| | - Hiroyuki Arai
- Department of Geriatrics and Gerontology, Institute of Development, Aging and Cancer, Tohoku University, Sendai city, Miyagi prefecture, Japan
| |
Collapse
|
30
|
Babić M, Svob Štrac D, Mück-Šeler D, Pivac N, Stanić G, Hof PR, Simić G. Update on the core and developing cerebrospinal fluid biomarkers for Alzheimer disease. Croat Med J 2015; 55:347-65. [PMID: 25165049 PMCID: PMC4157375 DOI: 10.3325/cmj.2014.55.347] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Alzheimer disease (AD) is a complex neurodegenerative disorder, whose prevalence will dramatically rise by 2050. Despite numerous clinical trials investigating this disease, there is still no effective treatment. Many trials showed negative or inconclusive results, possibly because they recruited only patients with severe disease, who had not undergone disease-modifying therapies in preclinical stages of AD before severe degeneration occurred. Detection of AD in asymptomatic at risk individuals (and a few presymptomatic individuals who carry an autosomal dominant monogenic AD mutation) remains impractical in many of clinical situations and is possible only with reliable biomarkers. In addition to early diagnosis of AD, biomarkers should serve for monitoring disease progression and response to therapy. To date, the most promising biomarkers are cerebrospinal fluid (CSF) and neuroimaging biomarkers. Core CSF biomarkers (amyloid β1-42, total tau, and phosphorylated tau) showed a high diagnostic accuracy but were still unreliable for preclinical detection of AD. Hence, there is an urgent need for detection and validation of novel CSF biomarkers that would enable early diagnosis of AD in asymptomatic individuals. This article reviews recent research advances on biomarkers for AD, focusing mainly on the CSF biomarkers. In addition to core CSF biomarkers, the potential usefulness of novel CSF biomarkers is discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Goran Simić
- Goran Šimić, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000 Zagreb, Croatia,
| |
Collapse
|
31
|
Harada R, Okamura N, Furumoto S, Furukawa K, Ishiki A, Tomita N, Hiraoka K, Watanuki S, Shidahara M, Miyake M, Ishikawa Y, Matsuda R, Inami A, Yoshikawa T, Tago T, Funaki Y, Iwata R, Tashiro M, Yanai K, Arai H, Kudo Y. [(18)F]THK-5117 PET for assessing neurofibrillary pathology in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2015; 42:1052-61. [PMID: 25792456 DOI: 10.1007/s00259-015-3035-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/03/2015] [Indexed: 11/25/2022]
Abstract
PURPOSE Visualization of the spatial distribution of neurofibrillary tangles would help in the diagnosis, prevention and treatment of dementia. The purpose of the study was to evaluate the clinical utility of [(18)F]THK-5117 as a highly selective tau imaging radiotracer. METHODS We initially evaluated in vitro binding of [(3)H]THK-5117 in post-mortem brain tissues from patients with Alzheimer's disease (AD). In clinical PET studies, [(18)F]THK-5117 retention in eight patients with AD was compared with that in six healthy elderly controls. Ten subjects underwent an additional [(11)C]PiB PET scan within 2 weeks. RESULTS In post-mortem brain samples, THK-5117 bound selectively to neurofibrillary deposits, which differed from the binding target of PiB. In clinical PET studies, [(18)F]THK-5117 binding in the temporal lobe clearly distinguished patients with AD from healthy elderly subjects. Compared with [(11)C]PiB, [(18)F]THK-5117 retention was higher in the medial temporal cortex. CONCLUSION These findings suggest that [(18)F]THK-5117 provides regional information on neurofibrillary pathology in living subjects.
Collapse
Affiliation(s)
- Ryuichi Harada
- Division of Neuro-imaging, Institute of Development, Aging and Cancer, Tohoku University, Sendai, 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Lewczuk P, Mroczko B, Fagan A, Kornhuber J. Biomarkers of Alzheimer's disease and mild cognitive impairment: a current perspective. Adv Med Sci 2015; 60:76-82. [PMID: 25579841 DOI: 10.1016/j.advms.2014.11.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/12/2014] [Accepted: 11/28/2014] [Indexed: 11/16/2022]
Abstract
A growing body of evidence supports the application of the neurochemical dementia diagnostics (NDD) biomarkers for the diagnosis of dementing conditions. Biomarkers of Alzheimer's disease (AD) were recently classified as these reflecting amyloid β pathology (decreased CSF concentrations of Aβ42 and/or positive Aβ PET scan) and these reflecting neurodegeneration (increased CSF Tau concentrations, decreased uptake of FDG on FDG-PET, and cerebral atrophy on structural MRI). Particularly important seems the role of the biomarkers in the early diagnosis of AD, as the first pathophysiologic events observable in the CSF and amyloid β-PET occur years and perhaps decades before the onset of the earliest clinical symptoms. Therefore, the NDD tools enable the diagnosis of AD already in the early preclinical stage. This review summarizes pathophysiology underlying the CSF biomarkers, following a discussion of their role in the current guidelines for the diagnostic procedures.
Collapse
Affiliation(s)
- Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok, Poland; Department of Biochemical Diagnostics, University Hospital in Bialystok, Bialystok, Poland
| | - Anne Fagan
- The Knight Alzheimer's Disease Research Center, Department of Neurology, Washington University, St. Louis, MO, USA
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
33
|
Cavaleri F. Paradigm shift redefining molecular, metabolic and structural events in Alzheimer's disease involves a proposed contribution by transition metals. Defined lengthy preclinical stage provides new hope to circumvent advancement of disease- and age-related neurodegeneration. Med Hypotheses 2015; 84:460-9. [PMID: 25691377 DOI: 10.1016/j.mehy.2015.01.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 01/30/2015] [Indexed: 12/28/2022]
Abstract
It is estimated that 5.5 Million North Americans suffer from varying degrees of Alzheimer's disease (AD) and by the year 2050 it may be one in 85 people globally (100 Million). It will be shown that heavy metal toxicity plays a significant role in sporadic AD. Although current literature speaks to involvement of metal ions (via Fenton reaction), studies and reviewers have yet to link cellular events including known structural changes such as amyloid plaque development to this metal toxicity the way it is proposed here. Contrary to the current AD model which positions BACE1 (β-secretase) as an aberrant or AD-advancing enzyme, it is proposed herein that the neuron's protective counteraction to this metal toxicity is, in fact, a justified increase in BACE1 activity and amyloid precursor protein (APP) processing to yield more secreted APP (sAPP) and β-amyloid peptide in response to metal toxicity. This new perspective which justifies a functional role for APP, BACE1 enzyme activity and the peptide products from this activity may at first appear to be counterintuitive. Compelling evidence, however, is presented and a mechanism is shown herein that validate BACE1 recruitment and the resulting β-amyloid protein as strategic countermeasures serving the cell effectively against neuro-impeding disease. It is proposed that β-amyloid peptide chelates and sequesters free heavy metals in the extracellular medium to aggregate as amyloid plaque while unchelated β-amyloid migrates across the cell membrane to chelate intracellular free divalent metals. The sequestered intracellular metal is subsequently chaperoned as a metallo-peptide to cross the plasma membrane and aggregate as amyloid plaques extracellularly. The BACE1 countermeasure is not genetic or metabolic aberration; and this novel conclusion demonstrates that it must not be inhibited as currently targeted. APP, BACE1, β-amyloid peptide, and sAPP play positive roles against the preclinical oxidative load that predates AD symptoms for as long as 20 years. A healthy neuron may tolerate free metal toxicity, such as iron in the case of injury-induced amyloid, for as long as twenty years due to this very BACE1 activity. In later stages, the uncontrolled metals and ROS are compounded by other factors which together overcome this BACE1/β-amyloid protein countermeasure. This results in a sudden increase in IL-1 leading to Tau's hyperphosphorylation as cited and eventually to Tau dissociation from the microtubule cytoskeleton interrupting cell trafficking. At this later stage of AD the β-amyloid protein which once served as a vehicle to escort toxic metals to the extracellular medium and a trap to form a relatively benign extraneuronal disposal site is no longer translocated due to interruption of trafficking and now accumulates intracellularly facilitating hyper-oxidative ROS levels and contributes to irreversible neuron apoptosis.
Collapse
Affiliation(s)
- Franco Cavaleri
- Brain Research Center, UBC Hospital, 2211 Wesbrook Mall, Vancouver, BC V6T 2B5, Canada.
| |
Collapse
|
34
|
Schade S, Mollenhauer B. Biomarkers in biological fluids for dementia with Lewy bodies. ALZHEIMERS RESEARCH & THERAPY 2014; 6:72. [PMID: 25478030 PMCID: PMC4255553 DOI: 10.1186/s13195-014-0072-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dementia with Lewy bodies (DLB) has become the second most common neurodegenerative dementia due to demographic ageing. Differential diagnosis is still troublesome especially in early stages of the disease, since there is a great clinical and neuropathological overlap primarily with Alzheimer's disease and Parkinson's disease. Therefore, more specific biomarkers, not only for scientific reasons but also for clinical therapeutic decision-making, are urgently needed. In this review, we summarize the knowledge on fluid biomarkers for DLB, derived predominantly from cerebrospinal fluid. We discuss the value of well-defined markers (β-amyloid, (phosphorylated) tau, α-synuclein) as well as some promising 'upcoming' substances, which still have to be further evaluated.
Collapse
Affiliation(s)
- Sebastian Schade
- Paracelsus-Elena-Klinik, Klinikstraße 16, Kassel, D-34128, Germany ; Department of Clinical Neurophysiology, University Medical Center, Georg-August University, Robert-Koch Straße 40, Göttingen, 37075, Germany
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Klinikstraße 16, Kassel, D-34128, Germany ; Department of Neurosurgery, University Medical Center, Georg-August University, Robert-Koch Straße 40, Göttingen, 37075, Germany ; Department of Neuropathology, University Medical Center, Georg-August University, Robert-Koch Straße 40, Göttingen, 37075, Germany
| |
Collapse
|
35
|
Inoue H, Hiradate Y, Shirakata Y, Kanai K, Kosaka K, Gotoh A, Fukuda Y, Nakai Y, Uchida T, Sato E, Tanemura K. Site-specific phosphorylation of Tau protein is associated with deacetylation of microtubules in mouse spermatogenic cells during meiosis. FEBS Lett 2014; 588:2003-8. [DOI: 10.1016/j.febslet.2014.04.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 03/26/2014] [Accepted: 04/10/2014] [Indexed: 11/26/2022]
|
36
|
Cerebrospinal fluid biomarkers of Alzheimer's disease. Neurosci Bull 2014; 30:233-42. [PMID: 24733653 DOI: 10.1007/s12264-013-1412-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 01/23/2014] [Indexed: 10/25/2022] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disorder that takes about a decade to develop, making early diagnosis possible. Clinically, the diagnosis of AD is complicated, costly, and inaccurate, so it is urgent to find specific biomarkers. Due to its multifactorial nature, a panel of biomarkers for the multiple pathologies of AD, such as cerebral amyloidogenesis, neuronal dysfunction, synapse loss, oxidative stress, and inflammation, are most promising for accurate diagnosis. Highly sensitive and high-throughput proteomic techniques can be applied to develop a panel of novel biomarkers for AD. In this review, we discuss the metabolism and diagnostic performance of the well-established core candidate cerebrospinal fluid (CSF) biomarkers (β-amyloid, total tau, and hyperphosphorylated tau). Meanwhile, novel promising CSF biomarkers, especially those identified by proteomics, updated in the last five years are also extensively discussed. Furthermore, we provide perspectives on how biomarker discovery for AD is evolving.
Collapse
|
37
|
Liu B, Tang Y, Shen Y, Cen L, Han M. Cerebrospinal fluid τ protein in differential diagnosis of Alzheimer's disease and vascular dementia in Chinese population: a meta-analysis. Am J Alzheimers Dis Other Demen 2014; 29:116-22. [PMID: 24164930 PMCID: PMC10852876 DOI: 10.1177/1533317513507374] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND To assess whether biomarker τ protein could differentiate between Alzheimer's disease (AD) and vascular dementia (VaD). METHODS We conducted a comprehensive search to identify studies on τ protein, patients with AD, and patients with VaD. Cerebrospinal fluid (CSF) τ protein levels were compared to discriminate among patients with AD, healthy controls, and patients with VaD by a meta-analysis. RESULTS Patients with AD exhibit significantly higher CSF τ protein levels than healthy controls or patients with VaD in the Chinese population. CONCLUSION Our findings suggested that CSF τ protein levels were found to be significantly associated with AD in the Chinese population. Measurement of τ protein could help in attenuating the strict distinction between AD and VaD.
Collapse
Affiliation(s)
- Bo Liu
- The Cadre Ward in Department of Neurology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Yulan Tang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuefei Shen
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Luan Cen
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Min Han
- The Cadre Ward in Department of Neurology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| |
Collapse
|
38
|
Schmidt O, Schulenborg T, Meyer HE, Marcus K, Hamacher M. How proteomics reveals potential biomarkers in brain diseases. Expert Rev Proteomics 2014; 2:901-13. [PMID: 16307519 DOI: 10.1586/14789450.2.6.901] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The brain is complex, and so are the proteomics studies of brain tissue and its diseases, including Alzheimer's Disease, Parkinson's Disease and schizophrenia. In this review, general considerations and strategies of proteomics technologies, the advantages and challenges as well as the special needs for brain tissue are described and summarized. In addition, the results of the first studies are presented including a quality evaluation of the candidate proteins for these diseases. A paragraph is dedicated to the efforts of standardization in this field.
Collapse
Affiliation(s)
- Oliver Schmidt
- Ruhr-Universitaet Bochum, Medizinisches Proteom-Center, ZKF E.141, Universitaetsstrasse 150, D-44801 Bochum, Germany.
| | | | | | | | | |
Collapse
|
39
|
Chiu MJ, Yang SY, Horng HE, Yang CC, Chen TF, Chieh JJ, Chen HH, Chen TC, Ho CS, Chang SF, Liu HC, Hong CY, Yang HC. Combined plasma biomarkers for diagnosing mild cognition impairment and Alzheimer's disease. ACS Chem Neurosci 2013; 4:1530-6. [PMID: 24090201 DOI: 10.1021/cn400129p] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A highly sensitive immunoassay, the immunomagnetic reduction, is used to measure several biomarkers for plasma that is related to Alzheimer's disease (AD). These biomarkers include Aβ-40, Aβ-42, and tau proteins. The samples are composed of four groups: healthy controls (n=66), mild cognitive impairment (MCI, n=22), very mild dementia (n=23), and mild-to-serve dementia, all due to AD (n=22). It is found that the concentrations of both Aβ-42 and tau protein for the healthy controls are significantly lower than those of all of the other groups. The sensitivity and the specificity of plasma Aβ-42 and tau protein in differentiating MCI from AD are all around 0.9 (0.88-0.97). However, neither plasma Aβ-42 nor tau-protein concentration is an adequate parameter to distinguish MCI from AD. A parameter is proposed, which is the product of plasma Aβ-42 and tau-protein levels, to differentiate MCI from AD. The sensitivity and specificity are found to be 0.80 and 0.82, respectively. It is concluded that the use of combined plasma biomarkers not only allows the differentiation of the healthy controls and patients with AD in both the prodromal phase and the dementia phase, but it also allows AD in the prodromal phase to be distinguished from that in the dementia phase.
Collapse
Affiliation(s)
- 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
| | - Shieh-Yueh Yang
- Institute
of Electro-optical Science and Technology, National Taiwan Normal University, Taipei 116, Taiwan
- MagQu Co., Ltd., Xindian District, New Taipei
City 231, Taiwan
| | - Herng-Er Horng
- Institute
of Electro-optical Science and Technology, National Taiwan Normal University, Taipei 116, Taiwan
| | - Che-Chuan Yang
- MagQu Co., Ltd., Xindian District, New Taipei
City 231, Taiwan
| | - Ta-Fu Chen
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Jen-Je Chieh
- Institute
of Electro-optical Science and Technology, National Taiwan Normal University, Taipei 116, Taiwan
| | - Hsin-Hsien Chen
- Institute
of Electro-optical Science and Technology, National Taiwan Normal University, Taipei 116, Taiwan
| | - Ting-Chi Chen
- MagQu Co., Ltd., Xindian District, New Taipei
City 231, Taiwan
| | - Chia-Shin Ho
- MagQu Co., Ltd., Xindian District, New Taipei
City 231, Taiwan
| | - Shuo-Fen Chang
- MagQu Co., Ltd., Xindian District, New Taipei
City 231, Taiwan
| | - Hao Chun Liu
- Institute
of Electro-optical Science and Technology, National Taiwan Normal University, Taipei 116, Taiwan
| | - Chin-Yih Hong
- Graduate
Institute of Bio-medical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - Hong-Chang Yang
- Department
of Electro-optical Engineering, Kun Shan University, Yongkang District, Tainan City 710, Taiwan
| |
Collapse
|
40
|
Characterization of novel CSF Tau and ptau biomarkers for Alzheimer's disease. PLoS One 2013; 8:e76523. [PMID: 24116116 PMCID: PMC3792042 DOI: 10.1371/journal.pone.0076523] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 08/31/2013] [Indexed: 12/02/2022] Open
Abstract
Cerebral spinal fluid (CSF) Aβ42, tau and p181tau are widely accepted biomarkers of Alzheimer’s disease (AD). Numerous studies show that CSF tau and p181tau levels are elevated in mild-to-moderate AD compared to age-matched controls. In addition, these increases might predict preclinical AD in cognitively normal elderly. Despite their importance as biomarkers, the molecular nature of CSF tau and ptau is not known. In the current study, reverse-phase high performance liquid chromatography was used to enrich and concentrate tau prior to western-blot analysis. Multiple N-terminal and mid-domain fragments of tau were detected in pooled CSF with apparent sizes ranging from <20 kDa to ~40 kDa. The pattern of tau fragments in AD and control samples were similar. In contrast, full-length tau and C-terminal-containing fragments were not detected. To quantify levels, five tau ELISAs and three ptau ELISAs were developed to detect different overlapping regions of the protein. The discriminatory potential of each assay was determined using 20 AD and 20 age-matched control CSF samples. Of the tau ELISAs, the two assays specific for tau containing N-terminal sequences, amino acids 9-198 (numbering based on tau 441) and 9-163, exhibited the most significant differences between AD and control samples. In contrast, CSF tau was not detected with an ELISA specific for a more C-terminal region (amino acids 159-335). Significant discrimination was also observed with ptau assays measuring amino acids 159-p181 and 159-p231. Interestingly, the discriminatory potential of p181 was reduced when measured in the context of tau species containing amino acids 9-p181. Taken together, these results demonstrate that tau in CSF occurs as a series of fragments and that discrimination of AD from control is dependent on the subset of tau species measured. These assays provide novel tools to investigate CSF tau and ptau as biomarkers for other neurodegenerative diseases.
Collapse
|
41
|
Kandimalla RJL, Prabhakar S, Wani WY, Kaushal A, Gupta N, Sharma DR, Grover VK, Bhardwaj N, Jain K, Gill KD. CSF p-Tau levels in the prediction of Alzheimer's disease. Biol Open 2013; 2:1119-24. [PMID: 24244848 PMCID: PMC3828758 DOI: 10.1242/bio.20135447] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/09/2013] [Indexed: 11/20/2022] Open
Abstract
The two hallmarks of Alzheimer's disease (AD) are neurofibrillary tangles and amyloid plaques. Neurofibrillary tangles are formed due to the hyperphosphorylation of tau protein. There is an urgent need to develop a reliable biomarker for the diagnosis of AD. Cerebrospinal fluid (CSF) is surrounding the brain and reflects the major neuropathological features in the AD brain. Diagnosis, disease progression and drug actions rely on the AD biomarkers. Mainly CSF tau and phosphorylated tau (p-Tau) have been observed to serve the purpose for early AD. Keeping in view the early appearance of p-Tau in CSF, we analyzed p-Tau levels in 23 AD, 23 Non AD type dementia (NAD), 23 Neurological control (NC) and 23 Healthy control (HC) North Indian patients. The levels of p-Tau were found to be increased in AD patients (67.87±18.05 pg/ml, SEM 3.76) compared with NAD (47.55±7.85 pg/ml, SEM 1.64), NC (34.42±4.51 pg/ml, SEM 0.94) and HC (27.09±7.18 pg/ml, SEM 1.50). The resulting sensitivity for AD with NAD was 80.27% whereas with respect to the NAD, NC and HC was 85.40%. Therefore elevated levels of p-Tau in AD can be exploited as a predictive biomarker in North Indian AD patients.
Collapse
Affiliation(s)
- Ramesh J L Kandimalla
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research , Chandigarh , India 160 012
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Randall C, Mosconi L, de Leon M, Glodzik L. Cerebrospinal fluid biomarkers of Alzheimer's disease in healthy elderly. FRONT BIOSCI-LANDMRK 2013; 18:1150-73. [PMID: 23747874 PMCID: PMC3904672 DOI: 10.2741/4170] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Numerous studies have shown that Alzheimer's Disease (AD) pathology begins before the onset of clinical symptoms. Because therapies are likely to be more effective if they are implemented early in the disease progression, it is necessary to identify reliable biomarkers to detect AD pathology in the early stages of the disease, ideally in presymptomatic individuals. Recent research has identified three candidate cerebrospinal fluid (CSF) biomarkers that reflect AD pathology: amyloid beta, total tau protein (t-tau), and tau protein phosphorylated at AD-specific epitopes (p-tau). They are useful in supporting the AD diagnosis and have predictive value for AD when patients are in the stage of mild cognitive impairment (MCI). However, their predictive utility in cognitively healthy subjects is still being evaluated. We conducted a review of studies published between 1993 and 2011 and summarized their findings on the role of CSF biomarkers for AD in healthy elderly.
Collapse
Affiliation(s)
- Catherine Randall
- Center for Brain Health, 145 East 32nd Street, 5th floor. New York, NY 10016
| | - Lisa Mosconi
- Center for Brain Health, 145 East 32nd Street, 5th floor. New York, NY 10016
| | - Mony de Leon
- Center for Brain Health, 145 East 32nd Street, 5th floor. New York, NY 10016
| | - Lidia Glodzik
- Center for Brain Health, 145 East 32nd Street, 5th floor. New York, NY 10016
| |
Collapse
|
43
|
Ikeda M, Yonemura K, Kakuda S, Tashiro Y, Fujita Y, Takai E, Hashimoto Y, Makioka K, Furuta N, Ishiguro K, Maruki R, Yoshida J, Miyaguchi O, Tsukie T, Kuwano R, Yamazaki T, Yamaguchi H, Amari M, Takatama M, Harigaya Y, Okamoto K. Cerebrospinal fluid levels of phosphorylated tau and Aβ1-38/Aβ1-40/Aβ1-42 in Alzheimer's disease with PS1 mutations. Amyloid 2013; 20:107-12. [PMID: 23638752 DOI: 10.3109/13506129.2013.790810] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We studied seven cases of Alzheimer's disease (AD). Six of the patients had presenilin 1 (PS1) mutations (PS1AD). Three novel PS1 mutations (T99A, H131R and L219R) and three other missense mutations (M233L, H163R and V272A) were found in the PS1AD group. We measured the levels of phosphorylated tau (ptau-181, ptau-199) and Aβ (Aβ1-42, Aβ1-40 and Aβ1-38) in the cerebrospinal fluid (CSF) of PS1AD patients, early-onset sporadic AD (EOSAD), late-onset sporadic AD (LOSAD) and non-demented subjects (ND). The CSF levels of Aβ1-42 in the three AD groups were significantly lower than those of the ND group (p < 0.0001). CSF levels of Aβ1-42 in the PS1AD group were significantly lower than those in the two sporadic AD groups. The Aβ1-40 and Aβ1-38 levels in the CSF of the PS1AD group were significantly lower than those of the three other groups (p < 0.0001, respectively). The levels of Aβ1-40, Aβ1-38 and Aβ1-42 in the CSF of the PS1AD group remained lower than those of the ND group for 4 years. Not only CSF Aβ1-42, but also Aβ1-40 and Aβ1-38 decreased in the advanced stages of PS1AD.
Collapse
Affiliation(s)
- Masaki Ikeda
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Alzheimer's disease biomarkers: correspondence between human studies and animal models. Neurobiol Dis 2013; 56:116-30. [PMID: 23631871 DOI: 10.1016/j.nbd.2013.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/11/2013] [Accepted: 04/18/2013] [Indexed: 01/05/2023] Open
Abstract
Alzheimer's disease (AD) represents an escalating global threat as life expectancy and disease prevalence continue to increase. There is a considerable need for earlier diagnoses to improve clinical outcomes. Fluid biomarkers measured from cerebrospinal fluid (CSF) and blood, or imaging biomarkers have considerable potential to assist in the diagnosis and management of AD. An additional important utility of biomarkers is in novel therapeutic development and clinical trials to assess efficacy and side effects of therapeutic interventions. Because many biomarkers are initially examined in animal models, the extent to which markers translate from animals to humans is an important issue. The current review highlights many existing and pipeline biomarker approaches, focusing on the degree of correspondence between AD patients and animal models. The review also highlights the need for greater translational correspondence between human and animal biomarkers.
Collapse
|
45
|
Xia CF, Arteaga J, Chen G, Gangadharmath U, Gomez LF, Kasi D, Lam C, Liang Q, Liu C, Mocharla VP, Mu F, Sinha A, Su H, Szardenings AK, Walsh JC, Wang E, Yu C, Zhang W, Zhao T, Kolb HC. [18
F]T807, a novel tau positron emission tomography imaging agent for Alzheimer's disease. Alzheimers Dement 2013; 9:666-76. [DOI: 10.1016/j.jalz.2012.11.008] [Citation(s) in RCA: 448] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 10/26/2012] [Accepted: 11/21/2012] [Indexed: 11/15/2022]
Affiliation(s)
- Chun-Fang Xia
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Janna Arteaga
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Gang Chen
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Umesh Gangadharmath
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Luis F. Gomez
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Dhanalakshmi Kasi
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Chung Lam
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Qianwa Liang
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Changhui Liu
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Vani P. Mocharla
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Fanrong Mu
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Anjana Sinha
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Helen Su
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - A. Katrin Szardenings
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Joseph C. Walsh
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Eric Wang
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Chul Yu
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Wei Zhang
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Tieming Zhao
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| | - Hartmuth C. Kolb
- Molecular Imaging Biomarker Research; Siemens Medical Solution USA, Inc; Culver City CA USA
| |
Collapse
|
46
|
Mullane K, Williams M. Alzheimer's therapeutics: continued clinical failures question the validity of the amyloid hypothesis-but what lies beyond? Biochem Pharmacol 2012. [PMID: 23178653 DOI: 10.1016/j.bcp.2012.11.014] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The worldwide incidence of Alzheimer's disease (AD) is increasing with estimates that 115 million individuals will have AD by 2050, creating an unsustainable healthcare challenge due to a lack of effective treatment options highlighted by multiple clinical failures of agents designed to reduce the brain amyloid burden considered synonymous with the disease. The amyloid hypothesis that has been the overarching focus of AD research efforts for more than two decades has been questioned in terms of its causality but has not been unequivocally disproven despite multiple clinical failures, This is due to issues related to the quality of compounds advanced to late stage clinical trials and the lack of validated biomarkers that allow the recruitment of AD patients into trials before they are at a sufficiently advanced stage in the disease where therapeutic intervention is deemed futile. Pursuit of a linear, reductionistic amyloidocentric approach to AD research, which some have compared to a religious faith, has resulted in other, equally plausible but as yet unvalidated AD hypotheses being underfunded leading to a disastrous roadblock in the search for urgently needed AD therapeutics. Genetic evidence supporting amyloid causality in AD is reviewed in the context of the clinical failures, and progress in tau-based and alternative approaches to AD, where an evolving modus operandi in biomedical research fosters excessive optimism and a preoccupation with unproven, and often ephemeral, biomarker/genome-based approaches that override transparency, objectivity and data-driven decision making, resulting in low probability environments where data are subordinate to self propagating hypotheses.
Collapse
|
47
|
|
48
|
Bibl M, Esselmann H, Wiltfang J. Neurochemical biomarkers in Alzheimer's disease and related disorders. Ther Adv Neurol Disord 2012; 5:335-48. [PMID: 23139704 PMCID: PMC3487531 DOI: 10.1177/1756285612455367] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Neurochemical biomarkers for diagnosing dementias are currently under intensive investigation and the field is rapidly expanding. The main protagonists and the best defined among them are cerebrospinal fluid levels of Aβ42, tau and its phosphorylated forms (p-tau). In addition, novel cerebrospinal fluid biomarkers are emerging and their multiparametric assessment seems most promising for increasing the accuracy in neurochemical dementia diagnostics. The combined assessment of Aβ42 and p-tau has recently shown value for diagnosing prodromal states of Alzheimer's dementia, that is, mild cognitive impairment. Disease-specific biomarkers for other degenerative dementias are still missing, but some progress has recently been made. As lumbar puncture is an additional burden for the patient, blood-based neurochemical biomarkers are definitely warranted and promising new discoveries have been made in this direction. These diagnostic developments have implicit therapeutic consequences and give rise to new requirements for future neurochemical dementia diagnostics.
Collapse
Affiliation(s)
- Mirko Bibl
- Department of Psychiatry, Psychotherapy and Addiction Medicine, Kliniken Essen-Mitte; University of Duisburg-Essen, Essen, Germany
| | | | | |
Collapse
|
49
|
Bekris LM, Millard S, Lutz F, Li G, Galasko DR, Farlow MR, Quinn JF, Kaye JA, Leverenz JB, Tsuang DW, Yu CE, Peskind ER. Tau phosphorylation pathway genes and cerebrospinal fluid tau levels in Alzheimer's disease. Am J Med Genet B Neuropsychiatr Genet 2012; 159B:874-83. [PMID: 22927204 PMCID: PMC3626266 DOI: 10.1002/ajmg.b.32094] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 08/03/2012] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is characterized by the presence in the brain of amyloid plaques, consisting predominately of the amyloid β peptide (Aβ), and neurofibrillary tangles, consisting primarily of tau. Hyper-phosphorylated-tau (p-tau) contributes to neuronal damage, and both p-tau and total-tau (t-tau) levels are elevated in AD cerebrospinal fluid (CSF) compared to cognitively normal controls. Our hypothesis was that increased ratios of CSF phosphorylated-tau levels relative to total-tau levels correlate with regulatory region genetic variation of kinase or phosphatase genes biologically associated with the phosphorylation status of tau. Eighteen SNPs located within 5' and 3' regions of 5 kinase and 4 phosphatase genes, as well as two SNPs within regulatory regions of the MAPT gene were chosen for this analysis. The study sample consisted of 101 AD patients and 169 cognitively normal controls. Rs7768046 in the FYN kinase gene and rs913275 in the PPP2R4 phosphatase gene were both associated with CSF p-tau and t-tau levels in AD. These SNPs were also differentially associated with either CSF t-tau (rs7768046) or CSF p-tau (rs913275) relative to t-tau levels in AD compared to controls. These results suggest that rs7768046 and rs913275 both influence CSF tau levels in an AD-associated manner.
Collapse
Affiliation(s)
- Lynn M. Bekris
- Geriatric Research, Education, and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, Washington,Department of Medicine, University of Washington, Seattle, Washington,Correspondence to: Lynn M. Bekris, Ph.D., Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, VAPSHCS GRECC S182B, 1660 South Columbian Way, Seattle, WA 98108.
| | - Steve Millard
- Northwest Network VISN-20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, Washington
| | - Franziska Lutz
- Geriatric Research, Education, and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, Washington,Department of Medicine, University of Washington, Seattle, Washington
| | - Gail Li
- Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Doug R. Galasko
- Department of Neurosciences, University of California at San Diego and VA Medical Center San Diego, San Diego, California
| | - Martin R. Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health and Science University, Portland, Oregon,Portland VA Medical Center, Portland, Oregon
| | - Jeffrey A. Kaye
- Department of Neurology, Oregon Health and Science University, Portland, Oregon,Portland VA Medical Center, Portland, Oregon
| | - James B. Leverenz
- Northwest Network VISN-20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, Washington,Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington,Department of Neurology, University of Washington School of Medicine, Seattle, Washington,Northwest Network VISN-20 Parkinson’s Disease Research, Education and Clinical Center (PADRECC), Roseburg, Oregon
| | - Debby W. Tsuang
- Northwest Network VISN-20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, Washington,Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Chang-En Yu
- Geriatric Research, Education, and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, Washington,Department of Medicine, University of Washington, Seattle, Washington
| | - Elaine R. Peskind
- Northwest Network VISN-20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, Washington,Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
50
|
Waedt J, Kleinow M, Kornhuber J, Lewczuk P. Neurochemical dementia diagnostics for Alzheimer’s disease and other dementias: an ISO 15189 perspective. Biomark Med 2012; 6:685-90. [DOI: 10.2217/bmm.12.63] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Dementia is one of the most common causes of health problems in the elderly populations of Western industrialized countries. A combined analysis of cerebrospinal fluid-based neurochemical dementia diagnostics biomarkers (amyloid-β peptides, total tau and phosphorylated forms of tau) provides sensitivity and specificity in the range of 85% for the diagnosis of Alzheimer’s disease, the most common cause of dementia. The alterations occur very early in the course of neurodegeneration, enabling medical follow-up of persons with increased risk of developing dementia. With a growing number of laboratories performing neurochemical dementia diagnostics routinely, it is important to standardize protocols and laboratory performance to enable comparisons of results and their interpretations. Together with the recently published expert guidelines for sample handling and preparation, as well as the interpretation (post-analytical) algorithms developed by experienced centers, ISO 15189 norm provides an extremely useful tool for standardization of neurochemical dementia diagnostics.
Collapse
Affiliation(s)
- Johanna Waedt
- Department of Psychiatry & Psychotherapy, Universitätsklinikum Erlangen & Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martina Kleinow
- Department of Psychiatry & Psychotherapy, Universitätsklinikum Erlangen & Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry & Psychotherapy, Universitätsklinikum Erlangen & Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Piotr Lewczuk
- Department of Psychiatry & Psychotherapy, Universitätsklinikum Erlangen & Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|