1
|
Huang Y, Tang J, Chen G, Wu Q, Wang Y, Chen J, Chen S, Liu J, Huang X. HDL-Apolipoprotein in Alzheimer's Disease Revisited: From Periphery to CNS. Aging Med (Milton) 2025; 8:164-177. [PMID: 40353050 PMCID: PMC12064998 DOI: 10.1002/agm2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/14/2024] [Accepted: 01/23/2025] [Indexed: 05/14/2025] Open
Abstract
High-density lipoprotein (HDL), as a crucial component of lipid metabolism, have roles in regulating Alzheimer's disease (AD) core pathology amyloid β (Aβ) and phosphorylated tau (p-tau) through its apolipoproteins, which are associated with brain structures, cognition, and risk of dementia. The pool of HDL apolipoproteins-in the brain and in the periphery-has its own distinct origin, composition, and regulatory mechanisms. It remains unclear whether these apolipoproteins in the periphery and CNS play distinct roles in the pathogenesis of AD. Specifically, this review focus on the distinct associations of apolipoprotein AI and apolipoprotein E-the major components of HDL in the blood and CSF-with pathological proteins, brain integrity, cognition, and dementia progression in AD. We summarize and examine the current state of knowledge on the values of these apolipoproteins in AD pathogenesis and clinical potential.
Collapse
Affiliation(s)
- Yihong Huang
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Jingyi Tang
- Department of NeurologySun Yat‐Sen Memorial Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Guohua Chen
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Qiangqiang Wu
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Yongfei Wang
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Jianjun Chen
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Simei Chen
- Department of NeurologyHoujie Hospital and Clinical College of Guangdong Medical UniversityDongguanGuangdongChina
| | - Jun Liu
- Department of NeurologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xiaoyun Huang
- Department of NeurologySun Yat‐Sen Memorial Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
2
|
Repetto L, Chen J, Yang Z, Zhai R, Timmers PRHJ, Feng X, Li T, Yao Y, Maslov D, Timoshchuk A, Tu F, Twait EL, May-Wilson S, Muckian MD, Prins BP, Png G, Kooperberg C, Johansson Å, Hillary RF, Wheeler E, Pan L, He Y, Klasson S, Ahmad S, Peters JE, Gilly A, Karaleftheri M, Tsafantakis E, Haessler J, Gyllensten U, Harris SE, Wareham NJ, Göteson A, Lagging C, Ikram MA, van Duijn CM, Jern C, Landén M, Langenberg C, Deary IJ, Marioni RE, Enroth S, Reiner AP, Dedoussis G, Zeggini E, Sharapov S, Aulchenko YS, Butterworth AS, Mälarstig A, Wilson JF, Navarro P, Shen X. The genetic landscape of neuro-related proteins in human plasma. Nat Hum Behav 2024; 8:2222-2234. [PMID: 39210026 DOI: 10.1038/s41562-024-01963-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/22/2024] [Indexed: 09/04/2024]
Abstract
Understanding the genetic basis of neuro-related proteins is essential for dissecting the molecular basis of human behavioural traits and the disease aetiology of neuropsychiatric disorders. Here the SCALLOP Consortium conducted a genome-wide association meta-analysis of over 12,000 individuals for 184 neuro-related proteins in human plasma. The analysis identified 125 cis-regulatory protein quantitative trait loci (cis-pQTL) and 164 trans-pQTL. The mapped pQTL capture on average 50% of each protein's heritability. At the cis-pQTL, multiple proteins shared a genetic basis with human behavioural traits such as alcohol and food intake, smoking and educational attainment, as well as neurological conditions and psychiatric disorders such as pain, neuroticism and schizophrenia. Integrating with established drug information, the causal inference analysis validated 52 out of 66 matched combinations of protein targets and diseases or side effects with available drugs while suggesting hundreds of repurposing and new therapeutic targets.
Collapse
Affiliation(s)
- Linda Repetto
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- Health Data Science Centre, Fondazione Human Technopole, Milan, Italy
| | - Jiantao Chen
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhijian Yang
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, China
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ranran Zhai
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, China
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Paul R H J Timmers
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Xiao Feng
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, China
| | - Ting Li
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yue Yao
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, China
| | - Denis Maslov
- MSU Institute for Artificial Intelligence, Lomonosov Moscow State University, Moscow, Russia
| | - Anna Timoshchuk
- MSU Institute for Artificial Intelligence, Lomonosov Moscow State University, Moscow, Russia
| | - Fengyu Tu
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Emma L Twait
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Sebastian May-Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Marisa D Muckian
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Bram P Prins
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Grace Png
- Institute of Translational Genomics, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM), TUM School of Medicine and Health, Munich, Germany
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Åsa Johansson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Eleanor Wheeler
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Lu Pan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yazhou He
- Department of Epidemiology and Medical Statistics, Division of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Sofia Klasson
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Shahzad Ahmad
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - James E Peters
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, UK
| | - Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | | | | | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ulf Gyllensten
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Sarah E Harris
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Andreas Göteson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lagging
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | | | - Christina Jern
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mikael Landén
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Computational Medicine, Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Ian J Deary
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Stefan Enroth
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Alexander P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Center and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens, Greece
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine and Health, Munich, Germany
| | - Sodbo Sharapov
- MSU Institute for Artificial Intelligence, Lomonosov Moscow State University, Moscow, Russia
- Biostatistics Unit-Population and Medical Genomics Programme, Genomics Research Centre, Fondazione Human Technopole, Milan, Italy
| | - Yurii S Aulchenko
- MSU Institute for Artificial Intelligence, Lomonosov Moscow State University, Moscow, Russia
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Adam S Butterworth
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Anders Mälarstig
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Emerging Science and Innovation, Pfizer Worldwide Research, Development and Medical, Cambridge, UK
| | - James F Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Pau Navarro
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Xia Shen
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China.
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK.
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, China.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
3
|
Koychev I, Reid G, Nguyen M, Mentz RJ, Joyce D, Shah SH, Holman RR. Inflammatory proteins associated with Alzheimer's disease reduced by a GLP1 receptor agonist: a post hoc analysis of the EXSCEL randomized placebo controlled trial. Alzheimers Res Ther 2024; 16:212. [PMID: 39358806 PMCID: PMC11448378 DOI: 10.1186/s13195-024-01573-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists are a viable option for the prevention of Alzheimer's disease (AD) but the mechanisms of this potential disease modifying action are unclear. We investigated the effects of once-weekly exenatide (EQW) on AD associated proteomic clusters. METHODS The Exenatide Study of Cardiovascular Event Lowering study compared the cardiovascular effects of EQW 2 mg with placebo in 13,752 people with type 2 diabetes mellitus. 4,979 proteins were measured (Somascan V0.4) on baseline and 1-year plasma samples of 3,973 participants. C-reactive protein (CRP), ficolin-2 (FCN2), plasminogen activator inhibitor 1 (PAI-1), soluble vascular cell adhesion protein 1 (sVCAM1) and 4 protein clusters were tested in multivariable mixed models. RESULTS EQW affected FCN2 (Cohen's d -0.019), PAI-1 (Cohen's d -0.033), sVCAM-1 (Cohen's d 0.035) and a cytokine-cytokine cluster (Cohen's d 0.037) significantly compared with placebo. These effects were sustained in individuals over the age of 65 but not in those under 65. CONCLUSIONS EQW treatment was associated with significant change in inflammatory proteins associated with AD. TRIAL REGISTRATION EXSCEL is registered on ClinicalTrials.gov: NCT01144338 on 10th of June 2010.
Collapse
Affiliation(s)
- Ivan Koychev
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxford, UK.
| | - Graham Reid
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Maggie Nguyen
- Duke Center for Precision Health, Duke University School of Medicine, Durham, NC, USA
| | | | - Dan Joyce
- Department of Psychiatry, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Svati H Shah
- Duke Center for Precision Health, Duke University School of Medicine, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Rury R Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
4
|
Vilkaite G, Vogel J, Mattsson-Carlgren N. Integrating amyloid and tau imaging with proteomics and genomics in Alzheimer's disease. Cell Rep Med 2024; 5:101735. [PMID: 39293391 PMCID: PMC11525023 DOI: 10.1016/j.xcrm.2024.101735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/28/2024] [Accepted: 08/20/2024] [Indexed: 09/20/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and is characterized by the aggregation of β-amyloid (Aβ) and tau in the brain. Breakthroughs in disease-modifying treatments targeting Aβ bring new hope for the management of AD. But to effectively modify and someday even prevent AD, a better understanding is needed of the biological mechanisms that underlie and link Aβ and tau in AD. Developments of high-throughput omics, including genomics, proteomics, and transcriptomics, together with molecular imaging of Aβ and tau with positron emission tomography (PET), allow us to discover and understand the biological pathways that regulate the aggregation and spread of Aβ and tau in living humans. The field of integrated omics and PET studies of Aβ and tau in AD is growing rapidly. We here provide an update of this field, both in terms of biological insights and in terms of future clinical implications of integrated omics-molecular imaging studies.
Collapse
Affiliation(s)
- Gabriele Vilkaite
- Department of Clinical Sciences Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Jacob Vogel
- Department of Clinical Sciences Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
5
|
Upadhyay A, Chhangani D, Rao NR, Kofler J, Vassar R, Rincon-Limas DE, Savas JN. Amyloid fibril proteomics of AD brains reveals modifiers of aggregation and toxicity. Mol Neurodegener 2023; 18:61. [PMID: 37710351 PMCID: PMC10503190 DOI: 10.1186/s13024-023-00654-z] [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: 01/25/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND The accumulation of amyloid beta (Aβ) peptides in fibrils is prerequisite for Alzheimer's disease (AD). Our understanding of the proteins that promote Aβ fibril formation and mediate neurotoxicity has been limited due to technical challenges in isolating pure amyloid fibrils from brain extracts. METHODS To investigate how amyloid fibrils form and cause neurotoxicity in AD brain, we developed a robust biochemical strategy. We benchmarked the success of our purifications using electron microscopy, amyloid dyes, and a large panel of Aβ immunoassays. Tandem mass-spectrometry based proteomic analysis workflows provided quantitative measures of the amyloid fibril proteome. These methods allowed us to compare amyloid fibril composition from human AD brains, three amyloid mouse models, transgenic Aβ42 flies, and Aβ42 seeded cultured neurons. RESULTS Amyloid fibrils are primarily composed by Aβ42 and unexpectedly harbor Aβ38 but generally lack Aβ40 peptides. Multidimensional quantitative proteomics allowed us to redefine the fibril proteome by identifying 20 new amyloid-associated proteins. Notably, we confirmed 57 previously reported plaque-associated proteins. We validated a panel of these proteins as bona fide amyloid-interacting proteins using antibodies and orthogonal proteomic analysis. One metal-binding chaperone metallothionein-3 is tightly associated with amyloid fibrils and modulates fibril formation in vitro. Lastly, we used a transgenic Aβ42 fly model to test if knock down or over-expression of fibril-interacting gene homologues modifies neurotoxicity. Here, we could functionally validate 20 genes as modifiers of Aβ42 toxicity in vivo. CONCLUSIONS These discoveries and subsequent confirmation indicate that fibril-associated proteins play a key role in amyloid formation and AD pathology.
Collapse
Affiliation(s)
- Arun Upadhyay
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Deepak Chhangani
- Department of Neurology, McKnight Brain Institute, and Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32611, USA
| | - Nalini R Rao
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Julia Kofler
- Department of Pathology, Division of Neuropathology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Robert Vassar
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Diego E Rincon-Limas
- Department of Neurology, McKnight Brain Institute, and Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32611, USA
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32611, USA
- Genetics Institute, University of Florida, Gainesville, FL, 32611, USA
| | - Jeffrey N Savas
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
6
|
Repetto L, Chen J, Yang Z, Zhai R, Timmers PRHJ, Li T, Twait EL, May-Wilson S, Muckian MD, Prins BP, Png G, Kooperberg C, Johansson Å, Hillary RF, Wheeler E, Pan L, He Y, Klasson S, Ahmad S, Peters JE, Gilly A, Karaleftheri M, Tsafantakis E, Haessler J, Gyllensten U, Harris SE, Wareham NJ, Göteson A, Lagging C, Ikram MA, van Duijn CM, Jern C, Landén M, Langenberg C, Deary IJ, Marioni RE, Enroth S, Reiner AP, Dedoussis G, Zeggini E, Butterworth AS, Mälarstig A, Wilson JF, Navarro P, Shen X. Unraveling Neuro-Proteogenomic Landscape and Therapeutic Implications for Human Behaviors and Psychiatric Disorders. RESEARCH SQUARE 2023:rs.3.rs-2720355. [PMID: 37034613 PMCID: PMC10081382 DOI: 10.21203/rs.3.rs-2720355/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Understanding the genetic basis of neuro-related proteins is essential for dissecting the molecular basis of human behavioral traits and the disease etiology of neuropsychiatric disorders. Here, the SCALLOP Consortium conducted a genome-wide association meta-analysis of over 12,500 individuals for 184 neuro-related proteins in human plasma. The analysis identified 117 cis-regulatory protein quantitative trait loci (cis-pQTL) and 166 trans-pQTL. The mapped pQTL capture on average 50% of each protein's heritability. Mendelian randomization analyses revealed multiple proteins showing potential causal effects on neuro-related traits such as sleeping, smoking, feelings, alcohol intake, mental health, and psychiatric disorders. Integrating with established drug information, we validated 13 out of 13 matched combinations of protein targets and diseases or side effects with available drugs, while suggesting hundreds of re-purposing and new therapeutic targets. This consortium effort provides a large-scale proteogenomic resource for biomedical research on human behaviors and other neuro-related phenotypes.
Collapse
Affiliation(s)
- Linda Repetto
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
| | - Jiantao Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhijian Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ranran Zhai
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Paul R. H. J. Timmers
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, MRC Institute of Genetics Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Ting Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Emma L. Twait
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrechtand Utrecht University, Utrecht, Netherlands
| | - Sebastian May-Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Marisa D. Muckian
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Bram P. Prins
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Grace Png
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM), School of Medicine, 81675 Munich, Germany
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle USA
| | - Åsa Johansson
- Dept. Immunology, Genetics and Pathology, Science for life laboratory, Uppsala University, Sweden
| | - Robert F. Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, EH4 2XU, United Kingdom
| | - Eleanor Wheeler
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Lu Pan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yazhou He
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Sofia Klasson
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Shahzad Ahmad
- Department of Epidemiology, ErasmusMC, Rotterdam, The Netherlands
| | - James E. Peters
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | | | | | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle USA
| | - Ulf Gyllensten
- Dept. Immunology, Genetics and Pathology, Science for life laboratory, Uppsala University, Sweden
| | - Sarah E. Harris
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, EH8 9JZ, United Kingdom
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Andreas Göteson
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lagging
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Genetics and Genomics, Gothenburg, Sweden
| | | | | | - Christina Jern
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mikael Landén
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Computational Medicine, Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, Germany
| | - Ian J. Deary
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, EH8 9JZ, United Kingdom
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, EH4 2XU, United Kingdom
| | - Stefan Enroth
- Dept. Immunology, Genetics and Pathology, Science for life laboratory, Uppsala University, Sweden
| | - Alexander P. Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Center and Department of Epidemiology, University of Washington, Seattle USA
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens, Greece
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine, Munich, Germany
| | - Adam S. Butterworth
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Anders Mälarstig
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Emerging Science and Innovation, Pfizer Worldwide Research, Development and Medical, Cambridge, UK
| | - James F. Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, MRC Institute of Genetics Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Pau Navarro
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, MRC Institute of Genetics Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Xia Shen
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Repetto L, Chen J, Yang Z, Zhai R, Timmers PRHJ, Li T, Twait EL, May-Wilson S, Muckian MD, Prins BP, Png G, Kooperberg C, Johansson Å, Hillary RF, Wheeler E, Pan L, He Y, Klasson S, Ahmad S, Peters JE, Gilly A, Karaleftheri M, Tsafantakis E, Haessler J, Gyllensten U, Harris SE, Wareham NJ, Göteson A, Lagging C, Ikram MA, van Duijn CM, Jern C, Landén M, Langenberg C, Deary IJ, Marioni RE, Enroth S, Reiner AP, Dedoussis G, Zeggini E, Butterworth AS, Mälarstig A, Wilson JF, Navarro P, Shen X. Genetic mechanisms of 184 neuro-related proteins in human plasma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.10.23285650. [PMID: 36824751 PMCID: PMC9949195 DOI: 10.1101/2023.02.10.23285650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Understanding the genetic basis of neuro-related proteins is essential for dissecting the disease etiology of neuropsychiatric disorders and other complex traits and diseases. Here, the SCALLOP Consortium conducted a genome-wide association meta-analysis of over 12,500 individuals for 184 neuro-reiated proteins in human plasma. The analysis identified 117 cis-regulatory protein quantitative trait loci (cis-pQTL) and 166 trans-pQTL. The mapped pQTL capture on average 50% of each protein's heritability. Mendelian randomization analyses revealed multiple proteins showing potential causal effects on neuro-reiated traits as well as complex diseases such as hypertension, high cholesterol, immune-related disorders, and psychiatric disorders. Integrating with established drug information, we validated 13 combinations of protein targets and diseases or side effects with available drugs, while suggesting hundreds of re-purposing and new therapeutic targets for diseases and comorbidities. This consortium effort provides a large-scale proteogenomic resource for biomedical research.
Collapse
Affiliation(s)
- Linda Repetto
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
| | - Jiantao Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhijian Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ranran Zhai
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Paul R. H. J. Timmers
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, MRC Institute of Genetics Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Ting Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Emma L. Twait
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Sebastian May-Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Marisa D. Muckian
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Bram P. Prins
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Grace Png
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM), School of Medicine, 81675 Munich, Germany
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle USA
| | - Åsa Johansson
- Dept. Immunology, Genetics and Pathology, Science for life laboratory, Uppsala University, Sweden
| | - Robert F. Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, EH4 2XU, United Kingdom
| | - Eleanor Wheeler
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Lu Pan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yazhou He
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Sofia Klasson
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Shahzad Ahmad
- Department of Epidemiology, ErasmusMC, Rotterdam, The Netherlands
| | - James E. Peters
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | | | | | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle USA
| | - Ulf Gyllensten
- Dept. Immunology, Genetics and Pathology, Science for life laboratory, Uppsala University, Sweden
| | - Sarah E. Harris
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, EH8 9JZ, United Kingdom
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Andreas Göteson
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lagging
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Genetics and Genomics, Gothenburg, Sweden
| | | | | | - Christina Jern
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mikael Landén
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Computational Medicine, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Germany
| | - Ian J. Deary
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, EH8 9JZ, United Kingdom
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, EH4 2XU, United Kingdom
| | - Stefan Enroth
- Dept. Immunology, Genetics and Pathology, Science for life laboratory, Uppsala University, Sweden
| | - Alexander P. Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Center and Department of Epidemiology, University of Washington, Seattle USA
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens, Greece
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine, Munich, Germany
| | - Adam S. Butterworth
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Anders Mälarstig
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Emerging Science and Innovation, Pfizer Worldwide Research, Development and Medical, Cambridge, UK
| | - James F. Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, MRC Institute of Genetics Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Pau Navarro
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, MRC Institute of Genetics Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Xia Shen
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Division of Public Health Sciences, Fred Hutchinson Cancer Center and Department of Epidemiology, University of Washington, Seattle USA
| |
Collapse
|
8
|
Brosseron F, Maass A, Kleineidam L, Ravichandran KA, Kolbe CC, Wolfsgruber S, Santarelli F, Häsler LM, McManus R, Ising C, Röske S, Peters O, Cosma NC, Schneider LS, Wang X, Priller J, Spruth EJ, Altenstein S, Schneider A, Fliessbach K, Wiltfang J, Schott BH, Buerger K, Janowitz D, Dichgans M, Perneczky R, Rauchmann BS, Teipel S, Kilimann I, Görß D, Laske C, Munk MH, Düzel E, Yakupow R, Dobisch L, Metzger CD, Glanz W, Ewers M, Dechent P, Haynes JD, Scheffler K, Roy N, Rostamzadeh A, Spottke A, Ramirez A, Mengel D, Synofzik M, Jucker M, Latz E, Jessen F, Wagner M, Heneka MT. Serum IL-6, sAXL, and YKL-40 as systemic correlates of reduced brain structure and function in Alzheimer's disease: results from the DELCODE study. Alzheimers Res Ther 2023; 15:13. [PMID: 36631909 PMCID: PMC9835320 DOI: 10.1186/s13195-022-01118-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/06/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Neuroinflammation constitutes a pathological hallmark of Alzheimer's disease (AD). Still, it remains unresolved if peripheral inflammatory markers can be utilized for research purposes similar to blood-based beta-amyloid and neurodegeneration measures. We investigated experimental inflammation markers in serum and analyzed interrelations towards AD pathology features in a cohort with a focus on at-risk stages of AD. METHODS Data of 74 healthy controls (HC), 99 subjective cognitive decline (SCD), 75 mild cognitive impairment (MCI), 23 AD relatives, and 38 AD subjects were obtained from the DELCODE cohort. A panel of 20 serum biomarkers was determined using immunoassays. Analyses were adjusted for age, sex, APOE status, and body mass index and included correlations between serum and CSF marker levels and AD biomarker levels. Group-wise comparisons were based on screening diagnosis and routine AD biomarker-based schematics. Structural imaging data were combined into composite scores representing Braak stage regions and related to serum biomarker levels. The Preclinical Alzheimer's Cognitive Composite (PACC5) score was used to test for associations between the biomarkers and cognitive performance. RESULTS Each experimental marker displayed an individual profile of interrelations to AD biomarkers, imaging, or cognition features. Serum-soluble AXL (sAXL), IL-6, and YKL-40 showed the most striking associations. Soluble AXL was significantly elevated in AD subjects with pathological CSF beta-amyloid/tau profile and negatively related to structural imaging and cognitive function. Serum IL-6 was negatively correlated to structural measures of Braak regions, without associations to corresponding IL-6 CSF levels or other AD features. Serum YKL-40 correlated most consistently to CSF AD biomarker profiles and showed the strongest negative relations to structure, but none to cognitive outcomes. CONCLUSIONS Serum sAXL, IL-6, and YKL-40 relate to different AD features, including the degree of neuropathology and cognitive functioning. This may suggest that peripheral blood signatures correspond to specific stages of the disease. As serum markers did not reflect the corresponding CSF protein levels, our data highlight the need to interpret serum inflammatory markers depending on the respective protein's specific biology and cellular origin. These marker-specific differences will have to be considered to further define and interpret blood-based inflammatory profiles for AD research.
Collapse
Affiliation(s)
- Frederic Brosseron
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Anne Maass
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Luca Kleineidam
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Kishore Aravind Ravichandran
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Carl-Christian Kolbe
- grid.15090.3d0000 0000 8786 803XInstitute of Innate Immunity, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany ,grid.420044.60000 0004 0374 4101Bayer AG, Alfred-Nobel-Straße 50, 40789 Monheim am Rhein, Germany
| | - Steffen Wolfsgruber
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Francesco Santarelli
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Lisa M. Häsler
- grid.10392.390000 0001 2190 1447Hertie Institute for Clinical Brain Research, Department Cellular Neurology, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany
| | - Róisín McManus
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Christina Ising
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany ,grid.452408.fExcellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Köln, Germany
| | - Sandra Röske
- grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Oliver Peters
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, 10117 Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Nicoleta-Carmen Cosma
- grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Luisa-Sophie Schneider
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, 10117 Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Xiao Wang
- grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Josef Priller
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, 10117 Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany ,grid.6936.a0000000123222966Department of Psychiatry and Psychotherapy, Technical University Munich, 81675 Munich, Germany
| | - Eike J. Spruth
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, 10117 Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Slawek Altenstein
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, 10117 Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Anja Schneider
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Klaus Fliessbach
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jens Wiltfang
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075 Göttingen, Germany ,grid.7450.60000 0001 2364 4210Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany ,grid.7311.40000000123236065Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Björn H. Schott
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075 Göttingen, Germany ,grid.7450.60000 0001 2364 4210Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany ,grid.418723.b0000 0001 2109 6265Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Katharina Buerger
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany ,grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Daniel Janowitz
- grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Martin Dichgans
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany ,grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Robert Perneczky
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany ,grid.411095.80000 0004 0477 2585Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany ,grid.7445.20000 0001 2113 8111Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK ,grid.11835.3e0000 0004 1936 9262Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Boris-Stephan Rauchmann
- grid.411095.80000 0004 0477 2585Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Stefan Teipel
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Gehlsheimer Str. 20, 18147 Rostock, Germany ,grid.413108.f0000 0000 9737 0454Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Ingo Kilimann
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Gehlsheimer Str. 20, 18147 Rostock, Germany ,grid.413108.f0000 0000 9737 0454Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Doreen Görß
- grid.413108.f0000 0000 9737 0454Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Christoph Laske
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany ,grid.10392.390000 0001 2190 1447Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Matthias H. Munk
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany ,grid.10392.390000 0001 2190 1447Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Emrah Düzel
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany ,grid.5807.a0000 0001 1018 4307Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Renat Yakupow
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Laura Dobisch
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Coraline D. Metzger
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany ,grid.5807.a0000 0001 1018 4307Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany ,grid.5807.a0000 0001 1018 4307Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany
| | - Wenzel Glanz
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Michael Ewers
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Peter Dechent
- grid.7450.60000 0001 2364 4210MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University, Goettingen, Germany
| | - John Dylan Haynes
- grid.6363.00000 0001 2218 4662Bernstein Center for Computational Neurosciences, Charité – Universitätsmedizin, Berlin, Germany
| | - Klaus Scheffler
- grid.10392.390000 0001 2190 1447Department for Biomedical Magnetic Resonance, University of Tübingen, 72076 Tübingen, Germany
| | - Nina Roy
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Ayda Rostamzadeh
- grid.6190.e0000 0000 8580 3777Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Annika Spottke
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.10388.320000 0001 2240 3300Department of Neurology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Alfredo Ramirez
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany ,grid.452408.fExcellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Köln, Germany ,grid.6190.e0000 0000 8580 3777Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany ,Department of Psychiatry & Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX USA
| | - David Mengel
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany ,grid.10392.390000 0001 2190 1447Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Matthis Synofzik
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany ,grid.10392.390000 0001 2190 1447Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Mathias Jucker
- grid.10392.390000 0001 2190 1447Hertie Institute for Clinical Brain Research, Department Cellular Neurology, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, 72076 Tübingen, Germany
| | - Eicke Latz
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XInstitute of Innate Immunity, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Frank Jessen
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.452408.fExcellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Köln, Germany ,grid.6190.e0000 0000 8580 3777Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Michael Wagner
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Michael T. Heneka
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany ,grid.16008.3f0000 0001 2295 9843Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, 4362 Esch-sur- Alzette, Luxembourg
| | | |
Collapse
|
9
|
Aerqin Q, Wang ZT, Wu KM, He XY, Dong Q, Yu JT. Omics-based biomarkers discovery for Alzheimer's disease. Cell Mol Life Sci 2022; 79:585. [PMID: 36348101 PMCID: PMC11803048 DOI: 10.1007/s00018-022-04614-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorders presenting with the pathological hallmarks of amyloid plaques and tau tangles. Over the past few years, great efforts have been made to explore reliable biomarkers of AD. High-throughput omics are a technology driven by multiple levels of unbiased data to detect the complex etiology of AD, and it provides us with new opportunities to better understand the pathophysiology of AD and thereby identify potential biomarkers. Through revealing the interaction networks between different molecular levels, the ultimate goal of multi-omics is to improve the diagnosis and treatment of AD. In this review, based on the current AD pathology and the current status of AD diagnostic biomarkers, we summarize how genomics, transcriptomics, proteomics and metabolomics are all conducing to the discovery of reliable AD biomarkers that could be developed and used in clinical AD management.
Collapse
Affiliation(s)
- Qiaolifan Aerqin
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Kai-Min Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Xiao-Yu He
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
10
|
Mofrad RB, Del Campo M, Peeters CFW, Meeter LHH, Seelaar H, Koel-Simmelink M, Ramakers IHGB, Middelkoop HAM, De Deyn PP, Claassen JAHR, van Swieten JC, Bridel C, Hoozemans JJM, Scheltens P, van der Flier WM, Pijnenburg YAL, Teunissen CE. Plasma proteome profiling identifies changes associated to AD but not to FTD. Acta Neuropathol Commun 2022; 10:148. [PMID: 36273219 PMCID: PMC9587555 DOI: 10.1186/s40478-022-01458-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Frontotemporal dementia (FTD) is caused by frontotemporal lobar degeneration (FTLD), characterized mainly by inclusions of Tau (FTLD-Tau) or TAR DNA binding43 (FTLD-TDP) proteins. Plasma biomarkers are strongly needed for specific diagnosis and potential treatment monitoring of FTD. We aimed to identify specific FTD plasma biomarker profiles discriminating FTD from AD and controls, and between FTD pathological subtypes. In addition, we compared plasma results with results in post-mortem frontal cortex of FTD cases to understand the underlying process. METHODS Plasma proteins (n = 1303) from pathologically and/or genetically confirmed FTD patients (n = 56; FTLD-Tau n = 16; age = 58.2 ± 6.2; 44% female, FTLD-TDP n = 40; age = 59.8 ± 7.9; 45% female), AD patients (n = 57; age = 65.5 ± 8.0; 39% female), and non-demented controls (n = 148; 61.3 ± 7.9; 41% female) were measured using an aptamer-based proteomic technology (SomaScan). In addition, exploratory analysis in post-mortem frontal brain cortex of FTD (n = 10; FTLD-Tau n = 5; age = 56.2 ± 6.9, 60% female, and FTLD-TDP n = 5; age = 64.0 ± 7.7, 60% female) and non-demented controls (n = 4; age = 61.3 ± 8.1; 75% female) were also performed. Differentially regulated plasma and tissue proteins were identified by global testing adjusting for demographic variables and multiple testing. Logistic lasso regression was used to identify plasma protein panels discriminating FTD from non-demented controls and AD, or FTLD-Tau from FTLD-TDP. Performance of the discriminatory plasma protein panels was based on predictions obtained from bootstrapping with 1000 resampled analysis. RESULTS Overall plasma protein expression profiles differed between FTD, AD and controls (6 proteins; p = 0.005), but none of the plasma proteins was specifically associated to FTD. The overall tissue protein expression profile differed between FTD and controls (7-proteins; p = 0.003). There was no difference in overall plasma or tissue expression profile between FTD subtypes. Regression analysis revealed a panel of 12-plasma proteins discriminating FTD from AD with high accuracy (AUC: 0.99). No plasma protein panels discriminating FTD from controls or FTD pathological subtypes were identified. CONCLUSIONS We identified a promising plasma protein panel as a minimally-invasive tool to aid in the differential diagnosis of FTD from AD, which was primarily associated to AD pathophysiology. The lack of plasma profiles specifically associated to FTD or its pathological subtypes might be explained by FTD heterogeneity, calling for FTD studies using large and well-characterize cohorts.
Collapse
Affiliation(s)
- R Babapour Mofrad
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands.,Alzheimer Center and Department of Neurology Amsterdam, Department of Neurology, Neuroscience Campus Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M Del Campo
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands.,Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain.,Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - C F W Peeters
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Mathematical and Statistical Methods Group (Biometris), Wageningen University and Research Wageningen, Wageningen, The Netherlands
| | - L H H Meeter
- Alzheimer Center Erasmus MC and Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - H Seelaar
- Alzheimer Center Rotterdam and Department of Neurology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - M Koel-Simmelink
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - I H G B Ramakers
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - H A M Middelkoop
- Institute of Psychology, Health, Medical and Neuropsychology Unit, Leiden University, Leiden, the Netherlands.,Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - P P De Deyn
- Laboratory of Neurochemistry and Behavior, Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Alzheimer Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J A H R Claassen
- Department of Geriatric Medicine, Radboud University Medical Center, Radboudumc Alzheimer Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - J C van Swieten
- Alzheimer Center Erasmus MC and Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - C Bridel
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - J J M Hoozemans
- Department of Pathology, Amsterdam University Medical Centers Location VUmc, Amsterdam, The Netherlands
| | - P Scheltens
- Alzheimer Center and Department of Neurology Amsterdam, Department of Neurology, Neuroscience Campus Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - W M van der Flier
- Alzheimer Center and Department of Neurology Amsterdam, Department of Neurology, Neuroscience Campus Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Y A L Pijnenburg
- Alzheimer Center and Department of Neurology Amsterdam, Department of Neurology, Neuroscience Campus Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Understanding carotenoid biosynthetic pathway control points using metabolomic analysis and natural genetic variation. Methods Enzymol 2022; 671:127-151. [DOI: 10.1016/bs.mie.2022.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
12
|
Endres K. Apolipoprotein A1, the neglected relative of Apolipoprotein E and its potential role in Alzheimer's disease. Neural Regen Res 2021; 16:2141-2148. [PMID: 33818485 PMCID: PMC8354123 DOI: 10.4103/1673-5374.310669] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/22/2020] [Accepted: 02/02/2021] [Indexed: 01/23/2023] Open
Abstract
Lipoproteins are multi-molecule assemblies with the primary function of transportation and processing of lipophilic substances within aqueous bodily fluids (blood, cerebrospinal fluid). Nevertheless, they also exert other physiological functions such as immune regulation. In particular, neurons are both sensitive to uncontrolled responses of the immune system and highly dependent on a controlled and sufficient supply of lipids. For this reason, the role of certain lipoproteins and their protein-component (apolipoproteins, Apo's) in neurological diseases is perceivable. ApoE, for example, is well-accepted as one of the major risk factors for sporadic Alzheimer's disease with a protective allele variant (ε2) and a risk-causing allele variant (ε4). ApoA1, the major protein component of high-density lipoproteins, is responsible for transportation of excess cholesterol from peripheral tissues to the liver. The protein is synthesized in the liver and intestine but also can enter the brain via the choroid plexus and thereby might have an impact on brain lipid homeostasis. This review focuses on the role of ApoA1 in Alzheimer's disease and discusses whether its role within this neurodegenerative disorder is specific or represents a general neuroprotective mechanism.
Collapse
Affiliation(s)
- Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Untere Zahlbacher Str. 8, 55131 Mainz, Germany
| |
Collapse
|
13
|
Laudanski K. Persistence of Lipoproteins and Cholesterol Alterations after Sepsis: Implication for Atherosclerosis Progression. Int J Mol Sci 2021; 22:ijms221910517. [PMID: 34638860 PMCID: PMC8508791 DOI: 10.3390/ijms221910517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Sepsis is one of the most common critical care illnesses with increasing survivorship. The quality of life in sepsis survivors is adversely affected by several co-morbidities, including increased incidence of dementia, stroke, cardiac disease and at least temporary deterioration in cognitive dysfunction. One of the potential explanations for their progression is the persistence of lipid profile abnormalities induced during acute sepsis into recovery, resulting in acceleration of atherosclerosis. (2) Methods: This is a targeted review of the abnormalities in the long-term lipid profile abnormalities after sepsis; (3) Results: There is a well-established body of evidence demonstrating acute alteration in lipid profile (HDL-c ↓↓, LDL-C -c ↓↓). In contrast, a limited number of studies demonstrated depression of HDL-c levels with a concomitant increase in LDL-C -c in the wake of sepsis. VLDL-C -c and Lp(a) remained unaltered in few studies as well. Apolipoprotein A1 was altered in survivors suggesting abnormalities in lipoprotein metabolism concomitant to overall lipoprotein abnormalities. However, most of the studies were limited to a four-month follow-up and patient groups were relatively small. Only one study looked at the atherosclerosis progression in sepsis survivors using clinical correlates, demonstrating an acceleration of plaque formation in the aorta, and a large metanalysis suggested an increase in the risk of stroke or acute coronary event between 3% to 9% in sepsis survivors. (4) Conclusions: The limited evidence suggests an emergence and persistence of the proatherogenic lipid profile in sepsis survivors that potentially contributes, along with other factors, to the clinical sequel of atherosclerosis.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA 19104, USA; ; Tel.: +1-215-662-8200
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Leonard Davis Institute of Healthcare Economics, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Cianflone A, Coppola L, Mirabelli P, Salvatore M. Predictive Accuracy of Blood-Derived Biomarkers for Amyloid-β Brain Deposition Along with the Alzheimer's Disease Continuum: A Systematic Review. J Alzheimers Dis 2021; 84:393-407. [PMID: 34542072 DOI: 10.3233/jad-210496] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND An amyloid-β (Aβ) positron emission tomography (Aβ-PET) scan of the human brain could lead to an early diagnosis of Alzheimer's disease (AD) and estimate disease progression. However, Aβ-PET imaging is expensive, invasive, and rarely applicable to cognitively normal subjects at risk for dementia. The identification of blood biomarkers predictive of Aβ brain deposition could help the identification of subjects at risk for dementia and could be helpful for the prognosis of AD progression. OBJECTIVE This study aimed to analyze the prognostic accuracy of blood biomarkers in predicting Aβ-PET status along with progression toward AD. METHODS In accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we searched bibliographic databases from 2010 to 2020. The quality of the included studies was assessed by the Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2) tool. RESULTS A total of 8 studies were retrieved. The prognostic accuracy of Aβ-PET status was calculated by obtaining ROCs for the following biomarkers: free, total, and bound Aβ42 and Aβ40; Aβ42/40 ratio; neurofilaments (NFL); total tau (T-tau); and phosphorylated-tau181 (P-tau181). Higher and lower plasma baseline levels of P-tau181 and the Aβ42/40 ratio, respectively, showed consistently good prognostication of Aβ-PET brain accumulation. Only P-tau181 was shown to predict AD progression. CONCLUSION In conclusion, the Aβ42/40 ratio and plasma P-tau181 were shown to predict Aβ-PET status. Plasma P-tau181 could also be a preclinical biomarker for AD progression.
Collapse
|
15
|
Shi L, Buckley NJ, Bos I, Engelborghs S, Sleegers K, Frisoni GB, Wallin A, Lléo A, Popp J, Martinez-Lage P, Legido-Quigley C, Barkhof F, Zetterberg H, Visser PJ, Bertram L, Lovestone S, Nevado-Holgado AJ. Plasma Proteomic Biomarkers Relating to Alzheimer's Disease: A Meta-Analysis Based on Our Own Studies. Front Aging Neurosci 2021; 13:712545. [PMID: 34366831 PMCID: PMC8335587 DOI: 10.3389/fnagi.2021.712545] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/21/2021] [Indexed: 01/21/2023] Open
Abstract
Background and Objective: Plasma biomarkers for the diagnosis and stratification of Alzheimer's disease (AD) are intensively sought. However, no plasma markers are well established so far for AD diagnosis. Our group has identified and validated various blood-based proteomic biomarkers relating to AD pathology in multiple cohorts. The study aims to conduct a meta-analysis based on our own studies to systematically assess the diagnostic performance of our previously identified blood biomarkers. Methods: To do this, we included seven studies that our group has conducted during the last decade. These studies used either Luminex xMAP or ELISA to measure proteomic biomarkers. As proteins measured in these studies differed, we selected protein based on the criteria that it must be measured in at least four studies. We then examined biomarker performance using random-effect meta-analyses based on the mean difference between biomarker concentrations in AD and controls (CTL), AD and mild cognitive impairment (MCI), MCI, and CTL as well as MCI converted to dementia (MCIc) and non-converted (MCInc) individuals. Results: An overall of 2,879 subjects were retrieved for meta-analysis including 1,053 CTL, 895 MCI, 882 AD, and 49 frontotemporal dementia (FTD) patients. Six proteins were measured in at least four studies and were chosen for meta-analyses for AD diagnosis. Of them, three proteins had significant difference between AD and controls, among which alpha-2-macroglobulin (A2M) and ficolin-2 (FCN2) increased in AD while fibrinogen gamma chain (FGG) decreased in AD compared to CTL. Furthermore, FGG significantly increased in FTD compared to AD. None of the proteins passed the significance between AD and MCI, or MCI and CTL, or MCIc and MCInc, although complement component 4 (CC4) tended to increase in MCIc individuals compared to MCInc. Conclusions: The results suggest that A2M, FCN2, and FGG are promising biomarkers to discriminate AD patients from controls, which are worthy of further validation.
Collapse
Affiliation(s)
- Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Noel J Buckley
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Isabelle Bos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, Netherlands.,Alzheimer Center, VU University Medical Center, Amsterdam, Netherlands
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Department of Neurology, Universitair Ziekenhuis Brussel and Center for Neurociences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Kristel Sleegers
- Complex Genetics Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.,Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Anders Wallin
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Alberto Lléo
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Julius Popp
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland.,Geriatric Psychiatry, Department of Mental Health and Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | | | - Cristina Legido-Quigley
- Kings College London, London, United Kingdom.,The Systems Medicine Group, Steno Diabetes Center, Gentofte, Denmark
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands.,UCL Institutes of Neurology and Healthcare Engineering, London, United Kingdom
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute at UCL, London, United Kingdom.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, Netherlands.,Alzheimer Center, VU University Medical Center, Amsterdam, Netherlands
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany.,Department of Psychology, University of Oslo, Oslo, Norway
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Janssen R&D, High Wycombe, United Kingdom
| | | |
Collapse
|
16
|
Ashton NJ, Leuzy A, Karikari TK, Mattsson-Carlgren N, Dodich A, Boccardi M, Corre J, Drzezga A, Nordberg A, Ossenkoppele R, Zetterberg H, Blennow K, Frisoni GB, Garibotto V, Hansson O. The validation status of blood biomarkers of amyloid and phospho-tau assessed with the 5-phase development framework for AD biomarkers. Eur J Nucl Med Mol Imaging 2021; 48:2140-2156. [PMID: 33677733 PMCID: PMC8175325 DOI: 10.1007/s00259-021-05253-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE The development of blood biomarkers that reflect Alzheimer's disease (AD) pathophysiology (phosphorylated tau and amyloid-β) has offered potential as scalable tests for dementia differential diagnosis and early detection. In 2019, the Geneva AD Biomarker Roadmap Initiative included blood biomarkers in the systematic validation of AD biomarkers. METHODS A panel of experts convened in November 2019 at a two-day workshop in Geneva. The level of maturity (fully achieved, partly achieved, preliminary evidence, not achieved, unsuccessful) of blood biomarkers was assessed based on the Biomarker Roadmap methodology and discussed fully during the workshop which also evaluated cerebrospinal fluid (CSF) and positron emission tomography (PET) biomarkers. RESULTS Plasma p-tau has shown analytical validity (phase 2 primary aim 1) and first evidence of clinical validity (phase 3 primary aim 1), whereas the maturity level for Aβ remains to be partially achieved. Full and partial achievement has been assigned to p-tau and Aβ, respectively, in their associations to ante-mortem measures (phase 2 secondary aim 2). However, only preliminary evidence exists for the influence of covariates, assay comparison and cut-off criteria. CONCLUSIONS Despite the relative infancy of blood biomarkers, in comparison to CSF biomarkers, much has already been achieved for phases 1 through 3 - with p-tau having greater success in detecting AD and predicting disease progression. However, sufficient data about the effect of covariates on the biomarker measurement is lacking. No phase 4 (real-world performance) or phase 5 (assessment of impact/cost) aim has been tested, thus not achieved.
Collapse
Affiliation(s)
- N J Ashton
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden.
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - A Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - T K Karikari
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
| | - N Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - A Dodich
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- Center for Neurocognitive Rehabilitation (CeRiN), CIMeC, University of Trento, Trento, Italy
| | - M Boccardi
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Rostock, Germany
- LANVIE - Laboratory of Neuroimaging of Aging, University of Geneva, Geneva, Switzerland
| | - J Corre
- Centre National de la Recherche Scientifique, Montpellier, France
| | - A Drzezga
- Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - A Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Aging, Karolinska University Hospital Stockholm, Stockholm, Sweden
| | - R Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - H Zetterberg
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - K Blennow
- Institute of Neuroscience & Physiology, Department of Psychiatry & Neurochemistry, Sahlgrenska Academy, University of Gothenburg, House V3/SU, SE-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - G B Frisoni
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Rostock, Germany
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - V Garibotto
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- Diagnostic Department, University Hospitals of Geneva, Geneva, Switzerland
| | - O Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
- UK Dementia Research Institute at UCL, London, UK.
- Memory Clinic, Skåne University Hospital, SE-205 02, Malmö, Sweden.
| |
Collapse
|
17
|
A multicentre validation study of the diagnostic value of plasma neurofilament light. Nat Commun 2021; 12:3400. [PMID: 34099648 PMCID: PMC8185001 DOI: 10.1038/s41467-021-23620-z] [Citation(s) in RCA: 297] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
Increased cerebrospinal fluid neurofilament light (NfL) is a recognized biomarker for neurodegeneration that can also be assessed in blood. Here, we investigate plasma NfL as a marker of neurodegeneration in 13 neurodegenerative disorders, Down syndrome, depression and cognitively unimpaired controls from two multicenter cohorts: King’s College London (n = 805) and the Swedish BioFINDER study (n = 1,464). Plasma NfL was significantly increased in all cortical neurodegenerative disorders, amyotrophic lateral sclerosis and atypical parkinsonian disorders. We demonstrate that plasma NfL is clinically useful in identifying atypical parkinsonian disorders in patients with parkinsonism, dementia in individuals with Down syndrome, dementia among psychiatric disorders, and frontotemporal dementia in patients with cognitive impairment. Data-driven cut-offs highlighted the fundamental importance of age-related clinical cut-offs for disorders with a younger age of onset. Finally, plasma NfL performs best when applied to indicate no underlying neurodegeneration, with low false positives, in all age-related cut-offs. Cerebrospinal fluid neurofilament light (NfL) is a biomarker for neurodegeneration that can also be assessed in blood. Here the authors show in a validation study the potential for plasma NfL as a biomarker for several neurodegenerative diseases.
Collapse
|
18
|
Sidorina A, Catesini G, Levi Mortera S, Marzano V, Putignani L, Boenzi S, Taurisano R, Garibaldi M, Deodato F, Dionisi-Vici C. Combined proteomic and lipidomic studies in Pompe disease allow a better disease mechanism understanding. J Inherit Metab Dis 2021; 44:705-717. [PMID: 33325062 DOI: 10.1002/jimd.12344] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022]
Abstract
Pompe disease (PD) is caused by deficiency of the enzyme acid α-glucosidase resulting in glycogen accumulation in lysosomes. Clinical symptoms include skeletal myopathy, respiratory failure, and cardiac hypertrophy. We studied plasma proteomic and lipidomic profiles in 12 PD patients compared to age-matched controls. The proteomic profiles were analyzed by nLC-MS/MS SWATH method. Wide-targeted lipidomic analysis was performed by LC-IMS/MS, allowing to quantify >1100 lipid species, spanning 13 classes. Significant differences were found for 16 proteins, with four showing the most relevant changes (GPLD1, PON1, LDHB, PKM). Lipidomic analysis showed elevated levels of three phosphatidylcholines and of the free fatty acid 22:4, and reduced levels of six lysophosphatidylcholines. Up-regulated glycolytic enzymes (LDHB and PKM) are involved in autophagy and glycogen metabolism, while down-regulated PON1 and GPLD1 combined with lipidomic data indicate an abnormal phospholipid metabolism. Reduced GPLD1 and dysregulation of lipids with acyl-chains characteristic of GPI-anchor structure suggest the potential involvement of GPI-anchor system in PD. Results of proteomic analysis displayed the involvement of multiple cellular functions affecting inflammatory, immune and antioxidant responses, autophagy, Ca2+ -homeostasis, and cell adhesion. The combined multi-omic approach revealed new biosignatures in PD, providing novel insights in disease pathophysiology with potential future clinical application.
Collapse
Affiliation(s)
- Anna Sidorina
- Division of Metabolism, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Giulio Catesini
- Division of Metabolism, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | | | - Valeria Marzano
- Unit of Human Microbiome, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Lorenza Putignani
- Unit of Human Microbiome, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
- Department of Diagnostic and Laboratory Medicine, Unit of Parasitology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Sara Boenzi
- Division of Metabolism, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Roberta Taurisano
- Division of Metabolism, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Matteo Garibaldi
- Department of Neurosciences, Mental Health and Sensory Organs NESMOS, Faculty of Medicine and Psychology, SAPIENZA University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Federica Deodato
- Division of Metabolism, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Carlo Dionisi-Vici
- Division of Metabolism, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| |
Collapse
|
19
|
Ashford MT, Veitch DP, Neuhaus J, Nosheny RL, Tosun D, Weiner MW. The search for a convenient procedure to detect one of the earliest signs of Alzheimer's disease: A systematic review of the prediction of brain amyloid status. Alzheimers Dement 2021; 17:866-887. [PMID: 33583100 DOI: 10.1002/alz.12253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/10/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Convenient, cost-effective tests for amyloid beta (Aβ) are needed to identify those at higher risk for developing Alzheimer's disease (AD). This systematic review evaluates recent models that predict dichotomous Aβ. (PROSPERO: CRD42020144734). METHODS We searched Embase and identified 73 studies from 29,581 for review. We assessed study quality using established tools, extracted information, and reported results narratively. RESULTS We identified few high-quality studies due to concerns about Aβ determination and analytical issues. The most promising convenient, inexpensive classifiers consist of age, apolipoprotein E genotype, cognitive measures, and/or plasma Aβ. Plasma Aβ may be sufficient if pre-analytical variables are standardized and scalable assays developed. Some models lowered costs associated with clinical trial recruitment or clinical screening. DISCUSSION Conclusions about models are difficult due to study heterogeneity and quality. Promising prediction models used demographic, cognitive/neuropsychological, imaging, and plasma Aβ measures. Further studies using standardized Aβ determination, and improved model validation are required.
Collapse
Affiliation(s)
- Miriam T Ashford
- Department of Veterans Affairs Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA.,Department of Veterans Affairs Medical Center, Center for Imaging and Neurodegenerative Diseases, San Francisco, California, USA
| | - Dallas P Veitch
- Department of Veterans Affairs Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - John Neuhaus
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Rachel L Nosheny
- Department of Veterans Affairs Medical Center, Center for Imaging and Neurodegenerative Diseases, San Francisco, California, USA.,Department of Psychiatry, University of California San Francisco, San Francisco, California, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Michael W Weiner
- Department of Veterans Affairs Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA.,Department of Veterans Affairs Medical Center, Center for Imaging and Neurodegenerative Diseases, San Francisco, California, USA.,Department of Psychiatry, University of California San Francisco, San Francisco, California, USA.,Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA.,Department of Medicine, University of California San Francisco, San Francisco, California, USA.,Department of Neurology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
20
|
Tosun D, Veitch D, Aisen P, Jack CR, Jagust WJ, Petersen RC, Saykin AJ, Bollinger J, Ovod V, Mawuenyega KG, Bateman RJ, Shaw LM, Trojanowski JQ, Blennow K, Zetterberg H, Weiner MW. Detection of β-amyloid positivity in Alzheimer's Disease Neuroimaging Initiative participants with demographics, cognition, MRI and plasma biomarkers. Brain Commun 2021; 3:fcab008. [PMID: 33842885 PMCID: PMC8023542 DOI: 10.1093/braincomms/fcab008] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 01/18/2023] Open
Abstract
In vivo gold standard for the ante-mortem assessment of brain β-amyloid pathology is currently β-amyloid positron emission tomography or cerebrospinal fluid measures of β-amyloid42 or the β-amyloid42/β-amyloid40 ratio. The widespread acceptance of a biomarker classification scheme for the Alzheimer's disease continuum has ignited interest in more affordable and accessible approaches to detect Alzheimer's disease β-amyloid pathology, a process that often slows down the recruitment into, and adds to the cost of, clinical trials. Recently, there has been considerable excitement concerning the value of blood biomarkers. Leveraging multidisciplinary data from cognitively unimpaired participants and participants with mild cognitive impairment recruited by the multisite biomarker study of Alzheimer's Disease Neuroimaging Initiative, here we assessed to what extent plasma β-amyloid42/β-amyloid40, neurofilament light and phosphorylated-tau at threonine-181 biomarkers detect the presence of β-amyloid pathology, and to what extent the addition of clinical information such as demographic data, APOE genotype, cognitive assessments and MRI can assist plasma biomarkers in detecting β-amyloid-positivity. Our results confirm plasma β-amyloid42/β-amyloid40 as a robust biomarker of brain β-amyloid-positivity (area under curve, 0.80-0.87). Plasma phosphorylated-tau at threonine-181 detected β-amyloid-positivity only in the cognitively impaired with a moderate area under curve of 0.67, whereas plasma neurofilament light did not detect β-amyloid-positivity in either group of participants. Clinical information as well as MRI-score independently detected positron emission tomography β-amyloid-positivity in both cognitively unimpaired and impaired (area under curve, 0.69-0.81). Clinical information, particularly APOE ε4 status, enhanced the performance of plasma biomarkers in the detection of positron emission tomography β-amyloid-positivity by 0.06-0.14 units of area under curve for cognitively unimpaired, and by 0.21-0.25 units for cognitively impaired; and further enhancement of these models with an MRI-score of β-amyloid-positivity yielded an additional improvement of 0.04-0.11 units of area under curve for cognitively unimpaired and 0.05-0.09 units for cognitively impaired. Taken together, these multi-disciplinary results suggest that when combined with clinical information, plasma phosphorylated-tau at threonine-181 and neurofilament light biomarkers, and an MRI-score could effectively identify β-amyloid+ cognitively unimpaired and impaired (area under curve, 0.80-0.90). Yet, when the MRI-score is considered in combination with clinical information, plasma phosphorylated-tau at threonine-181 and plasma neurofilament light have minimal added value for detecting β-amyloid-positivity. Our systematic comparison of β-amyloid-positivity detection models identified effective combinations of demographics, APOE, global cognition, MRI and plasma biomarkers. Promising minimally invasive and low-cost predictors such as plasma biomarkers of β-amyloid42/β-amyloid40 may be improved by age and APOE genotype.
Collapse
Affiliation(s)
- Duygu Tosun
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Dallas Veitch
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Paul Aisen
- Alzheimer’s Therapeutic Research Institute (ATRI), Keck School of Medicine, University of Southern California, San Diego, CA, USA
| | | | - William J Jagust
- School of Public Health and Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Ronald C Petersen
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James Bollinger
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Vitaliy Ovod
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Kwasi G Mawuenyega
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Michael W Weiner
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
21
|
Differential Expression of mRNAs in Peripheral Blood Related to Prodrome and Progression of Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4505720. [PMID: 33204697 PMCID: PMC7648929 DOI: 10.1155/2020/4505720] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/25/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a chronic progressive neurodegenerative disease that affects the quality of life of elderly individuals, while the pathogenesis of AD is still unclear. Based on the bioinformatics analysis of differentially expressed genes (DEGs) in peripheral blood samples, we investigated genes related to mild cognitive impairment (MCI), AD, and late-stage AD that might be used for predicting the conversions. Methods. We obtained the DEGs in MCI, AD, and advanced AD patients from the Gene Expression Omnibus (GEO) database. A Venn diagram was used to identify the intersecting genes. Gene Ontology (GO) and Kyoto Gene and Genomic Encyclopedia (KEGG) were used to analyze the functions and pathways of the intersecting genes. Protein-protein interaction (PPI) networks were constructed to visualize the network of the proteins coded by the related genes. Hub genes were selected based on the PPI network. Results. Bioinformatics analysis indicated that there were 61 DEGs in both the MCI and AD groups and 27 the same DEGs among the three groups. Using GO and KEGG analyses, we found that these genes were related to the function of mitochondria and ribosome. Hub genes were determined by bioinformatics software based on the PPI network. Conclusions. Mitochondrial and ribosomal dysfunction in peripheral blood may be early signs in AD patients and related to the disease progression. The identified hub genes may provide the possibility for predicting AD progression or be the possible targets for treatments.
Collapse
|
22
|
Diniz Pereira J, Gomes Fraga V, Morais Santos AL, Carvalho MDG, Caramelli P, Braga Gomes K. Alzheimer's disease and type 2 diabetes mellitus: A systematic review of proteomic studies. J Neurochem 2020; 156:753-776. [PMID: 32909269 DOI: 10.1111/jnc.15166] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/15/2020] [Accepted: 08/25/2020] [Indexed: 12/16/2022]
Abstract
Similar to dementia, the risk for developing type 2 diabetes mellitus (T2DM) increases with age, and T2DM also increases the risk for dementia, particularly Alzheimer's disease (AD). Although T2DM is primarily a peripheral disorder and AD is a central nervous system disease, both share some common features as they are chronic and complex diseases, and both show involvement of oxidative stress and inflammation in their progression. These characteristics suggest that T2DM may be associated with AD, which gave rise to a new term, type 3 diabetes (T3DM). In this study, we searched for matching peripheral proteomic biomarkers of AD and T2DM based in a systematic review of the available literature. We identified 17 common biomarkers that were differentially expressed in both patients with AD or T2DM when compared with healthy controls. These biomarkers could provide a useful workflow for screening T2DM patients at risk to develop AD.
Collapse
Affiliation(s)
- Jessica Diniz Pereira
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vanessa Gomes Fraga
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anna Luiza Morais Santos
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria das Graças Carvalho
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Paulo Caramelli
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karina Braga Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
23
|
Pekkala T, Hall A, Ngandu T, van Gils M, Helisalmi S, Hänninen T, Kemppainen N, Liu Y, Lötjönen J, Paajanen T, Rinne JO, Soininen H, Kivipelto M, Solomon A. Detecting Amyloid Positivity in Elderly With Increased Risk of Cognitive Decline. Front Aging Neurosci 2020; 12:228. [PMID: 32848707 PMCID: PMC7406705 DOI: 10.3389/fnagi.2020.00228] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/29/2020] [Indexed: 12/28/2022] Open
Abstract
The importance of early interventions in Alzheimer's disease (AD) emphasizes the need to accurately and efficiently identify at-risk individuals. Although many dementia prediction models have been developed, there are fewer studies focusing on detection of brain pathology. We developed a model for identification of amyloid-PET positivity using data on demographics, vascular factors, cognition, APOE genotype, and structural MRI, including regional brain volumes, cortical thickness and a visual medial temporal lobe atrophy (MTA) rating. We also analyzed the relative importance of different factors when added to the overall model. The model used baseline data from the Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) exploratory PET sub-study. Participants were at risk for dementia, but without dementia or cognitive impairment. Their mean age was 71 years. Participants underwent a brain 3T MRI and PiB-PET imaging. PiB images were visually determined as positive or negative. Cognition was measured using a modified version of the Neuropsychological Test Battery. Body mass index (BMI) and hypertension were used as cardiovascular risk factors in the model. Demographic factors included age, gender and years of education. The model was built using the Disease State Index (DSI) machine learning algorithm. Of the 48 participants, 20 (42%) were rated as Aβ positive. Compared with the Aβ negative group, the Aβ positive group had a higher proportion of APOE ε4 carriers (53 vs. 14%), lower executive functioning, lower brain volumes, and higher visual MTA rating. AUC [95% CI] for the complete model was 0.78 [0.65-0.91]. MRI was the most effective factor, especially brain volumes and visual MTA rating but not cortical thickness. APOE was nearly as effective as MRI in improving detection of amyloid positivity. The model with the best performance (AUC 0.82 [0.71-0.93]) was achieved by combining APOE and MRI. Our findings suggest that combining demographic data, vascular risk factors, cognitive performance, APOE genotype, and brain MRI measures can help identify Aβ positivity. Detecting amyloid positivity could reduce invasive and costly assessments during the screening process in clinical trials.
Collapse
Affiliation(s)
- Timo Pekkala
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Anette Hall
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Tiia Ngandu
- Public Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland.,Division of Clinical Geriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden
| | - Mark van Gils
- VTT Technical Research Centre of Finland Ltd., Tampere, Finland
| | - Seppo Helisalmi
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Tuomo Hänninen
- Neurocenter/Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Nina Kemppainen
- Turku PET Centre, University of Turku, Turku, Finland.,Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| | - Yawu Liu
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland.,Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | | | - Teemu Paajanen
- Finnish Institute of Occupational Health, Helsinki, Finland
| | - Juha O Rinne
- Turku PET Centre, University of Turku, Turku, Finland.,Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| | - Hilkka Soininen
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland.,Neurocenter/Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Miia Kivipelto
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland.,Division of Clinical Geriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden.,Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.,Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Alina Solomon
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland.,Division of Clinical Geriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Kim JW, Byun MS, Lee JH, Yi D, Jeon SY, Sohn BK, Lee JY, Shin SA, Kim YK, Kang KM, Sohn CH, Lee DY. Serum albumin and beta-amyloid deposition in the human brain. Neurology 2020; 95:e815-e826. [PMID: 32690787 PMCID: PMC7605506 DOI: 10.1212/wnl.0000000000010005] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 01/27/2020] [Indexed: 01/21/2023] Open
Abstract
Objectives To investigate the relationships of serum albumin with in vivo Alzheimer disease (AD) pathologies, including cerebral β-amyloid (Aβ) protein deposition, neurodegeneration of AD-signature regions, and cerebral white matter hyperintensities (WMH), in the human brain. Methods A total of 396 older adults without dementia underwent comprehensive clinical assessments, measurement of serum albumin level, and multimodal brain imaging, including [11C] Pittsburgh compound B-PET, 18F-fluorodeoxyglucose-PET, and MRI. Serum albumin was categorized as follows: <4.4 g/dL (low albumin), 4.4 to 4.5 g/dL (middle albumin), and >4.5 g/dL (high albumin; used as a reference category). Aβ positivity, AD-signature region cerebral glucose metabolism (AD-CM), AD-signature region cortical thickness (AD-CT), and WMH volume were used as outcome measures. Results Serum albumin level (as a continuous variable) was inversely associated with Aβ deposition and Aβ positivity. The low albumin group showed a significantly higher Aβ positivity rate compared to the high albumin group (odds ratio 3.40, 95% confidence interval 1.67–6.92, p = 0.001), while the middle albumin group showed no difference (odds ratio 1.74, 95% confidence interval 0.80–3.77, p = 0.162). Neither serum albumin level (as a continuous variable) nor albumin categories were related to AD-CM, AD-CT, or WMH volume. Conclusions Low serum albumin may increase the risk of AD dementia by elevating amyloid accumulation. In terms of AD prevention, more attention needs to be paid to avoid a low serum albumin level, even within the clinical normal range, by clinicians.
Collapse
Affiliation(s)
- Jee Wook Kim
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea
| | - Min Soo Byun
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea
| | - Jun Ho Lee
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea.
| | - Dahyun Yi
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea
| | - So Yeon Jeon
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea
| | - Bo Kyung Sohn
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea
| | - Jun-Young Lee
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea.
| | - Seong A Shin
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea
| | - Yu Kyeong Kim
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea
| | - Koung Mi Kang
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea
| | - Chul-Ho Sohn
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea
| | - Dong Young Lee
- From the Department of Neuropsychiatry (J.W.K.), Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, Gyeonggi-do; Department of Psychiatry (J.W.K.), Hallym University College of Medicine, Chuncheon, Gangwan-do; Institute of Human Behavioral Medicine (M.S.B., D.Y., D.Y.L.), Medical Research Center Seoul National University; Departments of Neuropsychiatry (J.H.L., D.Y.L.) and Radiology (K.M.K., C.-H.S.), Seoul National University Hospital; Department of Psychiatry (S.Y.J.), Chungnam National University Hospital, Daejeon; Sanggye Paik Hospital (B.K.S.), Department of Psychiatry, Inje University College of Medicine; Departments of Neuropsychiatry (J.-Y.L.) and Nuclear Medicine (S.A.S., Y.K.K.), SMG-SNU Boramae Medical Center; and Department of Psychiatry (J.-Y.L., D.Y.L.), Seoul National University College of Medicine, Republic of Korea.
| | | |
Collapse
|
25
|
Delabar JM, Ortner M, Simon S, Wijkhuisen A, Feraudet‐Tarisse C, Pegon J, Vidal E, Hirschberg Y, Dubois B, Potier M. Altered age-linked regulation of plasma DYRK1A in elderly cognitive complainers (INSIGHT-preAD study) with high brain amyloid load. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12046. [PMID: 32642550 PMCID: PMC7331462 DOI: 10.1002/trc2.12046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION An effective therapy has not yet been developed for Alzheimer's disease (AD), in part because pathological changes occur years before clinical symptoms manifest. We recently showed that decreased plasma DYRK1A identifies individuals with mild cognitive impairment (MCI) or AD, and that aged mice have higher DYRK1A levels. METHODS We assessed DYRK1A in plasma in young/aged controls and in elderly cognitive complainers with low (L) and high (H) brain amyloid load. RESULTS DYRK1A level increases with age in humans. However, plasma from elderly individuals reporting cognitive complaints showed that the H group had the same DYRK1A level as young adults, suggesting that the age-associated DYRK1A increase is blocked in this group. L and H groups had similar levels of clusterin. DISCUSSION These results are reflective of early changes in the brain. These observations suggest that plasma DYRK1A and not clusterin could be used to classify elderly memory complainers for risk for amyloid beta pathology.
Collapse
Affiliation(s)
- Jean M. Delabar
- INSERM U 1127, CNRS UMR 7225UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et la Moelle épinière, ICMSorbonne UniversitésParisFrance
- Brain & Spine Institute (ICM) CNRS UMR7225INSERM UMRS 975ParisFrance
| | - Marion Ortner
- Department of Psychiatry and Psychotherapy, School of Medicine, Klinikum rechts der IsarTechnical University of MunichMunichGermany
| | - Stephanie Simon
- CEA, DSV, iBiTec‐SLaboratoire d'études et de recherches en immunoanalyseGif‐sur‐YvetteFrance
| | - Anne Wijkhuisen
- CEA, DSV, iBiTec‐SLaboratoire d'études et de recherches en immunoanalyseGif‐sur‐YvetteFrance
| | - Cecile Feraudet‐Tarisse
- CEA, DSV, iBiTec‐SLaboratoire d'études et de recherches en immunoanalyseGif‐sur‐YvetteFrance
| | - Jonathan Pegon
- INSERM U 1127, CNRS UMR 7225UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et la Moelle épinière, ICMSorbonne UniversitésParisFrance
- Brain & Spine Institute (ICM) CNRS UMR7225INSERM UMRS 975ParisFrance
| | - Emma Vidal
- INSERM U 1127, CNRS UMR 7225UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et la Moelle épinière, ICMSorbonne UniversitésParisFrance
- Brain & Spine Institute (ICM) CNRS UMR7225INSERM UMRS 975ParisFrance
| | - Yael Hirschberg
- INSERM U 1127, CNRS UMR 7225UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et la Moelle épinière, ICMSorbonne UniversitésParisFrance
- Brain & Spine Institute (ICM) CNRS UMR7225INSERM UMRS 975ParisFrance
| | - Bruno Dubois
- Department of NeurologyCenter of excellence of neurodegenerative disease (CoEN) and National Reference Center for Rare or Early Dementias Pitié‐Salpêtrière Hospital, AP‐HPInstitute of Memory and Alzheimer's Disease (IM2A)Boulevard de l'hôpitalParisFrance
| | - Marie‐Claude Potier
- INSERM U 1127, CNRS UMR 7225UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et la Moelle épinière, ICMSorbonne UniversitésParisFrance
- Brain & Spine Institute (ICM) CNRS UMR7225INSERM UMRS 975ParisFrance
| |
Collapse
|
26
|
Diouf I, Bush AI, Ayton S. Cerebrospinal fluid ceruloplasmin levels predict cognitive decline and brain atrophy in people with underlying β-amyloid pathology. Neurobiol Dis 2020; 139:104810. [PMID: 32087292 PMCID: PMC7150625 DOI: 10.1016/j.nbd.2020.104810] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 02/03/2020] [Accepted: 02/18/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES The mechanisms leading to neurodegeneration in Alzheimer's disease (AD) may involve oxidative stress and neuroinflammation. Ceruloplasmin (Cp) is a circulating protein that intersects both these pathways, since its expression is increased during the acute phase response, and the protein acts to lower pro-oxidant iron in cells. Since the role of Cp in AD, and its potential for use as a biomarker is not established, we investigated CSF Cp and its association with longitudinal outcome measures related to AD. METHODS This was an observational study of 268 people from the Alzheimer's Disease Neuroimaging (ADNI) cohort. Subjects were classified clinically as having AD, mild cognitive impairment (MCI) or were cognitively normal (CN), and were also classified as being positive for β-amyloid using established thresholds in the CSF t-tau/Aβ42 ratio. Subjects underwent cognitive tests and MRI studies every 6 months for 2 years, then yearly thereafter for up to 6 years. RESULTS At baseline, CSF Cp was not associated with clinical or pathological diagnosis, but we found an unexpected association between CSF Cp and levels of CSF apolipoprotein E. In longitudinal analysis, high level of CSF Cp was associated with accelerated cognitive decline (as assessed by ADAS-Cog, CDR-SB, and MMSE) and ventricular volume enlargement in people classified as MCI and who had underlying β-amyloid pathology. CONCLUSION These results raise new questions into the role of Cp in neuroinflammation, oxidative stress, and APOE pathways involved in AD, and reveal the potential for this protein to be used as a biomarker in disease prognostication.
Collapse
Affiliation(s)
- Ibrahima Diouf
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia; CSIRO Health and Biosecurity/Australian E-Health Research Centre, Brisbane, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
27
|
Ashton NJ, Hye A, Rajkumar AP, Leuzy A, Snowden S, Suárez-Calvet M, Karikari TK, Schöll M, La Joie R, Rabinovici GD, Höglund K, Ballard C, Hortobágyi T, Svenningsson P, Blennow K, Zetterberg H, Aarsland D. An update on blood-based biomarkers for non-Alzheimer neurodegenerative disorders. Nat Rev Neurol 2020; 16:265-284. [PMID: 32322100 DOI: 10.1038/s41582-020-0348-0] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2020] [Indexed: 01/11/2023]
Abstract
Cerebrospinal fluid analyses and neuroimaging can identify the underlying pathophysiology at the earliest stage of some neurodegenerative disorders, but do not have the scalability needed for population screening. Therefore, a blood-based marker for such pathophysiology would have greater utility in a primary care setting and in eligibility screening for clinical trials. Rapid advances in ultra-sensitive assays have enabled the levels of pathological proteins to be measured in blood samples, but research has been predominantly focused on Alzheimer disease (AD). Nonetheless, proteins that were identified as potential blood-based biomarkers for AD, for example, amyloid-β, tau, phosphorylated tau and neurofilament light chain, are likely to be relevant to other neurodegenerative disorders that involve similar pathological processes and could also be useful for the differential diagnosis of clinical symptoms. This Review outlines the neuropathological, clinical, molecular imaging and cerebrospinal fluid features of the most common neurodegenerative disorders outside the AD continuum and gives an overview of the current status of blood-based biomarkers for these disorders.
Collapse
Affiliation(s)
- Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Abdul Hye
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Anto P Rajkumar
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK.,Institute of Mental Health, University of Nottingham, Nottingham, UK
| | - Antoine Leuzy
- Clinical Memory Research Unit, Lund University, Malmö, Sweden
| | - Stuart Snowden
- Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Marc Suárez-Calvet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Catalonia, Spain.,Department of Neurology, Hospital del Mar, Barcelona, Catalonia, Spain
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Clinical Memory Research Unit, Lund University, Malmö, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Renaud La Joie
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Gil D Rabinovici
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Kina Höglund
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Disease Research, Neurogeriatrics Division, Karolinska Institutet, Novum, Huddinge, Stockholm, Sweden
| | | | - Tibor Hortobágyi
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,MTA-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, University of Debrecen, Debrecen, Hungary
| | - Per Svenningsson
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Kaj Blennow
- 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
| | - 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
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK. .,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK. .,Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.
| |
Collapse
|
28
|
Update on Treatments for Cognitive Decline in Alzheimer’s Disease. J Nurse Pract 2020. [DOI: 10.1016/j.nurpra.2019.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
29
|
Nandakumar A, Xing Y, Aranha RR, Faridi A, Kakinen A, Javed I, Koppel K, Pilkington EH, Purcell AW, Davis TP, Faridi P, Ding F, Ke PC. Human Plasma Protein Corona of Aβ Amyloid and Its Impact on Islet Amyloid Polypeptide Cross-Seeding. Biomacromolecules 2020; 21:988-998. [PMID: 31909987 PMCID: PMC7067050 DOI: 10.1021/acs.biomac.9b01650] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is the most severe form of neurological disorder, characterized by the presence of extracellular amyloid-β (Aβ) plaques and intracellular tau tangles. For decades, therapeutic strategies against the pathological symptoms of AD have often relied on the delivery of monoclonal antibodies to target specifically Aβ amyloid or oligomers, largely to no avail. Aβ can be traced in the brain as well as in cerebrospinal fluid and the circulation, giving rise to abundant opportunities to interact with their environmental proteins. Using liquid chromatography tandem-mass spectrometry, here we identified for the first time the protein coronae of the two major amyloid forms of Aβ-Aβ1-42 and Aβ1-40-exposed to human blood plasma. Out of the proteins identified in all groups, 58 proteins were unique to the Aβ1-42 samples and 31 proteins unique to the Aβ1-40 samples. Both fibrillar coronae consisted of proteins significant in complement activation, inflammation, and protein metabolic pathways involved in the pathology of AD. Structure-wise, the coronal proteins often possessed multidomains of high flexibility to maximize their association with the amyloid fibrils. The protein corona hindered recognition of Aβ1-42 fibrils by their structurally specific antibodies and accelerated the aggregation but not the β-cell toxicity of human islet amyloid polypeptide, the peptide associated with type 2 diabetes. This study highlights the importance of understanding the structural, functional, and pathological implications of the amyloid protein corona for the development of therapeutics against AD and a range of amyloid diseases.
Collapse
Affiliation(s)
- Aparna Nandakumar
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences , Monash University , 381 Royal Parade , Parkville , Victoria 3052 , Australia
| | - Yanting Xing
- Department of Physics and Astronomy , Clemson University , Clemson , South Carolina 29634 , United States
| | - Ritchlynn R Aranha
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute , Monash University , Clayton , Victoria 3800 , Australia
| | - Ava Faridi
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences , Monash University , 381 Royal Parade , Parkville , Victoria 3052 , Australia
| | - Aleksandr Kakinen
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences , Monash University , 381 Royal Parade , Parkville , Victoria 3052 , Australia
| | - Ibrahim Javed
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences , Monash University , 381 Royal Parade , Parkville , Victoria 3052 , Australia
| | - Kairi Koppel
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences , Monash University , 381 Royal Parade , Parkville , Victoria 3052 , Australia
| | - Emily H Pilkington
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences , Monash University , 381 Royal Parade , Parkville , Victoria 3052 , Australia
| | - Anthony Wayne Purcell
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute , Monash University , Clayton , Victoria 3800 , Australia
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences , Monash University , 381 Royal Parade , Parkville , Victoria 3052 , Australia
- Australian Institute for Bioengineering and Nanotechnology , The University of Queensland , Brisbane , Queensland 4072 , Australia
| | - Pouya Faridi
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute , Monash University , Clayton , Victoria 3800 , Australia
| | - Feng Ding
- Department of Physics and Astronomy , Clemson University , Clemson , South Carolina 29634 , United States
| | - Pu Chun Ke
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences , Monash University , 381 Royal Parade , Parkville , Victoria 3052 , Australia
| |
Collapse
|
30
|
Westwood S, Baird AL, Anand SN, Nevado-Holgado AJ, Kormilitzin A, Shi L, Hye A, Ashton NJ, Morgan AR, Bos I, Vos SJ, Baker S, Buckley NJ, Ten Kate M, Scheltens P, Teunissen CE, Vandenberghe R, Gabel S, Meersmans K, Engelborghs S, De Roeck EE, Sleegers K, Frisoni GB, Blin O, Richardson JC, Bordet R, Molinuevo JL, Rami L, Wallin A, Kettunen P, Tsolaki M, Verhey F, Lléo A, Sala I, Popp J, Peyratout G, Martinez-Lage P, Tainta M, Johannsen P, Freund-Levi Y, Frölich L, Dobricic V, Legido-Quigley C, Bertram L, Barkhof F, Zetterberg H, Morgan BP, Streffer J, Visser PJ, Lovestone S. Validation of Plasma Proteomic Biomarkers Relating to Brain Amyloid Burden in the EMIF-Alzheimer's Disease Multimodal Biomarker Discovery Cohort. J Alzheimers Dis 2020; 74:213-225. [PMID: 31985466 PMCID: PMC7175945 DOI: 10.3233/jad-190434] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We have previously investigated, discovered, and replicated plasma protein biomarkers for use to triage potential trials participants for PET or cerebrospinal fluid measures of Alzheimer's disease (AD) pathology. This study sought to undertake validation of these candidate plasma biomarkers in a large, multi-center sample collection. Targeted plasma analyses of 34 proteins with prior evidence for prediction of in vivo pathology were conducted in up to 1,000 samples from cognitively healthy elderly individuals, people with mild cognitive impairment, and in patients with AD-type dementia, selected from the EMIF-AD catalogue. Proteins were measured using Luminex xMAP, ELISA, and Meso Scale Discovery assays. Seven proteins replicated in their ability to predict in vivo amyloid pathology. These proteins form a biomarker panel that, along with age, could significantly discriminate between individuals with high and low amyloid pathology with an area under the curve of 0.74. The performance of this biomarker panel remained consistent when tested in apolipoprotein E ɛ4 non-carrier individuals only. This blood-based panel is biologically relevant, measurable using practical immunocapture arrays, and could significantly reduce the cost incurred to clinical trials through screen failure.
Collapse
Affiliation(s)
| | | | | | | | | | - Liu Shi
- Department of Psychiatry, University of Oxford, UK
| | - Abdul Hye
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Nicholas J. Ashton
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular & Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | - Isabelle Bos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Stephanie J.B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | | | | | - Mara Ten Kate
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Philip Scheltens
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | | | | | - Silvy Gabel
- University Hospital Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Belgium
| | - Karen Meersmans
- University Hospital Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Belgium
| | - Sebastiaan Engelborghs
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, University of Antwerp, Antwerp, Belgium
- Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Ellen E. De Roeck
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, University of Antwerp, Antwerp, Belgium
- Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Kristel Sleegers
- Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Belgium
| | - Giovanni B. Frisoni
- University of Geneva, Geneva, Switzerland
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Olivier Blin
- AIX Marseille University, INS, Ap-Hm, Marseille, France
| | | | | | - José L. Molinuevo
- Alzheimer’s Disease & Other Cognitive Disorders Unit, Hopsital Clínic-IDIBAPS, Barcelona, Spain
- Barcelona Beta Brain Research Center, Unversitat Pompeu Fabra, Barcelona, Spain
| | - Lorena Rami
- Barcelona Beta Brain Research Center, Unversitat Pompeu Fabra, Barcelona, Spain
| | - Anders Wallin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petronella Kettunen
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Magda Tsolaki
- 1st Department of Neurology, AHEPA University Hospital, Makedonia, Thessaloniki, Greece
| | - Frans Verhey
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Alberto Lléo
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Isabel Sala
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Julius Popp
- University Hospital of Lausanne, Lausanne, Switzerland
- Geriatric Psychiatry, Department of Mental Health and Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | | | - Pablo Martinez-Lage
- Geriatric Psychiatry, Department of Mental Health and Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | | | - Peter Johannsen
- Danish Dementia Research Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Yvonne Freund-Levi
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
- Department of Neurobiology, Caring Sciences and Society (NVS), Division of Clinical Geriatrics, Karolinska Institutet, and Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Zentralinstitut für Seelische Gesundheit, University of Heidelberg, Mannheim, Germany
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Cristina Legido-Quigley
- Kings College London, London, UK
- The Systems Medicine Group, Steno Diabetes Center, Gentofte, Denmark
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherland
- UCL Institutes of Neurology and Healthcare Engineering, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - B. Paul Morgan
- Dementia Research Institute Cardiff, Cardiff University, Cardiff, UK
| | - Johannes Streffer
- Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- UCB, Braine-l’Alleud, Belgium, formerly Janssen R&D, LLC. Beerse, Belgium at the Time of Study Conduct
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, UK
- Janssen R&D, UK formerly affiliation (1) at the Time of the Study Conduct
| |
Collapse
|
31
|
Startin CM, Ashton NJ, Hamburg S, Hithersay R, Wiseman FK, Mok KY, Hardy J, Lleó A, Lovestone S, Parnetti L, Zetterberg H, Hye A, Strydom A. Plasma biomarkers for amyloid, tau, and cytokines in Down syndrome and sporadic Alzheimer's disease. Alzheimers Res Ther 2019; 11:26. [PMID: 30902060 PMCID: PMC6429702 DOI: 10.1186/s13195-019-0477-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/21/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Down syndrome (DS), caused by chromosome 21 trisomy, is associated with an ultra-high risk of dementia due to Alzheimer's disease (AD), driven by amyloid precursor protein (APP) gene triplication. Understanding relevant molecular differences between those with DS, those with sporadic AD (sAD) without DS, and controls will aid in understanding AD development in DS. We explored group differences in plasma concentrations of amyloid-β peptides and tau (as their accumulation is a characteristic feature of AD) and cytokines (as the inflammatory response has been implicated in AD development, and immune dysfunction is common in DS). METHODS We used ultrasensitive assays to compare plasma concentrations of the amyloid-β peptides Aβ40 and Aβ42, total tau (t-tau), and the cytokines IL1β, IL10, IL6, and TNFα between adults with DS (n = 31), adults with sAD (n = 27), and controls age-matched to the group with DS (n = 27), and explored relationships between molecular concentrations and with age within each group. In the group with DS, we also explored relationships with neurofilament light (NfL) concentration, due to its potential use as a biomarker for AD in DS. RESULTS Aβ40, Aβ42, and IL1β concentrations were higher in DS, with a higher Aβ42/Aβ40 ratio in controls. The group with DS showed moderate positive associations between concentrations of t-tau and both Aβ42 and IL1β. Only NfL concentration in the group with DS showed a significant positive association with age. CONCLUSIONS Concentrations of Aβ40 and Aβ42 were much higher in adults with DS than in other groups, reflecting APP gene triplication, while no difference in the Aβ42/Aβ40 ratio between those with DS and sAD may indicate similar processing and deposition of Aβ40 and Aβ42 in these groups. Higher concentrations of IL1β in DS may reflect an increased vulnerability to infections and/or an increased prevalence of autoimmune disorders, while the positive association between IL1β and t-tau in DS may indicate IL1β is associated with neurodegeneration. Finally, NfL concentration may be the most suitable biomarker for dementia progression in DS. The identification of such a biomarker is important to improve the detection of dementia and monitor its progression, and for designing clinical intervention studies.
Collapse
Affiliation(s)
- Carla M. Startin
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
- Division of Psychiatry, University College London, London, UK
- The LonDownS Consortium (London Down Syndrome Consortium), London, UK
| | - Nicholas J. Ashton
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- NIHR Biomedical Research Centre for Mental Health, Biomedical Research Unit for Dementia at South London, and Maudsley NHS Foundation, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular & Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Sarah Hamburg
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
- Division of Psychiatry, University College London, London, UK
- The LonDownS Consortium (London Down Syndrome Consortium), London, UK
| | - Rosalyn Hithersay
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
- Division of Psychiatry, University College London, London, UK
- The LonDownS Consortium (London Down Syndrome Consortium), London, UK
| | - Frances K. Wiseman
- The LonDownS Consortium (London Down Syndrome Consortium), London, UK
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
| | - Kin Y. Mok
- The LonDownS Consortium (London Down Syndrome Consortium), London, UK
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, SAR People’s Republic of China
| | - John Hardy
- The LonDownS Consortium (London Down Syndrome Consortium), London, UK
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
- Reta Lila Weston Institute, Institute of Neurology, University College London, London, UK
| | - Alberto Lleó
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Lucilla Parnetti
- Centre for Memory Disturbances, Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
| | - Abdul Hye
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- NIHR Biomedical Research Centre for Mental Health, Biomedical Research Unit for Dementia at South London, and Maudsley NHS Foundation, London, UK
| | - André Strydom
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF UK
- Division of Psychiatry, University College London, London, UK
- The LonDownS Consortium (London Down Syndrome Consortium), London, UK
| |
Collapse
|