1
|
Wang X, Freiesleben SD, Schneider LS, Preis L, Priller J, Spruth EJ, Altenstein S, Schneider A, Fliessbach K, Wiltfang J, Hansen N, Jessen F, Rostamzadeh A, Duzel E, Glanz W, Incesoy EI, Buerger K, Janowitz D, Ewers M, Perneczky R, Rauchmann BS, Teipel SJ, Kilimann I, Goerss D, Laske C, Munk MHJ, Spottke A, Roy-Kluth N, Heneka MT, Brosseron F, Wagner M, Wolfsgruber S, Ramirez A, Kleineidam L, Stark M, Peters O. Association of Neurogranin and BACE1 With Clinical Cognitive Decline in Individuals With Subjective Cognitive Decline. Neurology 2024; 103:e209806. [PMID: 39303184 DOI: 10.1212/wnl.0000000000209806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND AND OBJECTIVES CSF biomarkers have immense diagnostic and prognostic potential for Alzheimer disease (AD). However, AD is still diagnosed relatively late in the disease process, sometimes even years after the initial manifestation of cognitive symptoms. Thus, further identification of biomarkers is required to detect related pathology in the preclinical stage and predict cognitive decline. Our study aimed to assess the association of neurogranin and β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) with cognitive decline in individuals with subjective cognitive decline (SCD). METHODS We enrolled participants with available neurogranin and BACE1 measurements in CSF from the DELCODE (DZNE-Longitudinal Cognitive Impairment and Dementia, Germany) cohort. The longitudinal change of Preclinical Alzheimer's Cognitive Composite score was assessed as the primary outcome in participants with SCD and controls. The secondary outcome was defined as conversion of SCD to mild cognitive impairment (MCI) during follow-up. Levels of neurogranin, BACE1, and neurogranin/BACE1 ratio across groups were compared by analysis of covariance after adjustment for demographics. The linear mixed-effects model and Cox regression analysis were applied to evaluate their association with cognitive decline and progression of SCD to MCI, respectively. RESULTS A total of 530 participants (mean age: 70.76 ± 6.01 years, 48.7% female) were analyzed in the study. The rate of cognitive decline was faster in individuals with SCD with higher neurogranin and neurogranin/BACE1 ratio (β = -0.138, SE = 0.065, p = 0.037, and β = -0.293, SE = 0.115, p = 0.013). Higher baseline neurogranin and neurogranin/BACE1 ratio were associated with an increased rate of conversion from SCD to MCI (hazard ratio [HR] 1.35 per SD, 95% CI 1.03-1.77, p = 0.028, and HR 1.53 per SD, 95% CI 1.13-2.07, p = 0.007). In addition, the impact of higher neurogranin levels on accelerating the rate of cognitive decline was more pronounced in the SCD group than in cognitively unimpaired controls (β = -0.077, SE = 0.033, p = 0.020). DISCUSSION Our findings suggest that CSF neurogranin and BACE1 begin to change in the preclinical stage of AD and they are associated with clinical progression in individuals with SCD.
Collapse
Affiliation(s)
- Xiao Wang
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Silka D Freiesleben
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Luisa-Sophie Schneider
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Lukas Preis
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Josef Priller
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Eike J Spruth
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Slawek Altenstein
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Anja Schneider
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Klaus Fliessbach
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Jens Wiltfang
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Niels Hansen
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Frank Jessen
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Ayda Rostamzadeh
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Emrah Duzel
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Wenzel Glanz
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Enise I Incesoy
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Katharina Buerger
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Daniel Janowitz
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Michael Ewers
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Robert Perneczky
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Boris-Stephan Rauchmann
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Stefan J Teipel
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Ingo Kilimann
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Doreen Goerss
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Christoph Laske
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Matthias H J Munk
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Annika Spottke
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Nina Roy-Kluth
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Michael T Heneka
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Frederic Brosseron
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Michael Wagner
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Steffen Wolfsgruber
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Alfredo Ramirez
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Luca Kleineidam
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Melina Stark
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| | - Oliver Peters
- From the Department of Psychiatry and Neurosciences (X.W., S.D.F., L.-S.S., L.P., O.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Experimental and Clinical Research Center (ECRC) (X.W., S.D.F., L.-S.S., L.P., O.P.); German Center for Neurodegenerative Diseases (DZNE) (S.D.F., J.P., E.J.S., S.A., O.P.); Department of Psychiatry and Psychotherapy (J.P., E.J.S., S.A.), Charité, Berlin; Department of Psychiatry and Psychotherapy (J.P.), School of Medicine, Technical University of Munich, Germany; University of Edinburgh and UK DRI (J.P.), United Kingdom; German Center for Neurodegenerative Diseases (DZNE) (A. Schneider, K.F., F.J., A. Spottke, N.R.-K., F.B., M.W., S.W., A. Ramirez, L.K., M.S.), Bonn; Department of Neurodegenerative Disease and Geriatric Psychiatry (A. Schneider, K.F., M.W., S.W., A. Ramirez, L.K., M.S.), University of Bonn Medical Center; German Center for Neurodegenerative Diseases (DZNE) (J.W.), Goettingen; Department of Psychiatry and Psychotherapy (J.W., N.H.), University Medical Center Goettingen, University of Goettingen, Germany; Neurosciences and Signaling Group (J.W.), Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Portugal; Department of Psychiatry (F.J., A. Rostamzadeh), Medical Faculty, University of Cologne; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) (F.J., A. Ramirez), University of Cologne, Köln; German Center for Neurodegenerative Diseases (DZNE) (E.D., W.G., E.I.I.), Magdeburg; Institute of Cognitive Neurology and Dementia Research (IKND) (E.D., E.I.I.), Otto-von-Guericke University, Magdeburg; Department for Psychiatry and Psychotherapy (E.I.I.), University Clinic Magdeburg; German Center for Neurodegenerative Diseases (DZNE) (K.B., M.E., R.P.), Munich; Institute for Stroke and Dementia Research (ISD) (K.B., D.J., M.E.), and Department of Psychiatry and Psychotherapy (R.P., B.-S.R.), University Hospital, LMU Munich; Munich Cluster for Systems Neurology (SyNergy) (R.P.), Germany; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R.), University of Sheffield, United Kingdom; Department of Neuroradiology (B.-S.R.), University Hospital, LMU Munich; German Center for Neurodegenerative Diseases (DZNE) (S.J.T., I.K., D.G.), Rostock; Department of Psychosomatic Medicine (S.J.T., I.K., D.G.), Rostock University Medical Center; German Center for Neurodegenerative Diseases (DZNE) (C.L., M.H.J.M.), Tübingen; Section for Dementia Research (C.L.), Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy (C.L., M.H.J.M.), University of Tübingen; Department of Neurology (A. Spottke), University of Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB) (M.T.H.), University of Luxembourg, Belvaux; Division of Neurogenetics and Molecular Psychiatry (A. Ramirez), Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; and Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (A. Ramirez), San Antonio, TX
| |
Collapse
|
2
|
Smith RG, Pishva E, Kouhsar M, Imm J, Dobricic V, Johannsen P, Wittig M, Franke A, Vandenberghe R, Schaeverbeke J, Freund‐Levi Y, Frölich L, Scheltens P, Teunissen CE, Frisoni G, Blin O, Richardson JC, Bordet R, Engelborghs S, de Roeck E, Martinez‐Lage P, Altuna M, Tainta M, Lleó A, Sala I, Popp J, Peyratout G, Winchester L, Nevado‐Holgado A, Verhey F, Tsolaki M, Andreasson U, Blennow K, Zetterberg H, Streffer J, Vos SJB, Lovestone S, Visser PJ, Bertram L, Lunnon K. Blood DNA methylomic signatures associated with CSF biomarkers of Alzheimer's disease in the EMIF-AD study. Alzheimers Dement 2024; 20:6722-6739. [PMID: 39193893 PMCID: PMC11485320 DOI: 10.1002/alz.14098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION We investigated blood DNA methylation patterns associated with 15 well-established cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease (AD) pathophysiology, neuroinflammation, and neurodegeneration. METHODS We assessed DNA methylation in 885 blood samples from the European Medical Information Framework for Alzheimer's Disease (EMIF-AD) study using the EPIC array. RESULTS We identified Bonferroni-significant differential methylation associated with CSF YKL-40 (five loci) and neurofilament light chain (NfL; seven loci) levels, with two of the loci associated with CSF YKL-40 levels correlating with plasma YKL-40 levels. A co-localization analysis showed shared genetic variants underlying YKL-40 DNA methylation and CSF protein levels, with evidence that DNA methylation mediates the association between genotype and protein levels. Weighted gene correlation network analysis identified two modules of co-methylated loci correlated with several amyloid measures and enriched in pathways associated with lipoproteins and development. DISCUSSION We conducted the most comprehensive epigenome-wide association study (EWAS) of AD-relevant CSF biomarkers to date. Future work should explore the relationship between YKL-40 genotype, DNA methylation, and protein levels in the brain. HIGHLIGHTS Blood DNA methylation was assessed in the EMIF-AD MBD study. Epigenome-wide association studies (EWASs) were performed for 15 Alzheimer's disease (AD)-relevant cerebrospinal fluid (CSF) biomarker measures. Five Bonferroni-significant loci were associated with YKL-40 levels and seven with neurofilament light chain (NfL). DNA methylation in YKL-40 co-localized with previously reported genetic variation. DNA methylation potentially mediates the effect of single-nucleotide polymorphisms (SNPs) in YKL-40 on CSF protein levels.
Collapse
Affiliation(s)
- Rebecca G. Smith
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Ehsan Pishva
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML)Maastricht UniversityMaastrichtThe Netherlands
| | - Morteza Kouhsar
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Jennifer Imm
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA)University of LübeckLübeckGermany
| | - Peter Johannsen
- Danish Dementia Research Centre, RigshospitaletCopenhagenDenmark
| | - Michael Wittig
- Institute of Clinical Molecular BiologyChristian‐Albrechts‐University of KielKielGermany
| | - Andre Franke
- Institute of Clinical Molecular BiologyChristian‐Albrechts‐University of KielKielGermany
| | - Rik Vandenberghe
- Laboratory for Cognitive NeurologyKU Leuven, Leuven Brain InstituteLeuvenBelgium
| | - Jolien Schaeverbeke
- Laboratory for Cognitive NeurologyKU Leuven, Leuven Brain InstituteLeuvenBelgium
| | - Yvonne Freund‐Levi
- Department of Clinical Science and EducationSödersjukhuset, Karolinska InstitutetStockholmSweden
- School of Medical SciencesÖrebro UniversityÖrebroSweden
- Department of GeriatricsSödertälje HospitalSödertäljeSweden
| | - Lutz Frölich
- Department of Geriatric PsychiatryCentral Institut of Mental HealthMedical Faculty Mannheim/Heidelberg UniversityMannheimGermany
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam NeuroscienceVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Laboratory Medicine, Amsterdam NeuroscienceVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Giovanni Frisoni
- Memory centerGeneva University and University Hospitals; on behalf of the AMYPAD consortiumGenevaSwitzerland
| | | | - Jill C. Richardson
- Neuroscience Therapeutic Area, GlaxoSmithKline R&DStevenageHertfordshireUK
| | | | - Sebastiaan Engelborghs
- Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N)Vrije Universiteit Brussel (VUB), JetteBrusselsBelgium
| | - Ellen de Roeck
- Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| | - Pablo Martinez‐Lage
- Center for Research and Advanced TherapiesFundación CITA‐Alzhéimer FundazioaSan SebastianGipuzkoaSpain
| | - Miren Altuna
- Center for Research and Advanced TherapiesFundación CITA‐Alzhéimer FundazioaSan SebastianGipuzkoaSpain
| | - Mikel Tainta
- Center for Research and Advanced TherapiesFundación CITA‐Alzhéimer FundazioaSan SebastianGipuzkoaSpain
| | - Alberto Lleó
- Servicio de Neurología, Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED)Hospital Sant PauBarcelonaSpain
| | - Isabel Sala
- Servicio de Neurología, Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED)Hospital Sant PauBarcelonaSpain
| | - Julius Popp
- University Hospital of Psychiatry Zürich, University of ZürichZürichSwitzerland
| | - Gwendoline Peyratout
- Department of PsychiatryUniversity Hospital of Lausanne (CHUV)LausanneSwitzerland
| | | | | | - Frans Verhey
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML)Maastricht UniversityMaastrichtThe Netherlands
| | - Magda Tsolaki
- 1st Department of NeurologySchool of MedicineLaboratory of Neurodegenerative DiseasesCenter for Interdisciplinary Research and InnovationAristotle University of Thessaloniki, and Alzheimer HellasThessalonikiGreece
| | - Ulf Andreasson
- Institute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgGöteborgSweden
| | - Kaj Blennow
- Institute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgGöteborgSweden
- Paris Brain InstituteICM, Pitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research CenterDivision of Life Sciences and Medicineand Department of NeurologyInstitute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiPR China
| | - Henrik Zetterberg
- Institute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgGöteborgSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative Diseases, N.T.ShatinHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public Health, University of Wisconsin‐MadisonMadisonWisconsinUSA
| | | | - Stephanie J. B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML)Maastricht UniversityMaastrichtThe Netherlands
| | - Simon Lovestone
- Department of PsychiatryUniversity Hospital of Lausanne (CHUV)LausanneSwitzerland
- Currently at: Johnson & Johnson Innovative MedicinesBeerseBelgium
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML)Maastricht UniversityMaastrichtThe Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam NeuroscienceVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA)University of LübeckLübeckGermany
| | - Katie Lunnon
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| |
Collapse
|
3
|
Muqaku B, Anderl-Straub S, Werner L, Nagl M, Otto M, Teunissen CE, Oeckl P. Contactin proteins in cerebrospinal fluid show different alterations in dementias. J Neurol 2024:10.1007/s00415-024-12694-6. [PMID: 39317877 DOI: 10.1007/s00415-024-12694-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND The proteins contactin (CNTN) 1-6 are synaptic proteins for which there is evidence that they are dysregulated in neurodegenerative dementias. Less is known about CNTN changes and differences in cerebrospinal fluid (CSF) of dementias, which can provide important information about alterations of the CNTN network and be of value for differential diagnosis. METHODS We developed a mass spectrometry-based multiple reaction monitoring (MRM) method to simultaneously determine all six CNTNs in CSF samples using stable isotope-labeled standard peptides. The analytical performance of the method was evaluated for peptide stability, dilution linearity and precision. CNTNs were measured in 82 CSF samples from patients with Alzheimer's disease (AD, n = 19), behavioural variant frontotemporal dementia (bvFTD, n = 18), Parkinson's disease dementia/dementia with Lewy bodies (PDD/DLB, n = 18) and non-neurodegenerative controls (n = 27) and compared with core AD biomarkers. RESULTS The MRM analysis revealed down-regulation of CNTN2 (fold change (FC) = 0.77), CNTN4 (FC = 0.75) and CNTN5 (FC = 0.67) in bvFTD and CNTN3 (FC = 0.72), CNTN4 (FC = 0.75) and CNTN5 (FC = 0.73) in PDD/DLB compared to AD. CNTN levels strongly correlated with each other in controls (r = 0.73), bvFTD (r = 0.86) and PDD/DLB (r = 0.70), but the correlation was significantly lower in AD (r = 0.41). CNTNs in AD did not show correlation even with core AD biomarkers. Combined use of CNTN1-6 levels increased diagnostic performance of AD core biomarkers. CONCLUSIONS Our data show CNTNs differentially altered in dementias and indicate CNTN homeostasis being selectively dysregulated in AD. The combined use of CNTNs with AD core biomarkers might help to improve differential diagnosis.
Collapse
Affiliation(s)
- Besnik Muqaku
- German Center for Neurodegenerative Diseases (DZNE E.V.), Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Sarah Anderl-Straub
- Department of Neurology, Ulm University Hospital, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Leonie Werner
- Department of Neurology, Ulm University Hospital, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Magdalena Nagl
- Department of Neurology, Ulm University Hospital, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Markus Otto
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam University Medical Centers (UMC), Amsterdam, The Netherlands
| | - Patrick Oeckl
- German Center for Neurodegenerative Diseases (DZNE E.V.), Helmholtzstr. 8/1, 89081, Ulm, Germany.
- Department of Neurology, Ulm University Hospital, Helmholtzstr. 8/1, 89081, Ulm, Germany.
| |
Collapse
|
4
|
Jurasova V, Andel R, Katonova A, Veverova K, Zuntychova T, Horakova H, Vyhnalek M, Kolarova T, Matoska V, Blennow K, Hort J. CSF neurogranin levels as a biomarker in Alzheimer's disease and frontotemporal lobar degeneration: a cross-sectional analysis. Alzheimers Res Ther 2024; 16:199. [PMID: 39242539 PMCID: PMC11378641 DOI: 10.1186/s13195-024-01566-w] [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: 07/05/2024] [Accepted: 08/24/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND There is initial evidence suggesting that biomarker neurogranin (Ng) may distinguish Alzheimer's disease (AD) from other neurodegenerative diseases. Therefore, we assessed (a) the discriminant ability of cerebrospinal fluid (CSF) Ng levels to distinguish between AD and frontotemporal lobar degeneration (FTLD) pathology and between different stages within the same disease, (b) the relationship between Ng levels and cognitive performance in both AD and FTLD pathology, and (c) whether CSF Ng levels vary by apolipoprotein E (APOE) polymorphism in the AD continuum. METHODS Participants with subjective cognitive decline (SCD) (n = 33), amnestic mild cognitive impairment (aMCI) due to AD (n = 109), AD dementia (n = 67), MCI due to FTLD (n = 25), and FTLD dementia (n = 29) were recruited from the Czech Brain Aging Study. One-way analysis of covariance (ANCOVA) assessed Ng levels in diagnostic subgroups. Linear regressions evaluated the relationship between CSF Ng levels, memory scores, and APOE polymorphism. RESULTS Ng levels were higher in aMCI-AD patients compared to MCI-FTLD (F[1, 134] = 15.16, p < .001), and in AD-dementia compared to FTLD-dementia (F[1, 96] = 4.60, p = .029). Additionally, Ng levels were higher in FTLD-dementia patients compared to MCI-FTLD (F[1, 54]= 4.35, p = .034), lower in SCD participants compared to aMCI-AD (F[1, 142] = 10.72, p = .001) and AD-dementia (F[1, 100] = 20.90, p < .001), and did not differ between SCD participants and MCI-FTLD (F[1, 58]= 1.02, p = .491) or FTLD-dementia (F[1, 62]= 2.27, p = .051). The main effect of diagnosis across the diagnostic subgroups on Aβ1-42/Ng ratio was significant too (F[4, 263]=, p < .001). We found a non-significant association between Ng levels and memory scores overall (β=-0.25, p = .154) or in AD diagnostic subgroups, and non-significant differences in this association between overall AD APOE ε4 carriers and non-carriers (β=-0.32, p = .358). CONCLUSIONS In this first study to-date to assess MCI and dementia due to AD or FTLD within one study, elevated CSF Ng appears to be an early biomarker of AD-related impairment, but its role as a biomarker appears to diminish after dementia diagnosis, whereby dementia-related underlying processes in AD and FTLD may begin to merge. The Aβ1-42/Ng ratio discriminated AD from FTLD patients better than Ng alone. CSF Ng levels were not related to memory in AD or FTLD, suggesting that Ng may be a marker of the biological signs of disease state rather than cognitive deficits.
Collapse
Affiliation(s)
- Vanesa Jurasova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic.
| | - Ross Andel
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Alzbeta Katonova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
| | - Katerina Veverova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
| | - Terezie Zuntychova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
| | - Hana Horakova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Martin Vyhnalek
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Tereza Kolarova
- Department of Clinical Biochemistry, Hematology and Immunology, Homolka Hospital, Prague, Czech Republic
| | - Vaclav Matoska
- Department of Clinical Biochemistry, Hematology and Immunology, Homolka Hospital, Prague, Czech Republic
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
5
|
Schauer SP, Cho CH, Novikova G, Roth GA, Lee J, Sharma AD, Foley AR, Ng C, Shen P, Choi M, Ma TP, Phu L, Budayeva HG, Cheung TK, Lalehzadeh G, Imperio J, Ngu H, Etxeberria A, Liang Y, Rezzonico MG, Dourado M, Huang K, Lai Z, Hokom M, Pandya NJ, Newton D, Abdel‐Haleem AM, Chan P, Lee D, Tassew NG, Sangaraju D, O'Connor D, Hötzel I, Stark KL, Chou C, Foreman O, Easton A, Wildsmith KR, Sperinde G, Rose CM, Friedman BA, Fuji RN, Weimer RM, Meilandt WJ, Sadekar S, Nugent AA, Biever A. Primate cerebrospinal fluid CHI3L1 reflects brain TREM2 agonism. Alzheimers Dement 2024; 20:5861-5888. [PMID: 39090679 PMCID: PMC11497760 DOI: 10.1002/alz.13921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Triggering receptor expressed on myeloid cells 2 (TREM2) agonists are being clinically evaluated as disease-modifying therapeutics for Alzheimer's disease. Clinically translatable pharmacodynamic (PD) biomarkers are needed to confirm drug activity and select the appropriate therapeutic dose in clinical trials. METHODS We conducted multi-omic analyses on paired non-human primate brain and cerebrospinal fluid (CSF), and stimulation of human induced pluripotent stem cell-derived microglia cultures after TREM2 agonist treatment, followed by validation of candidate fluid PD biomarkers using immunoassays. We immunostained microglia to characterize proliferation and clustering. RESULTS We report CSF soluble TREM2 (sTREM2) and CSF chitinase-3-like protein 1 (CHI3L1/YKL-40) as PD biomarkers for the TREM2 agonist hPara.09. The respective reduction of sTREM2 and elevation of CHI3L1 in brain and CSF after TREM2 agonist treatment correlated with transient microglia proliferation and clustering. DISCUSSION CSF CHI3L1 and sTREM2 reflect microglial TREM2 agonism and can be used as clinical PD biomarkers to monitor TREM2 activity in the brain. HIGHLIGHTS CSF soluble triggering receptor expressed on myeloid cells 2 (sTREM2) reflects brain target engagement for a novel TREM2 agonist, hPara.09. CSF chitinase-3-like protein 1 reflects microglial TREM2 agonism. Both can be used as clinical fluid biomarkers to monitor TREM2 activity in brain.
Collapse
Affiliation(s)
- Stephen P. Schauer
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Chang Hoon Cho
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Gloriia Novikova
- Department of BioinformaticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Gillie A. Roth
- Department of Preclinical and Translational Pharmacokinetics and PharmacodynamicsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Julie Lee
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Anup D. Sharma
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Alejandro R. Foley
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Carl Ng
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Philip Shen
- Department of Safety Assessment PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Meena Choi
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Taylur P. Ma
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Lilian Phu
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Hanna G. Budayeva
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Tommy K. Cheung
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Guita Lalehzadeh
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Jose Imperio
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Hai Ngu
- Department of Research PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Ainhoa Etxeberria
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Yuxin Liang
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | | | - Michelle Dourado
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Kevin Huang
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Zijuan Lai
- Department of Drug Metabolism and PharmacokineticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Martha Hokom
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Nikhil J. Pandya
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Dwight Newton
- Roche InformaticsHoffmann‐La Roche, Ltd.MississaugaOntarioCanada
| | | | - Pamela Chan
- Department of Biochemical and Cellular PharmacologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Donna Lee
- Department of Safety Assessment ToxicologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Nardos G. Tassew
- Department of Safety Assessment ToxicologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Dewakar Sangaraju
- Department of Drug Metabolism and PharmacokineticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Deborah O'Connor
- Department of ChemistryManufacturing, and ControlsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Isidro Hötzel
- Department of Antibody EngineeringGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Kimberly L. Stark
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Carolina Chou
- Department of Safety Assessment Nonclinical OperationsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Oded Foreman
- Department of Research PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Amy Easton
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Kristin R. Wildsmith
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Gizette Sperinde
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Christopher M. Rose
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Brad A. Friedman
- Department of BioinformaticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Reina N. Fuji
- Department of Safety Assessment PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Robby M. Weimer
- Department of Translational ImagingGenentech, Inc.South San FranciscoCaliforniaUSA
| | | | - Shraddha Sadekar
- Department of Preclinical and Translational Pharmacokinetics and PharmacodynamicsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Alicia A. Nugent
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Anne Biever
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| |
Collapse
|
6
|
Arslan B, Zetterberg H, Ashton NJ. Blood-based biomarkers in Alzheimer's disease - moving towards a new era of diagnostics. Clin Chem Lab Med 2024; 62:1063-1069. [PMID: 38253262 DOI: 10.1515/cclm-2023-1434] [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: 12/13/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024]
Abstract
Alzheimer's disease (AD), a primary cause of dementia globally, is traditionally diagnosed via cerebrospinal fluid (CSF) measures and positron emission tomography (PET). The invasiveness, cost, and limited accessibility of these methods have led to exploring blood-based biomarkers as a promising alternative for AD diagnosis and monitoring. Recent advancements in sensitive immunoassays have identified potential blood-based biomarkers, such as Aβ42/Aβ40 ratios and phosphorylated tau (p-tau) species. This paper briefly evaluates the clinical utility and reliability of these biomarkers across various AD stages, highlighting challenges like refining plasma Aβ42/Aβ40 assays and enhancing the precision of p-tau, particularly p-tau181, p-tau217, and p-tau231. The discussion also covers other plasma biomarkers like neurofilament light (NfL), glial fibrillary acidic protein (GFAP), and synaptic biomarkers, assessing their significance in AD diagnostics. The need for ongoing research and development of robust assays to match the performance of CSF and PET biomarkers is underscored. In summary, blood-based biomarkers are increasingly crucial in AD diagnosis, follow-up, prognostication, treatment response evaluation, and population screening, particularly in primary care settings. These developments are set to revolutionize AD diagnostics, offering earlier and more accessible detection and management options.
Collapse
Affiliation(s)
- Burak Arslan
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, P.R. China
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Old Age Psychiatry, Psychology & Neuroscience, King's College London, Institute of Psychiatry, London, UK
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, 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
|
7
|
Karlsson L, Vogel J, Arvidsson I, Åström K, Janelidze S, Blennow K, Palmqvist S, Stomrud E, Mattsson-Carlgren N, Hansson O. Cerebrospinal fluid reference proteins increase accuracy and interpretability of biomarkers for brain diseases. Nat Commun 2024; 15:3676. [PMID: 38693142 PMCID: PMC11063138 DOI: 10.1038/s41467-024-47971-5] [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: 06/13/2023] [Accepted: 04/17/2024] [Indexed: 05/03/2024] Open
Abstract
Cerebrospinal fluid (CSF) biomarkers reflect brain pathophysiology and are used extensively in translational research as well as in clinical practice for diagnosis of neurological diseases, e.g., Alzheimer's disease (AD). However, CSF biomarker concentrations may be influenced by non-disease related inter-individual variability. Here we use a data-driven approach to demonstrate the existence of inter-individual variability in mean standardized CSF protein levels. We show that these non-disease related differences cause many commonly reported CSF biomarkers to be highly correlated, thereby producing misleading results if not accounted for. To adjust for this inter-individual variability, we identified and evaluated high-performing reference proteins which improved the diagnostic accuracy of key CSF AD biomarkers. Our reference protein method attenuates the risk for false positive findings, and improves the sensitivity and specificity of CSF biomarkers, with broad implications for both research and clinical practice.
Collapse
Affiliation(s)
- Linda Karlsson
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden.
| | - Jacob Vogel
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Clinical Sciences, Clinical Memory Research Unit, SciLifeLab, Lund University, Lund, Sweden
| | - Ida Arvidsson
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Kalle Åström
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Sebastian Palmqvist
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
8
|
Wang S, Xie S, Zheng Q, Zhang Z, Wang T, Zhang G. Biofluid biomarkers for Alzheimer's disease. Front Aging Neurosci 2024; 16:1380237. [PMID: 38659704 PMCID: PMC11039951 DOI: 10.3389/fnagi.2024.1380237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease, with a complex pathogenesis and an irreversible course. Therefore, the early diagnosis of AD is particularly important for the intervention, prevention, and treatment of the disease. Based on the different pathophysiological mechanisms of AD, the research progress of biofluid biomarkers are classified and reviewed. In the end, the challenges and perspectives of future research are proposed.
Collapse
Affiliation(s)
- Sensen Wang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Sitan Xie
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| | - Qinpin Zheng
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Zhihui Zhang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Guirong Zhang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| |
Collapse
|
9
|
Tavares-Júnior JWL, Ciurleo GCV, Feitosa EDAAF, Oriá RB, Braga-Neto P. The Clinical Aspects of COVID and Alzheimer's Disease: A Round-Up of Where Things Stand and Are Headed. J Alzheimers Dis 2024; 99:1159-1171. [PMID: 38848177 DOI: 10.3233/jad-231368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
The link between long COVID-19 and brain/cognitive impairments is concerning and may foster a worrisome worldwide emergence of novel cases of neurodegenerative diseases with aging. This review aims to update the knowledge, crosstalk, and possible intersections between the Post-COVID Syndrome (PCS) and Alzheimer's disease (AD). References included in this review were obtained from PubMed searches conducted between October 2023 and November 2023. PCS is a very heterogenous and poorly understood disease with recent evidence of a possible association with chronic diseases such as AD. However, more scientific data is required to establish the link between PCS and AD.
Collapse
Affiliation(s)
| | - Gabriella Cunha Vieira Ciurleo
- Department of Clinical Medicine, Neurology Section, Faculty of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
- Department of Morphology and Institute of Biomedicine, Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, School of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | | | - Reinaldo B Oriá
- Department of Clinical Medicine, Neurology Section, Faculty of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
- Department of Morphology and Institute of Biomedicine, Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, School of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Pedro Braga-Neto
- Department of Clinical Medicine, Neurology Section, Faculty of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
- Center of Health Sciences, State University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
10
|
Vijiaratnam N, Foltynie T. How should we be using biomarkers in trials of disease modification in Parkinson's disease? Brain 2023; 146:4845-4869. [PMID: 37536279 PMCID: PMC10690028 DOI: 10.1093/brain/awad265] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/05/2023] Open
Abstract
The recent validation of the α-synuclein seed amplification assay as a biomarker with high sensitivity and specificity for the diagnosis of Parkinson's disease has formed the backbone for a proposed staging system for incorporation in Parkinson's disease clinical studies and trials. The routine use of this biomarker should greatly aid in the accuracy of diagnosis during recruitment of Parkinson's disease patients into trials (as distinct from patients with non-Parkinson's disease parkinsonism or non-Parkinson's disease tremors). There remain, however, further challenges in the pursuit of biomarkers for clinical trials of disease modifying agents in Parkinson's disease, namely: optimizing the distinction between different α-synucleinopathies; the selection of subgroups most likely to benefit from a candidate disease modifying agent; a sensitive means of confirming target engagement; and the early prediction of longer-term clinical benefit. For example, levels of CSF proteins such as the lysosomal enzyme β-glucocerebrosidase may assist in prognostication or allow enrichment of appropriate patients into disease modifying trials of agents with this enzyme as the target; the presence of coexisting Alzheimer's disease-like pathology (detectable through CSF levels of amyloid-β42 and tau) can predict subsequent cognitive decline; imaging techniques such as free-water or neuromelanin MRI may objectively track decline in Parkinson's disease even in its later stages. The exploitation of additional biomarkers to the α-synuclein seed amplification assay will, therefore, greatly add to our ability to plan trials and assess the disease modifying properties of interventions. The choice of which biomarker(s) to use in the context of disease modifying clinical trials will depend on the intervention, the stage (at risk, premotor, motor, complex) of the population recruited and the aims of the trial. The progress already made lends hope that panels of fluid biomarkers in tandem with structural or functional imaging may provide sensitive and objective methods of confirming that an intervention is modifying a key pathophysiological process of Parkinson's disease. However, correlation with clinical progression does not necessarily equate to causation, and the ongoing validation of quantitative biomarkers will depend on insightful clinical-genetic-pathophysiological comparisons incorporating longitudinal biomarker changes from those at genetic risk with evidence of onset of the pathophysiology and those at each stage of manifest clinical Parkinson's disease.
Collapse
Affiliation(s)
- Nirosen Vijiaratnam
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| |
Collapse
|
11
|
Abdelmoaty MM, Lu E, Kadry R, Foster EG, Bhattarai S, Mosley RL, Gendelman HE. Clinical biomarkers for Lewy body diseases. Cell Biosci 2023; 13:209. [PMID: 37964309 PMCID: PMC10644566 DOI: 10.1186/s13578-023-01152-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023] Open
Abstract
Synucleinopathies are a group of neurodegenerative disorders characterized by pathologic aggregates of neural and glial α-synuclein (α-syn) in the form of Lewy bodies (LBs), Lewy neurites, and cytoplasmic inclusions in both neurons and glia. Two major classes of synucleinopathies are LB disease and multiple system atrophy. LB diseases include Parkinson's disease (PD), PD with dementia, and dementia with LBs. All are increasing in prevalence. Effective diagnostics, disease-modifying therapies, and therapeutic monitoring are urgently needed. Diagnostics capable of differentiating LB diseases are based on signs and symptoms which might overlap. To date, no specific diagnostic test exists despite disease-specific pathologies. Diagnostics are aided by brain imaging and cerebrospinal fluid evaluations, but more accessible biomarkers remain in need. Mechanisms of α-syn evolution to pathologic oligomers and insoluble fibrils can provide one of a spectrum of biomarkers to link complex neural pathways to effective therapies. With these in mind, we review promising biomarkers linked to effective disease-modifying interventions.
Collapse
Affiliation(s)
- Mai M Abdelmoaty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Eugene Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rana Kadry
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Emma G Foster
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Shaurav Bhattarai
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
12
|
Pesämaa I, Müller SA, Robinson S, Darcher A, Paquet D, Zetterberg H, Lichtenthaler SF, Haass C. A microglial activity state biomarker panel differentiates FTD-granulin and Alzheimer's disease patients from controls. Mol Neurodegener 2023; 18:70. [PMID: 37775827 PMCID: PMC10543321 DOI: 10.1186/s13024-023-00657-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/13/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND With the emergence of microglia-modulating therapies there is an urgent need for reliable biomarkers to evaluate microglial activation states. METHODS Using mouse models and human induced pluripotent stem cell-derived microglia (hiMGL), genetically modified to yield the most opposite homeostatic (TREM2-knockout) and disease-associated (GRN-knockout) states, we identified microglia activity-dependent markers. Non-targeted mass spectrometry was used to identify proteomic changes in microglia and cerebrospinal fluid (CSF) of Grn- and Trem2-knockout mice. Additionally, we analyzed the proteome of GRN- and TREM2-knockout hiMGL and their conditioned media. Candidate marker proteins were tested in two independent patient cohorts, the ALLFTD cohort (GRN mutation carriers versus non-carriers), as well as the proteomic data set available from the EMIF-AD MBD study. RESULTS We identified proteomic changes between the opposite activation states in mouse microglia and CSF, as well as in hiMGL cell lysates and conditioned media. For further verification, we analyzed the CSF proteome of heterozygous GRN mutation carriers suffering from frontotemporal dementia (FTD). We identified a panel of six proteins (FABP3, MDH1, GDI1, CAPG, CD44, GPNMB) as potential indicators for microglial activation. Moreover, we confirmed three of these proteins (FABP3, GDI1, MDH1) to be significantly elevated in the CSF of Alzheimer's (AD) patients. Remarkably, each of these markers differentiated amyloid-positive cases with mild cognitive impairment (MCI) from amyloid-negative individuals. CONCLUSIONS The identified candidate proteins reflect microglia activity and may be relevant for monitoring the microglial response in clinical practice and clinical trials modulating microglial activity and amyloid deposition. Moreover, the finding that three of these markers differentiate amyloid-positive from amyloid-negative MCI cases in the AD cohort suggests that these proteins associate with a very early immune response to seeded amyloid. This is consistent with our previous findings in the Dominantly Inherited Alzheimer's Disease Network (DIAN) cohort, where soluble TREM2 increases as early as 21 years before symptom onset. Moreover, in mouse models for amyloidogenesis, seeding of amyloid is limited by physiologically active microglia further supporting their early protective role. The biological functions of some of our main candidates (FABP3, CD44, GPNMB) also further emphasize that lipid dysmetabolism may be a common feature of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ida Pesämaa
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians-University Munich, Munich, Germany
- Department of Psychiatry & Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Sophie Robinson
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians-University Munich, Munich, Germany
- Institute for Stroke and Dementia Research, University Hospital Munich, Ludwig-Maximilians- University Munich, Munich, Germany
| | - Alana Darcher
- Epileptology, University Hospital Bonn, Bonn, Germany
| | - Dominik Paquet
- Institute for Stroke and Dementia Research, University Hospital Munich, Ludwig-Maximilians- University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany
| | - Henrik Zetterberg
- Department of Psychiatry & Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, 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
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany.
- Biomedical Centre (BMC), Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
13
|
Zhang Y, Tian J, Ni J, Wei M, Li T, Shi J. Peripheral Blood and Cerebrospinal Fluid Levels of YKL-40 in Alzheimer's Disease: A Systematic Review and Meta-Analysis. Brain Sci 2023; 13:1364. [PMID: 37891733 PMCID: PMC10605482 DOI: 10.3390/brainsci13101364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
The pathogenesis associated with Alzheimer's disease (AD) is particularly complicated, and early diagnosis and course monitoring of the disease are not ideal based on the available core biomarkers. As a biomarker closely related to neuroinflammation, YKL-40 provides a potential scalable approach in AD, but its association remains controversial and inconclusive with AD. We conducted this study to assess the utility of YKL-40 levels in peripheral blood and cerebrospinal fluid (CSF) of AD patients and healthy controls (HCs) by meta-analysis. We systematically searched and screened relevant trials for comparing YKL-40 levels between AD patients and HCs in PubMed, Embase, Cochrane, and Web of Science, with a search deadline of 14 March 2023 for each database. A total of 17 eligible and relevant studies involving 1811 subjects, including 949 AD patients and 862 HCs, were included. The results showed that YKL-40 levels in the peripheral blood of AD patients and HCs did not possess significant differences. Subgroup analysis showed YKL-40 significantly differed in plasma (SMD = 0.527, 95%CI: [0.302, 0.752]; p = 0.000), but did not in serum. In the case of comparison with HCs, YKL-40 was significantly higher in CSF of AD patients (SMD = 0.893, 95%CI: [0.665, 1.121]; p = 0.000). Besides that, when we performed a combined analysis of total YKL-40 in both peripheral blood and CSF, overall YKL-40 concentrations were also significantly increased among AD patients (SMD = 0.608, 95%CI: [0.272, 0.943]; p = 0.000). YKL-40 provides support and rationale for the neuroinflammatory pathogenesis of AD. The significance of CSF levels of YKL-40 for early screening of AD is definite. Plasma levels of YKL-40 also appear to assist in discriminating AD patients from HCs, which facilitates early screening and monitoring of the natural course of AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Shi
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; (Y.Z.); (J.T.); (J.N.); (M.W.); (T.L.)
| |
Collapse
|
14
|
Loveland PM, Yu JJ, Churilov L, Yassi N, Watson R. Investigation of Inflammation in Lewy Body Dementia: A Systematic Scoping Review. Int J Mol Sci 2023; 24:12116. [PMID: 37569491 PMCID: PMC10418754 DOI: 10.3390/ijms241512116] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Inflammatory mechanisms are increasingly recognized as important contributors to the pathogenesis of neurodegenerative diseases, including Lewy body dementia (LBD). Our objectives were to, firstly, review inflammation investigation methods in LBD (dementia with Lewy bodies and Parkinson's disease dementia) and, secondly, identify alterations in inflammatory signals in LBD compared to people without neurodegenerative disease and other neurodegenerative diseases. A systematic scoping review was performed by searching major electronic databases (MEDLINE, Embase, Web of Science, and PSYCHInfo) to identify relevant human studies. Of the 2509 results screened, 80 studies were included. Thirty-six studies analyzed postmortem brain tissue, and 44 investigated living subjects with cerebrospinal fluid, blood, and/or brain imaging assessments. Largely cross-sectional data were available, although two longitudinal clinical studies investigated prodromal Lewy body disease. Investigations were focused on inflammatory immune cell activity (microglia, astrocytes, and lymphocytes) and inflammatory molecules (cytokines, etc.). Results of the included studies identified innate and adaptive immune system contributions to inflammation associated with Lewy body pathology and clinical disease features. Different signals in early and late-stage disease, with possible late immune senescence and dystrophic glial cell populations, were identified. The strength of these associations is limited by the varying methodologies, small study sizes, and cross-sectional nature of the data. Longitudinal studies investigating associations with clinical and other biomarker outcomes are needed to improve understanding of inflammatory activity over the course of LBD. This could identify markers of disease activity and support therapeutic development.
Collapse
Affiliation(s)
- Paula M. Loveland
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3000, Australia
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3000, Australia
| | - Jenny J. Yu
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3000, Australia
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3000, Australia
| | - Leonid Churilov
- Department of Neurology, Melbourne Brain Centre, The Royal Melbourne Hospital, University of Melbourne, Parkville 3000, Australia
- Melbourne Medical School, University of Melbourne, Parkville 3000, Australia
| | - Nawaf Yassi
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3000, Australia
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3000, Australia
- Department of Neurology, Melbourne Brain Centre, The Royal Melbourne Hospital, University of Melbourne, Parkville 3000, Australia
| | - Rosie Watson
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3000, Australia
- Department of Medicine, The Royal Melbourne Hospital, University of Melbourne, Parkville 3000, Australia
| |
Collapse
|
15
|
Doroszkiewicz J, Kulczyńska-Przybik A, Dulewicz M, Borawska R, Zajkowska M, Słowik A, Mroczko B. Potential Utility of Cerebrospinal Fluid Glycoprotein Nonmetastatic Melanoma Protein B as a Neuroinflammatory Diagnostic Biomarker in Mild Cognitive Impairment and Alzheimer's Disease. J Clin Med 2023; 12:4689. [PMID: 37510803 PMCID: PMC10380476 DOI: 10.3390/jcm12144689] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is a very common neurodegenerative disorder characterized by the gradual loss of neurons and extracellular amyloid-peptide buildup. There is compelling evidence that the disease process depends on neuroinflammatory alterations, such as the activation of astrocytes and microglia cells. A transmembrane glycoprotein known as glycoprotein nonmetastatic melanoma protein B (GPNMB) plays a neuroprotective role during the development of neurodegeneration. To the best of our knowledge, this is the first investigation discussing the potential clinical usefulness of this protein in the AD continuum, especially in the MCI (mild cognitive impairment) stage. A total of 71 patients with AD or MCI as well as controls were enrolled in this study. The concentrations of GPNMB, YKL-40, Aβ1-42 (amyloid beta 1-42), Tau, and pTau and the Aβ1-42/1-40 ratio in the CSF (cerebrospinal fluid) were tested using immunological methods. The concentrations of both GPNMB and YKL-40 in the cerebrospinal fluid were significantly higher in patients with AD and MCI compared to the controls. Moreover, both proteins were biochemically associated with classical biomarkers of AD and were especially associated with the Aβ1-42/1-40 ratio and Tau and pTau levels in the whole study group. Elevated concentrations of GPNMB were observed in the Aβ(+) group of AD patients compared to the Aβ(-) subjects. Additionally, the diagnostic performance (AUC value) of GPNMB was higher than that of amyloid β1-42 in MCI patients compared with controls. Our study indicates that GPNMB might be a promising neuroinflammatory biomarker for the early diagnosis and prognosis of the AD continuum, with potential utility as a therapeutic target.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | | | - Maciej Dulewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Renata Borawska
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Monika Zajkowska
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Agnieszka Słowik
- Department of Neurology, Jagiellonian University, 30-688 Cracow, Poland
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
16
|
Pesämaa I, Müller SA, Robinson S, Darcher A, Paquet D, Zetterberg H, Lichtenthaler SF, Haass C. A MICROGLIAL ACTIVITY STATE BIOMARKER PANEL DIFFERENTIATES FTD-GRANULIN AND ALZHEIMER'S DISEASE PATIENTS FROM CONTROLS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545187. [PMID: 37398209 PMCID: PMC10312678 DOI: 10.1101/2023.06.15.545187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Background With the emergence of microglia-modulating therapies there is an urgent need for reliable biomarkers to evaluate microglial activation states. Methods Using mouse models and human induced pluripotent stem cell-derived microglia (hiMGL), which were genetically modified to yield the most opposite homeostatic ( TREM2- knockout) and disease-associated ( GRN -knockout) states, we identified microglia activity-dependent markers. Non-targeted mass spectrometry was used to identify changes in microglial and cerebrospinal (CSF) proteome of Grn - and Trem2 -knockout mice. Additionally, we analyzed the proteome of GRN - and TREM2 -knockout hiMGL and their conditioned media. Candidate marker proteins were tested in two independent patient cohorts, the ALLFTD cohort with 11 GRN mutation carriers and 12 non-carriers, as well as the proteomic data set available from the European Medical Information Framework Alzheimer's Disease Multimodal Biomarker Discovery (EMIF-AD MBD). Findings We identified proteomic changes between the opposite activation states in mouse microglia and cerebrospinal fluid (CSF), as well as in hiMGL cell lysates and conditioned media. For further verification, we analyzed the CSF proteome of heterozygous GRN mutation carriers suffering from frontotemporal dementia (FTD). We identified a panel of six proteins (FABP3, MDH1, GDI1, CAPG, CD44, GPNMB) as potential indicators for microglial activation. Moreover, we confirmed three of these proteins (FABP3, GDI1, MDH1) to be significantly elevated in the CSF of AD patients. In AD, these markers differentiated amyloid-positive cases with mild cognitive impairment (MCI) from amyloid-negative individuals. Interpretation The identified candidate proteins reflect microglia activity and may be relevant for monitoring the microglial response in clinical practice and clinical trials modulating microglial activity and amyloid deposition. Moreover, the finding that three of these markers differentiate amyloid-positive from amyloid-negative MCI cases in the AD cohort suggests that these marker proteins associate with a very early immune response to seeded amyloid. This is consistent with our previous findings in the DIAN (Dominantly Inherited Alzheimer's Disease Network) cohort, where soluble TREM2 increases as early as 21 years before symptom onset. Moreover, in mouse models for amyloidogenesis, seeding of amyloid is limited by physiologically active microglia further supporting their early protective role. The biological functions of some of our main candidates (FABP3, CD44, GPNMB) also further emphasize that lipid dysmetabolism may be a common feature of neurodegenerative disorders. Funding This work was supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany's Excellence Strategy within the framework of the Munich Cluster for Systems Neurology (EXC 2145 SyNergy - ID 390857198 to CH, SFL and DP) and a Koselleck Project HA1737/16-1 (to CH).
Collapse
|
17
|
Russo C, Valle MS, Casabona A, Malaguarnera L. Chitinase Signature in the Plasticity of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24076301. [PMID: 37047273 PMCID: PMC10094409 DOI: 10.3390/ijms24076301] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Several reports have pointed out that Chitinases are expressed and secreted by various cell types of central nervous system (CNS), including activated microglia and astrocytes. These cells play a key role in neuroinflammation and in the pathogenesis of many neurodegenerative disorders. Increased levels of Chitinases, in particular Chitotriosidase (CHIT-1) and chitinase-3-like protein 1 (CHI3L1), have been found increased in several neurodegenerative disorders. Although having important biological roles in inflammation, to date, the molecular mechanisms of Chitinase involvement in the pathogenesis of neurodegenerative disorders is not well-elucidated. Several studies showed that some Chitinases could be assumed as markers for diagnosis, prognosis, activity, and severity of a disease and therefore can be helpful in the choice of treatment. However, some studies showed controversial results. This review will discuss the potential of Chitinases in the pathogenesis of some neurodegenerative disorders, such as Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, and multiple sclerosis, to understand their role as distinctive biomarkers of neuronal cell activity during neuroinflammatory processes. Knowledge of the role of Chitinases in neuronal cell activation could allow for the development of new methodologies for downregulating neuroinflammation and consequently for diminishing negative neurological disease outcomes.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Maria Stella Valle
- Laboratory of Neuro-Biomechanics, Section of Physiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
- Correspondence:
| | - Antonino Casabona
- Laboratory of Neuro-Biomechanics, Section of Physiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
18
|
Bernhardt AM, Tiedt S, Teupser D, Dichgans M, Meyer B, Gempt J, Kuhn PH, Simons M, Palleis C, Weidinger E, Nübling G, Holdt L, Hönikl L, Gasperi C, Giesbertz P, Müller SA, Breimann S, Lichtenthaler SF, Kuster B, Mann M, Imhof A, Barth T, Hauck SM, Zetterberg H, Otto M, Weichert W, Hemmer B, Levin J. A unified classification approach rating clinical utility of protein biomarkers across neurologic diseases. EBioMedicine 2023; 89:104456. [PMID: 36745974 PMCID: PMC9931915 DOI: 10.1016/j.ebiom.2023.104456] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/22/2022] [Accepted: 01/17/2023] [Indexed: 02/07/2023] Open
Abstract
A major evolution from purely clinical diagnoses to biomarker supported clinical diagnosing has been occurring over the past years in neurology. High-throughput methods, such as next-generation sequencing and mass spectrometry-based proteomics along with improved neuroimaging methods, are accelerating this development. This calls for a consensus framework that is broadly applicable and provides a spot-on overview of the clinical validity of novel biomarkers. We propose a harmonized terminology and a uniform concept that stratifies biomarkers according to clinical context of use and evidence levels, adapted from existing frameworks in oncology with a strong focus on (epi)genetic markers and treatment context. We demonstrate that this framework allows for a consistent assessment of clinical validity across disease entities and that sufficient evidence for many clinical applications of protein biomarkers is lacking. Our framework may help to identify promising biomarker candidates and classify their applications by clinical context, aiming for routine clinical use of (protein) biomarkers in neurology.
Collapse
Affiliation(s)
- Alexander M Bernhardt
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany; German Center for Neurodegenerative Diseases, Site Munich, Germany
| | - Steffen Tiedt
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Martin Dichgans
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Peer-Hendrik Kuhn
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases, Site Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Institute of Neuronal Cell Biology, Technical University Munich, 80802, Munich, Germany
| | - Carla Palleis
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany; German Center for Neurodegenerative Diseases, Site Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Endy Weidinger
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany; German Center for Neurodegenerative Diseases, Site Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Georg Nübling
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany; German Center for Neurodegenerative Diseases, Site Munich, Germany
| | - Lesca Holdt
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Lisa Hönikl
- Department of Neurosurgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Christiane Gasperi
- Department of Neurology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Pieter Giesbertz
- German Center for Neurodegenerative Diseases, Site Munich, Germany; Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases, Site Munich, Germany; Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stephan Breimann
- German Center for Neurodegenerative Diseases, Site Munich, Germany; Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany; Department of Bioinformatics, Wissenschaftszentrum Weihenstephan, Technical University of Munich, Freising, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases, Site Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany; German Cancer Consortium (DKTK), Munich Partner Site, Munich, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Axel Imhof
- Protein Analysis Unit, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Großhaderner Straße 9, 82152, Martinsried, Germany
| | - Teresa Barth
- Protein Analysis Unit, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Großhaderner Straße 9, 82152, Martinsried, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Centre Munich, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Markus Otto
- Department of Neurology, Halle University Hospital, Martin Luther University Halle/Wittenberg, Saale, Germany
| | - Wilko Weichert
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Bernhard Hemmer
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Neurology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany; German Center for Neurodegenerative Diseases, Site Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
19
|
Advanced Overview of Biomarkers and Techniques for Early Diagnosis of Alzheimer's Disease. Cell Mol Neurobiol 2023:10.1007/s10571-023-01330-y. [PMID: 36847930 DOI: 10.1007/s10571-023-01330-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023]
Abstract
The development of early non-invasive diagnosis methods and identification of novel biomarkers are necessary for managing Alzheimer's disease (AD) and facilitating effective prognosis and treatment. AD has multi-factorial nature and involves complex molecular mechanism, which causes neuronal degeneration. The primary challenges in early AD detection include patient heterogeneity and lack of precise diagnosis at the preclinical stage. Several cerebrospinal fluid (CSF) and blood biomarkers have been proposed to show excellent diagnosis ability by identifying tau pathology and cerebral amyloid beta (Aβ) for AD. Intense research endeavors are being made to develop ultrasensitive detection techniques and find potent biomarkers for early AD diagnosis. To mitigate AD worldwide, understanding various CSF biomarkers, blood biomarkers, and techniques that can be used for early diagnosis is imperative. This review attempts to provide information regarding AD pathophysiology, genetic and non-genetic factors associated with AD, several potential blood and CSF biomarkers, like neurofilament light, neurogranin, Aβ, and tau, along with biomarkers under development for AD detection. Besides, numerous techniques, such as neuroimaging, spectroscopic techniques, biosensors, and neuroproteomics, which are being explored to aid early AD detection, have been discussed. The insights thus gained would help in finding potential biomarkers and suitable techniques for the accurate diagnosis of early AD before cognitive dysfunction.
Collapse
|
20
|
TDP-43 Proteinopathy Specific Biomarker Development. Cells 2023; 12:cells12040597. [PMID: 36831264 PMCID: PMC9954136 DOI: 10.3390/cells12040597] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
TDP-43 is the primary or secondary pathological hallmark of neurodegenerative diseases, such as amyotrophic lateral sclerosis, half of frontotemporal dementia cases, and limbic age-related TDP-43 encephalopathy, which clinically resembles Alzheimer's dementia. In such diseases, a biomarker that can detect TDP-43 proteinopathy in life would help to stratify patients according to their definite diagnosis of pathology, rather than in clinical subgroups of uncertain pathology. For therapies developed to target pathological proteins that cause the disease a biomarker to detect and track the underlying pathology would greatly enhance such undertakings. This article reviews the latest developments and outlooks of deriving TDP-43-specific biomarkers from the pathophysiological processes involved in the development of TDP-43 proteinopathy and studies using biosamples from clinical entities associated with TDP-43 pathology to investigate biomarker candidates.
Collapse
|
21
|
Lin H, Zhang J, Dai Y, Liu H, He X, Chen L, Tao J, Li C, Liu W. Neurogranin as an important regulator in swimming training to improve the spatial memory dysfunction of mice with chronic cerebral hypoperfusion. JOURNAL OF SPORT AND HEALTH SCIENCE 2023; 12:116-129. [PMID: 35066217 PMCID: PMC9923430 DOI: 10.1016/j.jshs.2022.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/18/2021] [Accepted: 12/30/2021] [Indexed: 05/09/2023]
Abstract
BACKGROUND Vascular cognitive impairment caused by chronic cerebral hypoperfusion (CCH) has become a hot issue worldwide. Aerobic exercise positively contributes to the preservation or restoration of cognitive abilities; however, the specific mechanism has remained inconclusive. And recent studies found that neurogranin (Ng) is a potential biomarker for cognitive impairment. This study aims to investigate the underlying role of Ng in swimming training to improve cognitive impairment. METHODS To test this hypothesis, the clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) system was utilized to construct a strain of Ng conditional knockout (Ng cKO) mice, and bilateral common carotid artery stenosis (BCAS) surgery was performed to prepare the model. In Experiment 1, 2-month-old male and female transgenic mice were divided into a control group (wild-type littermate, n = 9) and a Ng cKO group (n = 9). Then, 2-month-old male and female C57BL/6 mice were divided into a sham group (C57BL/6, n = 12) and a BCAS group (n = 12). In Experiment 2, 2-month-old male and female mice were divided into a sham group (wild-type littermate, n = 12), BCAS group (n = 12), swim group (n = 12), BCAS + Ng cKO group (n = 12), and swim + Ng cKO group (n = 12). Then, 7 days after BCAS, mice were given swimming training for 5 weeks (1 week for adaptation and 4 weeks for training, 5 days a week, 60 min a day). After intervention, laser speckle was used to detect cerebral blood perfusion in the mice, and the T maze and Morris water maze were adopted to test their spatial memory. Furthermore, electrophysiology and Western blotting were conducted to record long-term potential and observe the expressions of Ca2+ pathway-related proteins, respectively. Immunohistochemistry was applied to analyze the expression of relevant markers in neuronal damage, inflammation, and white matter injury. RESULTS The figures showed that spatial memory impairment was detected in Ng cKO mice, and a sharp decline of cerebral blood flow and an impairment of progressive spatial memory were observed in BCAS mice. Regular swimming training improved the spatial memory impairment of BCAS mice. This was achieved by preventing long-term potential damage and reversing the decline of Ca2+ signal transduction pathway-related proteins. At the same time, the results suggested that swimming also led to improvements in neuronal death, inflammation, and white matter injury induced by CCH. Further study adopted the use of Ng cKO transgenic mice, and the results indicated that the positive effects of swimming training on cognitive impairments, synaptic plasticity, and related pathological changes caused by CCH could be abolished by the knockout of Ng. CONCLUSION Swimming training can mediate the expression of Ng to enhance hippocampal synaptic plasticity and improve related pathological changes induced by CCH, thereby ameliorating the spatial memory impairment of vascular cognitive impairment.
Collapse
Affiliation(s)
- Huawei Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Jiayong Zhang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Yaling Dai
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Huanhuan Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Xiaojun He
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Lewen Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Jing Tao
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Chaohui Li
- General surgery, Anxi General Hospital of Traditional Chinese Medicine, Quanzhou 362400, China
| | - Weilin Liu
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| |
Collapse
|
22
|
Shir D, Mielke MM, Hofrenning EI, Lesnick TG, Knopman DS, Petersen RC, Jack CR, Algeciras-Schimnich A, Vemuri P, Graff-Radford J. Associations of Neurodegeneration Biomarkers in Cerebrospinal Fluid with Markers of Alzheimer's Disease and Vascular Pathology. J Alzheimers Dis 2023; 92:887-898. [PMID: 36806507 PMCID: PMC10193844 DOI: 10.3233/jad-221015] [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] [Indexed: 02/19/2023]
Abstract
BACKGROUND The National Institute on Aging-Alzheimer's Association Research Framework proposes defining Alzheimer's disease by grouping imaging and fluid biomarkers by their respective pathologic processes. The AT(N) structure proposes several neurodegenerative fluid biomarkers (N) including total tau (t-tau), neurogranin (Ng), and neurofilament light chain (NfL). However, pathologic drivers influencing each biomarker remain unclear. OBJECTIVE To determine whether cerebrospinal fluid (CSF)-neurodegenerative biomarkers (N) map differentially to Alzheimer's disease pathology measured by Aβ42 (an indicator of amyloidosis, [A]), p-tau (an indicator of tau deposition, [T]), and MRI vascular pathology indicators (measured by white-matter integrity, infarcts, and microbleeds [V]). METHODS Participants were from Mayo Clinic Study of Aging (MCSA) with CSF measures of NfL, Ng, t-tau, Aβ42, and p-tau and available MRI brain imaging. Linear models assessed associations between CSF neurodegeneration (N) markers, amyloid markers (A), tau (T), and vascular pathology (V). RESULTS Participants (n = 408) had a mean age of 69.2±10.7; male, 217 (53.2%); cognitively unimpaired, 359 (88%). All three neurodegeneration biomarkers correlated with age (p < 0.001 for NfL and t-tau, p = 0.018 for Ng). Men had higher CSF-NfL levels; women had higher Ng (p < 0.001). NfL and t-tau levels correlated with infarcts (p = 0.009, p = 0.034 respectively); no biomarkers correlated with white-matter integrity. N biomarkers correlated with p-tau levels (T, p < 0.001). Higher Aβ42 levels associated with higher N-biomarker levels but only among cognitively unimpaired (A, p < 0.001). CONCLUSION The influence of vascular pathology in the general population on CSF (N) biomarkers is modest, with greater influence of infarcts than white-matter disruption. Neurodegeneration markers more closely correlated with tau than amyloid markers.
Collapse
Affiliation(s)
- Dror Shir
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Michelle M. Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota 55905, USA
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina, 27101
| | | | - Timothy G. Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - David S. Knopman
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Ronald C. Petersen
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55905, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Clifford R. Jack
- Department of Radiology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | |
Collapse
|
23
|
Wesenhagen KEJ, Tijms BM, Boonkamp L, Hoede PL, Goossens J, Dewit N, Scheltens P, Vanmechelen E, Visser PJ, Teunissen CE. P-tau subgroups in AD relate to distinct amyloid production and synaptic integrity profiles. Alzheimers Res Ther 2022; 14:95. [PMID: 35841015 PMCID: PMC9288016 DOI: 10.1186/s13195-022-01038-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022]
Abstract
Background We previously identified four Alzheimer’s disease (AD) subgroups with increasingly higher cerebrospinal fluid (CSF) levels of tau phosphorylated at threonine 181 (p-tau). These subgroups included individuals across the cognitive spectrum, suggesting p-tau subgroups could reflect distinct biological changes in AD, rather than disease severity. Therefore, in the current study, we further investigated which potential processes may be related with p-tau subgroups, by comparing individuals on CSF markers for presynaptic structure [vesicle-associated membrane protein 2 (VAMP2)], postsynaptic structure [neurogranin (NRGN)], axonal damage [neurofilament light (NfL)], and amyloid production [beta-secretase 1 (BACE1) and amyloid-beta 1–40 (Aβ40)]. Methods We selected 348 amyloid-positive (A+) individuals (53 preclinical, 102 prodromal, 193 AD dementia) and 112 amyloid-negative (A−) cognitively normal (CN) individuals from the Amsterdam Dementia Cohort (ADC). Individuals were labeled according to their p-tau subgroup (subgroup 1: p-tau ≤ 56 pg/ml; subgroup 2: 57–96 pg/ml; subgroup 3: 97–159 pg/ml; subgroup 4: > 159 pg/ml). CSF protein levels were measured with ELISA (NRGN, BACE1, Aβ40, NfL) or single-molecule array (Simoa) (VAMP2). We tested whether protein levels differed between the p-tau subgroups within A+ individuals with linear models corrected for age and sex and whether disease stage influenced these relationships. Results Among A+ individuals, higher p-tau subgroups showed a higher percentage of AD dementia [subgroup 1: n = 41/94 (44%); subgroup 2: n = 81/147 (55%); subgroup 3: n = 59/89 (66%); subgroup 4: n = 7/11 (64%)]. Relative to controls, subgroup 1 showed reduced CSF levels of BACE1, Aβ40, and VAMP2 and higher levels of NfL. Subgroups 2 to 4 showed gradually increased CSF levels of all measured proteins, either across the first three (NfL and Aβ40) or across all subgroups (VAMP2, NRGN, BACE1). The associations did not depend on the clinical stage (interaction p-values ranging between 0.19 and 0.87). Conclusions The results suggest that biological heterogeneity in p-tau levels in AD is related to amyloid metabolism and synaptic integrity independent of clinical stage. Biomarkers reflecting amyloid metabolism and synaptic integrity may be useful outcome measures in clinical trials targeting tau pathology.
Collapse
|
24
|
Abstract
Alzheimer's disease (AD) characterization has progressed from being indexed using clinical symptomatology followed by neuropathological examination at autopsy to in vivo signatures using cerebrospinal fluid (CSF) biomarkers and positron emission tomography. The core AD biomarkers reflect amyloid-β plaques (A), tau pathology (T) and neurodegeneration (N), following the ATN schedule, and are now being introduced into clinical routine practice. This is an important development, as disease-modifying treatments are now emerging. Further, there are now reproducible data on CSF biomarkers which reflect synaptic pathology, neuroinflammation and common co-pathologies. In addition, the development of ultrasensitive techniques has enabled the core CSF biomarkers of AD pathophysiology to be translated to blood (e.g., phosphorylated tau, amyloid-β and neurofilament light). In this chapter, we review where we stand with both core and novel CSF biomarkers, as well as the explosion of data on blood biomarkers. Also, we discuss potential applications in research aiming to better understand the disease, as well as possible use in routine clinical practice and therapeutic trials.
Collapse
Affiliation(s)
- Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Anders Elmgren
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, United Kingdom; UK Dementia Research Institute, University College London, London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| |
Collapse
|
25
|
Liu WL, Lin HW, Lin MR, Yu Y, Liu HH, Dai YL, Chen LW, Jia WW, He XJ, Li XL, Zhu JF, Xue XH, Tao J, Chen LD. Emerging blood exosome-based biomarkers for preclinical and clinical Alzheimer's disease: a meta-analysis and systematic review. Neural Regen Res 2022; 17:2381-2390. [PMID: 35535875 PMCID: PMC9120706 DOI: 10.4103/1673-5374.335832] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/29/2021] [Accepted: 10/10/2021] [Indexed: 11/10/2022] Open
Abstract
Blood exosomes, which are extracellular vesicles secreted by living cells into the circulating blood, are regarded as a relatively noninvasive novel tool for monitoring brain physiology and disease states. An increasing number of blood cargo-loaded exosomes are emerging as potential biomarkers for preclinical and clinical Alzheimer's disease. Therefore, we conducted a meta-analysis and systematic review of molecular biomarkers derived from blood exosomes to comprehensively analyze their diagnostic performance in preclinical Alzheimer's disease, mild cognitive impairment, and Alzheimer's disease. We performed a literature search in PubMed, Web of Science, Embase, and Cochrane Library from their inception to August 15, 2020. The research subjects mainly included Alzheimer's disease, mild cognitive impairment, and preclinical Alzheimer's disease. We identified 34 observational studies, of which 15 were included in the quantitative analysis (Newcastle-Ottawa Scale score 5.87 points) and 19 were used in the qualitative analysis. The meta-analysis results showed that core biomarkers including Aβ1-42, P-T181-tau, P-S396-tau, and T-tau were increased in blood neuron-derived exosomes of preclinical Alzheimer's disease, mild cognitive impairment, and Alzheimer's disease patients. Molecules related to additional risk factors that are involved in neuroinflammation (C1q), metabolism disorder (P-S312-IRS-1), neurotrophic deficiency (HGF), vascular injury (VEGF-D), and autophagy-lysosomal system dysfunction (cathepsin D) were also increased. At the gene level, the differential expression of transcription-related factors (REST) and microRNAs (miR-132) also affects RNA splicing, transport, and translation. These pathological changes contribute to neural loss and synaptic dysfunction. The data confirm that the above-mentioned core molecules and additional risk-related factors in blood exosomes can serve as candidate biomarkers for preclinical and clinical Alzheimer's disease. These findings support further development of exosome biomarkers for a clinical blood test for Alzheimer's disease. This meta-analysis was registered at the International Prospective Register of Systematic Reviews (Registration No. CRD4200173498, 28/04/2020).
Collapse
Affiliation(s)
- Wei-Lin Liu
- Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Hua-Wei Lin
- Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Miao-Ran Lin
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Yan Yu
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Huan-Huan Liu
- Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Ya-Ling Dai
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Le-Wen Chen
- Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Wei-Wei Jia
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Xiao-Jun He
- Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Xiao-Ling Li
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Jing-Fang Zhu
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Xie-Hua Xue
- Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Jing Tao
- Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Li-Dian Chen
- Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| |
Collapse
|
26
|
Piccoli T, Blandino V, Maniscalco L, Matranga D, Graziano F, Guajana F, Agnello L, Lo Sasso B, Gambino CM, Giglio RV, La Bella V, Ciaccio M, Colletti T. Biomarkers Related to Synaptic Dysfunction to Discriminate Alzheimer's Disease from Other Neurological Disorders. Int J Mol Sci 2022; 23:10831. [PMID: 36142742 PMCID: PMC9501545 DOI: 10.3390/ijms231810831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/07/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Recently, the synaptic proteins neurogranin (Ng) and α-synuclein (α-Syn) have attracted scientific interest as potential biomarkers for synaptic dysfunction in neurodegenerative diseases. In this study, we measured the CSF Ng and α-Syn concentrations in patients affected by AD (n = 69), non-AD neurodegenerative disorders (n-AD = 50) and non-degenerative disorders (n-ND, n = 98). The concentrations of CSF Ng and α-Syn were significantly higher in AD than in n-AD and n-ND. Moreover, the Aβ42/Ng and Aβ42/α-Syn ratios showed statistically significant differences between groups and discriminated AD patients from n-AD patients, better than Ng or α-Syn alone. Regression analyses showed an association of higher Ng concentrations with MMSE < 24, pathological Aβ 42/40 ratios, pTau, tTau and the ApoEε4 genotype. Aβ 42/Ng was associated with MMSE < 24, an AD-related FDG-PET pattern, the ApoEε4 genotype, pathological Aβ 42 levels and Aβ 42/40 ratios, pTau, and tTau. Moreover, APO-Eε4 carriers showed higher Ng concentrations than non-carriers. Our results support the idea that the Aβ 42/Ng ratio is a reliable index of synaptic dysfunction/degeneration able to discriminate AD from other neurological conditions.
Collapse
Affiliation(s)
- Tommaso Piccoli
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| | - Valeria Blandino
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| | - Laura Maniscalco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Domenica Matranga
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Fabiola Graziano
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| | - Fabrizio Guajana
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| | - Luisa Agnello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy
| | - Bruna Lo Sasso
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy
| | - Caterina Maria Gambino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy
| | - Rosaria Vincenza Giglio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy
| | - Vincenzo La Bella
- ALS Clinical Research Center and Laboratory of Neurochemistry, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| | - Marcello Ciaccio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy
| | - Tiziana Colletti
- ALS Clinical Research Center and Laboratory of Neurochemistry, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| |
Collapse
|
27
|
Mumtaz I, Ayaz MO, Khan MS, Manzoor U, Ganayee MA, Bhat AQ, Dar GH, Alghamdi BS, Hashem AM, Dar MJ, Ashraf GM, Maqbool T. Clinical relevance of biomarkers, new therapeutic approaches, and role of post-translational modifications in the pathogenesis of Alzheimer's disease. Front Aging Neurosci 2022; 14:977411. [PMID: 36158539 PMCID: PMC9490081 DOI: 10.3389/fnagi.2022.977411] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/18/2022] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that causes progressive loss of cognitive functions like thinking, memory, reasoning, behavioral abilities, and social skills thus affecting the ability of a person to perform normal daily functions independently. There is no definitive cure for this disease, and treatment options available for the management of the disease are not very effective as well. Based on histopathology, AD is characterized by the accumulation of insoluble deposits of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs). Although several molecular events contribute to the formation of these insoluble deposits, the aberrant post-translational modifications (PTMs) of AD-related proteins (like APP, Aβ, tau, and BACE1) are also known to be involved in the onset and progression of this disease. However, early diagnosis of the disease as well as the development of effective therapeutic approaches is impeded by lack of proper clinical biomarkers. In this review, we summarized the current status and clinical relevance of biomarkers from cerebrospinal fluid (CSF), blood and extracellular vesicles involved in onset and progression of AD. Moreover, we highlight the effects of several PTMs on the AD-related proteins, and provide an insight how these modifications impact the structure and function of proteins leading to AD pathology. Finally, for disease-modifying therapeutics, novel approaches, and targets are discussed for the successful treatment and management of AD.
Collapse
Affiliation(s)
- Ibtisam Mumtaz
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| | - Mir Owais Ayaz
- Laboratory of Cell and Molecular Biology, Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Centre for Scientific and Innovative Research, Ghaziabad, Utter Pradesh, India
| | - Mohamad Sultan Khan
- Neurobiology and Molecular Chronobiology Laboratory, Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Umar Manzoor
- Laboratory of Immune and Inflammatory Disease, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, South Korea
| | - Mohd Azhardin Ganayee
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, India
| | - Aadil Qadir Bhat
- Laboratory of Cell and Molecular Biology, Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Centre for Scientific and Innovative Research, Ghaziabad, Utter Pradesh, India
| | - Ghulam Hassan Dar
- Sri Pratap College, Cluster University Srinagar, Jammu and Kashmir, India
| | - Badrah S. Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pre-clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anwar M. Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohd Jamal Dar
- Laboratory of Cell and Molecular Biology, Department of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Centre for Scientific and Innovative Research, Ghaziabad, Utter Pradesh, India
| | - Gulam Md. Ashraf
- Pre-clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tariq Maqbool
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| |
Collapse
|
28
|
Zakharova NV, Bugrova AE, Indeykina MI, Fedorova YB, Kolykhalov IV, Gavrilova SI, Nikolaev EN, Kononikhin AS. Proteomic Markers and Early Prediction of Alzheimer's Disease. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:762-776. [PMID: 36171657 DOI: 10.1134/s0006297922080089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD) is the most common socially significant neurodegenerative pathology, which currently affects more than 30 million elderly people worldwide. Since the number of patients grows every year and may exceed 115 million by 2050, and due to the lack of effective therapies, early prediction of AD remains a global challenge, solution of which can contribute to the timely appointment of a preventive therapy in order to avoid irreversible changes in the brain. To date, clinical assays for the markers of amyloidosis in cerebrospinal fluid (CSF) have been developed, which, in conjunction with the brain MRI and PET studies, are used either to confirm the diagnosis based on obligate clinical criteria or to predict the risk of AD developing at the stage of mild cognitive impairment (MCI). However, the problem of predicting AD at the asymptomatic stage remains unresolved. In this regard, the search for new protein markers and studies of proteomic changes in CSF and blood plasma are of particular interest and may consequentially identify particular pathways involved in the pathogenesis of AD. Studies of specific proteomic changes in blood plasma deserve special attention and are of increasing interest due to the much less invasive method of sample collection as compared to CSF, which is important when choosing the object for large-scale screening. This review briefly summarizes the current knowledge on proteomic markers of AD and considers the prospects of developing reliable methods for early identification of AD risk factors based on the proteomic profile.
Collapse
Affiliation(s)
- Natalia V Zakharova
- Emanuel Institute for Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Anna E Bugrova
- Emanuel Institute for Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Maria I Indeykina
- Emanuel Institute for Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | | | | | | | - Evgeny N Nikolaev
- Skolkovo Institute of Science and Technology, Moscow, 121205, Russia
| | | |
Collapse
|
29
|
Zhou J, Benoit M, Sharoar MG. Recent advances in pre-clinical diagnosis of Alzheimer's disease. Metab Brain Dis 2022; 37:1703-1725. [PMID: 33900524 DOI: 10.1007/s11011-021-00733-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/05/2021] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is the most common dementia with currently no known cures or disease modifying treatments (DMTs), despite much time and effort from the field. Diagnosis and intervention of AD during the early pre-symptomatic phase of the disease is thought to be a more effective strategy. Therefore, the detection of biomarkers has emerged as a critical tool for monitoring the effect of new AD therapies, as well as identifying patients most likely to respond to treatment. The establishment of the amyloid/tau/neurodegeneration (A/T/N) framework in 2018 has codified the contexts of use of AD biomarkers in neuroimaging and bodily fluids for research and diagnostic purposes. Furthermore, a renewed drive for novel AD biomarkers and innovative methods of detection has emerged with the goals of adding additional insight to disease progression and discovery of new therapeutic targets. The use of biomarkers has accelerated the development of AD drugs and will bring new therapies to patients in need. This review highlights recent methods utilized to diagnose antemortem AD.
Collapse
Affiliation(s)
- John Zhou
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
- Molecular Medicine Program, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Marc Benoit
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Md Golam Sharoar
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA.
| |
Collapse
|
30
|
Hok-A-Hin YS, Hoozemans JJM, Hu WT, Wouters D, Howell JC, Rábano A, van der Flier WM, Pijnenburg YAL, Teunissen CE, Del Campo M. YKL-40 changes are not detected in post-mortem brain of patients with Alzheimer's disease and frontotemporal lobar degeneration. Alzheimers Res Ther 2022; 14:100. [PMID: 35879733 PMCID: PMC9310415 DOI: 10.1186/s13195-022-01039-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/22/2022] [Indexed: 12/12/2022]
Abstract
Background YKL-40 (Chitinase 3-like I) is increased in CSF of Alzheimer’s disease (AD) and frontotemporal lobar degeneration (FTLD) patients and is therefore considered a potential neuroinflammatory biomarker. Whether changed YKL-40 levels in the CSF reflect dysregulation of YKL-40 in the brain is not completely understood yet. We aimed to extensively analyze YKL-40 levels in the brain of AD and different FTLD pathological subtypes. The direct relationship between YKL-40 levels in post-mortem brain and ante-mortem CSF was examined in a small set of paired brain-CSF samples. Method YKL-40 was analyzed in post-mortem temporal and frontal cortex of non-demented controls and patients with AD and FTLD (including FTLD-Tau and FTLD-TDP) pathology by immunohistochemistry (temporal cortex: 51 controls and 56 AD and frontal cortex: 7 controls and 24 FTLD patients), western blot (frontal cortex: 14 controls, 5 AD and 67 FTLD patients), or ELISA (temporal cortex: 11 controls and 7 AD and frontal cortex: 14 controls, 5 AD and 67 FTLD patients). YKL-40 levels were also measured in paired post-mortem brain and ante-mortem CSF samples from dementia patients (n = 9, time-interval collection: 1.4 years) by ELISA. Results We observed that YKL-40 post-mortem brain levels were similar between AD, FTLD, and controls as shown by immunohistochemistry, western blot, and ELISA. Interestingly, strong YKL-40 immunoreactivity was observed in AD cases with cerebral amyloid angiopathy (CAA; n = 6). In paired CSF-brain samples, YKL-40 concentration was 8-times higher in CSF compared to brain. Conclusion Our data suggest that CSF YKL-40 changes may not reflect YKL-40 changes within AD and FTLD pathological brain areas. The YKL-40 reactivity associated with classical CAA hallmarks indicates a possible relationship between YKL-40, neuroinflammation, and vascular pathology. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-01039-y.
Collapse
Affiliation(s)
- Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Clinical Chemistry department, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Jeroen J M Hoozemans
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Amsterdam, The Netherlands
| | - William T Hu
- Department of Neurology, Center for Neurodegenerative Diseases Research, Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, USA
| | - Dorine Wouters
- Neurochemistry Laboratory, Clinical Chemistry department, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Amsterdam, The Netherlands
| | - Jennifer C Howell
- Department of Neurology, Center for Neurodegenerative Diseases Research, Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, USA
| | - Alberto Rábano
- CIEN Tissue Bank, Alzheimer's Centre Reina Sofía-CIEN Foundation, Madrid, Spain
| | - Wiesje M van der Flier
- Alzheimer Centre Amsterdam, Department of Neurology, Amsterdam Neuroscience, VU University Medical Centers, Amsterdam, The Netherlands.,Department of Epidemiology and Data Science, VU University Medical Centers, Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Centre Amsterdam, Department of Neurology, Amsterdam Neuroscience, VU University Medical Centers, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Clinical Chemistry department, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Amsterdam, The Netherlands
| | - Marta Del Campo
- Neurochemistry Laboratory, Clinical Chemistry department, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Amsterdam, The Netherlands.,Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
31
|
Hawksworth J, Fernández E, Gevaert K. A new generation of AD biomarkers: 2019 to 2021. Ageing Res Rev 2022; 79:101654. [PMID: 35636691 DOI: 10.1016/j.arr.2022.101654] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 05/17/2022] [Accepted: 05/25/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and cases are rising worldwide. The effort to fight this disease is hampered by a lack of disease-modifying treatments and the absence of an early, accurate diagnostic tool. Neuropathology begins years or decades before symptoms occur and, upon onset of symptoms, diagnosis can take a year or more. Such delays postpone treatment and make research into the early stages of the disease difficult. Ideally, clinicians require a minimally invasive test that can detect AD in its early stages, before cognitive symptoms occur. Advances in proteomic technologies have facilitated the study of promising biomarkers of AD. Over the last two years (2019-2021) studies have identified and validated many species which can be measured in cerebrospinal fluid (CSF), plasma, or in both fluids, and which have a high predictive value for AD. We herein discuss proteins which have been highlighted as promising biomarkers of AD in the last two years, and consider implications for future research within the research framework of the amyloid (A), tau (T), neurodegeneration (N) scoring system. We review recently identified species of amyloid and tau which may improve diagnosis when used in combination with current measures such as amyloid-beta-42 (Aβ42), total tau (t-tau) and phosphorylated tau (p-tau). In addition, several proteins have been identified as likely proxies for neurodegeneration, including neurofilament light (NfL), synaptosomal-associated protein 25 (SNAP-25) and neurogranin (NRGN). Finally, proteins originating from diverse processes such as neuroinflammation, lipid transport and mitochondrial dysfunction could aid in both AD diagnosis and patient stratification.
Collapse
|
32
|
Blanc F, Bousiges O. Biomarkers and diagnosis of dementia with Lewy bodies including prodromal: Practical aspects. Rev Neurol (Paris) 2022; 178:472-483. [PMID: 35491246 DOI: 10.1016/j.neurol.2022.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 10/18/2022]
Abstract
Dementia with Lewy Bodies (DLB) is a common form of cognitive neurodegenerative disease. More than half of the patients affected are not or misdiagnosed because of the clinical similarity with Alzheimer's disease (AD), Parkinson's disease but also psychiatric diseases such as depression or psychosis. In this review, we evaluate the interest of different biomarkers in the diagnostic process: cerebrospinal fluid (CSF), brain MRI, FP-CIT SPECT, MIBG SPECT, perfusion SPECT, FDG-PET by focusing more specifically on differential diagnosis between DLB and AD. FP-CIT SPECT is of high interest to discriminate DLB and AD, but not at the prodromal stage. Brain MRI has shown differences in group study with lower grey matter concentration of the Insula in prodromal DLB, but its interest in clinical routine is not demonstrated. Among the AD biomarkers (t-Tau, phospho-Tau181, Aβ42 and Aβ40) used routinely, t-Tau and phospho-Tau181 have shown excellent discrimination whatever the clinical stages severity. CSF Alpha-synuclein assay in the CSF has also an interest in the discrimination between DLB and AD but not in segregation between DLB and healthy elderly subjects. CSF synuclein RT-QuIC seems to be an excellent biomarker but its application in clinical routine remains to be demonstrated, given the non-automation of the process.
Collapse
Affiliation(s)
- F Blanc
- Hôpitaux Universitaire de Strasbourg, CM2R (Centre Mémoire de Ressource et de Recherche), Hôpital de jour, pôle de Gériatrie, Strasbourg, France; CNRS, laboratoire ICube UMR 7357 et FMTS (Fédération de Médecine Translationnelle de Strasbourg), équipe IMIS, Strasbourg, France.
| | - O Bousiges
- CNRS, laboratoire ICube UMR 7357 et FMTS (Fédération de Médecine Translationnelle de Strasbourg), équipe IMIS, Strasbourg, France; Hôpitaux Universitaire de Strasbourg, Laboratoire de Biochimie et Biologie Moléculaire, Strasbourg, France
| |
Collapse
|
33
|
Klyucherev TO, Olszewski P, Shalimova AA, Chubarev VN, Tarasov VV, Attwood MM, Syvänen S, Schiöth HB. Advances in the development of new biomarkers for Alzheimer's disease. Transl Neurodegener 2022; 11:25. [PMID: 35449079 PMCID: PMC9027827 DOI: 10.1186/s40035-022-00296-z] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 03/28/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is a complex, heterogeneous, progressive disease and is the most common type of neurodegenerative dementia. The prevalence of AD is expected to increase as the population ages, placing an additional burden on national healthcare systems. There is a large need for new diagnostic tests that can detect AD at an early stage with high specificity at relatively low cost. The development of modern analytical diagnostic tools has made it possible to determine several biomarkers of AD with high specificity, including pathogenic proteins, markers of synaptic dysfunction, and markers of inflammation in the blood. There is a considerable potential in using microRNA (miRNA) as markers of AD, and diagnostic studies based on miRNA panels suggest that AD could potentially be determined with high accuracy for individual patients. Studies of the retina with improved methods of visualization of the fundus are also showing promising results for the potential diagnosis of the disease. This review focuses on the recent developments of blood, plasma, and ocular biomarkers for the diagnosis of AD.
Collapse
Affiliation(s)
- Timofey O Klyucherev
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, Sweden.,Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Pawel Olszewski
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, Sweden
| | - Alena A Shalimova
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, Sweden.,Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vladimir N Chubarev
- Institute of Translational Medicine and Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vadim V Tarasov
- Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, Russia.,Institute of Translational Medicine and Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Misty M Attwood
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, Sweden
| | - Stina Syvänen
- Department of Public Health and Caring Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
34
|
Zou X, Yuan Y, Liao Y, Jiang C, Zhao F, Ding D, Gu Y, Chen L, Chu Y, Hsu Y, Liebig PA, Xu B, Mao Y. Moyamoya disease: A human model for chronic hypoperfusion and intervention in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12285. [PMID: 35415209 PMCID: PMC8985488 DOI: 10.1002/trc2.12285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/10/2022] [Accepted: 03/03/2022] [Indexed: 11/07/2022]
Abstract
Introduction Chronic cerebral hypoperfusion has been considered the etiology for sporadic Alzheimer's disease (AD). However, no valid clinical evidence exists due to the similar risk factors between cerebrovascular disease and AD. Methods We used moyamoya disease (MMD) as a model of chronic hypoperfusion and cognitive impairment, without other etiology interference. Results Based on the previous reports and preliminary findings, we hypothesized that chronic cerebral hypoperfusion could be an independent upstream crucial variable, resulting in AD, and induce pathological hallmarks such as amyloid beta peptide and hyperphosphorylated tau accumulation. Discussion Timely intervention with revascularisation would help reverse the brain damage with AD hallmarks and lead to cognitive improvement.
Collapse
Affiliation(s)
- Xiang Zou
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Yifan Yuan
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Yujun Liao
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Conglin Jiang
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Fan Zhao
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Ding Ding
- Huashan HospitalInstitute of NeurologyFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Yuxiang Gu
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Liang Chen
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
- Tianqiao and Chrissy Chen International Institute for Brain DiseasesShanghaiChina
| | - Ying‐Hua Chu
- MR CollaborationSiemens Healthineers Ltd.ShanghaiChina
| | - Yi‐Cheng Hsu
- MR CollaborationSiemens Healthineers Ltd.ShanghaiChina
| | | | - Bin Xu
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Ying Mao
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
- Huashan HospitalInstitute of NeurologyFudan UniversityShanghaiChina
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceSchool of Basic Medical Sciences and Institutes of Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
35
|
Liu P, Wang Y, Sun Y, Peng G. Neuroinflammation as a Potential Therapeutic Target in Alzheimer’s Disease. Clin Interv Aging 2022; 17:665-674. [PMID: 35520949 PMCID: PMC9064449 DOI: 10.2147/cia.s357558] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/12/2022] [Indexed: 12/14/2022] Open
Abstract
Although amyloid-β (Aβ) peptide accumulation is considered as a key early event in the pathogenesis of Alzheimer’s disease (AD), the precise pathophysiology of this deadly illness remains unclear and no effective remedies capable of inhibiting disease progression have been discovered. In addition to deposition of extracellular Aβ plaques and intracellular neurofibrillary tangles, neuroinflammation has been identified as the third core characteristic crucial in the pathogenesis of AD. More and more evidence from laboratory and clinical studies have suggested that anti-inflammatory treatments could defer or prevent the occurrence of AD. In this review, we will discuss multifaceted evidence of neuroinflammation presented in AD and the newly emerged anti-inflammatory targets both in pre-clinical and clinical AD.
Collapse
Affiliation(s)
- Ping Liu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Yunyun Wang
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- Department of Neurology, Shengzhou People’s Hospital, Shaoxing, People’s Republic of China
| | - Yan Sun
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Guoping Peng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- Correspondence: Guoping Peng, Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, People’s Republic of China, Tel +86 13588150613, Email
| |
Collapse
|
36
|
Morar U, Izquierdo W, Martin H, Forouzannezhad P, Zarafshan E, Unger E, Bursac Z, Cabrerizo M, Barreto A, Vaillancourt DE, DeKosky ST, Loewenstein D, Duara R, Adjouadi M. A study of the longitudinal changes in multiple cerebrospinal fluid and volumetric magnetic resonance imaging biomarkers on converter and non-converter Alzheimer's disease subjects with consideration for their amyloid beta status. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12258. [PMID: 35229014 PMCID: PMC8865744 DOI: 10.1002/dad2.12258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 06/14/2023]
Abstract
INTRODUCTION This study aims to determine whether newly introduced biomarkers Visinin-like protein-1 (VILIP-1), chitinase-3-like protein 1 (YKL-40), synaptosomal-associated protein 25 (SNAP-25), and neurogranin (NG) in cerebrospinal fluid are useful in evaluating the asymptomatic and early symptomatic stages of Alzheimer's disease (AD). It further aims to shed new insight into the differences between stable subjects and those who progress to AD by associating cerebrospinal fluid (CSF) biomarkers and specific magnetic resonance imaging (MRI) regions with disease progression, more deeply exploring how such biomarkers relate to AD pathology. METHODS We examined baseline and longitudinal changes over a 7-year span and the longitudinal interactions between CSF and MRI biomarkers for subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI). We stratified all CSF (140) and MRI (525) cohort participants into five diagnostic groups (including converters) further dichotomized by CSF amyloid beta (Aβ) status. Linear mixed models were used to compare within-person rates of change across diagnostic groups and to evaluate the association of CSF biomarkers as predictors of magnetic resonance imaging (MRI) biomarkers. CSF biomarkers and disease-prone MRI regions are assessed for CSF proteins levels and brain structural changes. RESULTS VILIP-1 and SNAP-25 displayed within-person increments in early symptomatic, amyloid-positive groups. CSF amyloid-positive (Aβ+) subjects showed elevated baseline levels of total tau (tTau), phospho-tau181 (pTau), VILIP-1, and NG. YKL-40, SNAP-25, and NG are positively intercorrelated. Aβ+ subjects showed negative MRI biomarker changes. YKL-40, tTau, pTau, and VILIP-1 are longitudinally associated with MRI biomarkers atrophy. DISCUSSION Converters (CNc, MCIc) highlight the evolution of biomarkers during the disease progression. Results show that underlying amyloid pathology is associated with accelerated cognitive impairment. CSF levels of Aβ42, pTau, tTau, VILIP-1, and SNAP-25 show utility to discriminate between mild cognitive impairment (MCI) converter and control subjects (CN). Higher levels of YKL-40 in the Aβ+ group were longitudinally associated with declines in temporal pole and entorhinal thickness. Increased levels of tTau, pTau, and VILIP-1 in the Aβ+ groups were longitudinally associated with declines in hippocampal volume. These CSF biomarkers should be used in assessing the characterization of the AD progression.
Collapse
Affiliation(s)
- Ulyana Morar
- Center for Advanced Technology and EducationDepartment of Electrical and Computer EngineeringFlorida International UniversityMiamiFloridaUSA
| | - Walter Izquierdo
- Center for Advanced Technology and EducationDepartment of Electrical and Computer EngineeringFlorida International UniversityMiamiFloridaUSA
| | - Harold Martin
- Center for Advanced Technology and EducationDepartment of Electrical and Computer EngineeringFlorida International UniversityMiamiFloridaUSA
| | - Parisa Forouzannezhad
- Center for Advanced Technology and EducationDepartment of Electrical and Computer EngineeringFlorida International UniversityMiamiFloridaUSA
| | - Elaheh Zarafshan
- Center for Advanced Technology and EducationDepartment of Electrical and Computer EngineeringFlorida International UniversityMiamiFloridaUSA
| | - Elona Unger
- College of PharmacyFlorida A&M UniversityTallahasseeFloridaUSA
| | - Zoran Bursac
- Department of BiostatisticsRobert Stempel College of Public HealthFlorida International UniversityMiami
| | - Mercedes Cabrerizo
- Center for Advanced Technology and EducationDepartment of Electrical and Computer EngineeringFlorida International UniversityMiamiFloridaUSA
| | - Armando Barreto
- Center for Advanced Technology and EducationDepartment of Electrical and Computer EngineeringFlorida International UniversityMiamiFloridaUSA
| | - David E. Vaillancourt
- Department of Neurology and McKnight Brain InstituteCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFloridaUSA
- Florida Alzheimer's Disease Research Center (ADRC)University of FloridaGainesvilleFloridaUSA
| | - Steven T. DeKosky
- Department of Neurology and McKnight Brain InstituteCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Florida Alzheimer's Disease Research Center (ADRC)University of FloridaGainesvilleFloridaUSA
| | - David Loewenstein
- Florida Alzheimer's Disease Research Center (ADRC)University of FloridaGainesvilleFloridaUSA
- Department of Psychiatry and Behavioral SciencesMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Ranjan Duara
- Florida Alzheimer's Disease Research Center (ADRC)University of FloridaGainesvilleFloridaUSA
- Wien Center for Alzheimer's Disease and Memory DisordersMount Sinai Medical CenterMiamiFloridaUSA
| | - Malek Adjouadi
- Center for Advanced Technology and EducationDepartment of Electrical and Computer EngineeringFlorida International UniversityMiamiFloridaUSA
- Florida Alzheimer's Disease Research Center (ADRC)University of FloridaGainesvilleFloridaUSA
| |
Collapse
|
37
|
Cervantes González A, Belbin O. Fluid markers of synapse degeneration in synucleinopathies. J Neural Transm (Vienna) 2022; 129:187-206. [PMID: 35147800 DOI: 10.1007/s00702-022-02467-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/20/2022] [Indexed: 01/06/2023]
Abstract
The abnormal accumulation of α-synuclein in the brain is a common feature of Parkinson's disease (PD), PD dementia (PDD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA), and synucleinopathies that present with overlapping but distinct clinical symptoms that include motor and cognitive deficits. Synapse degeneration is the crucial neuropathological event in these synucleinopathies and the neuropathological correlate of connectome dysfunction. The cognitive and motor deficits resulting from the connectome dysfunction are currently measured by scalar systems that are limited in their sensitivity and largely subjective. Ideally, a marker of synapse degeneration would correlate with measures of cognitive or motor impairment, and could therefore be used as a more objective, surrogate biomarker of the core clinical features of these diseases. Furthermore, an objective surrogate biomarker that can detect and monitor the progression of synapse degeneration would improve patient management and clinical trial design, and could provide a measure of therapeutic response. Here, we review the published findings relating to candidate biomarkers of synapse degeneration in PD, PDD, DLB, and MSA patient-derived biofluids and discuss the findings in the context of the mechanisms associated with α-synuclein-mediated synapse degeneration. Understanding these mechanisms is essential not only for discovery of biomarkers, but also to improve our understanding of the earliest changes in disease pathogenesis of synucleinopathies.
Collapse
Affiliation(s)
- Alba Cervantes González
- Neurology Department, Biomedical Research Institute Sant Pau (IIB Sant Pau) and Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Olivia Belbin
- Neurology Department, Biomedical Research Institute Sant Pau (IIB Sant Pau) and Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.
| |
Collapse
|
38
|
Karaboğa MNS, Sezgintürk MK. Biosensor approaches on the diagnosis of neurodegenerative diseases: Sensing the past to the future. J Pharm Biomed Anal 2022; 209:114479. [PMID: 34861607 DOI: 10.1016/j.jpba.2021.114479] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/05/2021] [Accepted: 11/14/2021] [Indexed: 12/12/2022]
Abstract
Early diagnosis of neurodegeneration-oriented diseases that develop with the aging world is essential for improving the patient's living conditions as well as the treatment of the disease. Alzheimer's and Parkinson's diseases are prominent examples of neurodegeneration characterized by dementia leading to the death of nerve cells. The clinical diagnosis of these diseases only after the symptoms appear, delays the treatment process. Detection of biomarkers, which are distinctive molecules in biological fluids, involved in neurodegeneration processes, has the potential to allow early diagnosis of neurodegenerative diseases. Studies on biosensors, whose main responsibility is to detect the target analyte with high specificity, has gained momentum in recent years with the aim of high detection of potential biomarkers of neurodegeneration process. This study aims to provide an overview of neuro-biosensors developed on the basis of biomarkers identified in biological fluids for the diagnosis of neurodegenerative diseases such as Alzheimer's disease (AD), and Parkinson's disease (PD), and to provide an overview of the urgent needs in this field, emphasizing the importance of early diagnosis in the general lines of the neurodegeneration pathway. In this review, biosensor systems developed for the detection of biomarkers of neurodegenerative diseases, especially in the last 5 years, are discussed.
Collapse
|
39
|
Mortamais M, Laure-Anne G, Balem M, Bars EL, de Champfleur NM, Bouyahia A, Chupin M, Perus L, Fisher C, Vellas B, Andrieu S, Mangin JF, Berr C, Gabelle A. Sulcal morphology as cognitive decline predictor in older adults with memory complaints. Neurobiol Aging 2022; 113:84-94. [DOI: 10.1016/j.neurobiolaging.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/08/2021] [Accepted: 02/08/2022] [Indexed: 11/16/2022]
|
40
|
Roveta F, Cermelli A, Boschi S, Ferrandes F, Grassini A, Marcinnò A, Spina M, Rubino E, Borsello T, Vercelli A, Rainero I. Synaptic Proteins as Fluid Biomarkers in Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2022; 90:1381-1393. [PMID: 36278349 DOI: 10.3233/jad-220515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Synaptic disruption precedes neuronal death and correlates with clinical features of Alzheimer's disease (AD). The identification of fluid biomarkers of synaptic damage is emerging as a goal for early and accurate diagnosis of the disease. OBJECTIVE To perform a systematic review and meta-analysis to determine whether fluid biomarkers of synaptic damage are impaired in AD. METHODS PubMed, Scopus, EMBASE, and Web of Science were searched for articles reporting synaptic proteins as fluid biomarkers in AD and cognitively unimpaired (CU) individuals. Pooled effect sizes were determined using the Hedge G method with random effects. Questions adapted from the Quality Assessment of Diagnostic Accuracy Studies were applied for quality assessment. A protocol for this study has been previously registered in PROSPERO (registration number: CRD42021277487). RESULTS The search strategy identified 204 articles that were assessed for eligibility. A total of 23 studies were included in the systematic review and 15 were included in the meta-analysis. For Neurogranin, 827 AD and 1,237 CU subjects were included in the meta-analysis, showing a significant increase in cerebrospinal fluid of patients with AD compared to CU individuals, with an effect size of 1.01 (p < 0.001). A significant increase in SNAP-25 and GAP-43 levels in CSF of patients with AD was observed. CONCLUSION Neurogranin, SNAP-25, and GAP-43 are possible biomarkers of synaptic damage in AD, and other potential synaptic biomarkers are emerging. This meta-analysis also revealed that there are still relatively few studies investigating these biomarkers in patients with AD or other dementias and showed wide heterogeneity in literature.
Collapse
Affiliation(s)
- Fausto Roveta
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Aurora Cermelli
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Silvia Boschi
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Fabio Ferrandes
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Alberto Grassini
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Andrea Marcinnò
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Margherita Spina
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Elisa Rubino
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| | - Tiziana Borsello
- Department of Pharmacological and Biomolecular Sciences University of Milano, Milan, Italy
- Mario Negri Institute for Pharmacological Research, University of Milano, Milan, Italy
| | - Alessandro Vercelli
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| | - Innocenzo Rainero
- Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy
| |
Collapse
|
41
|
Mavroudis I, Chowdhury R, Petridis F, Karantali E, Chatzikonstantinou S, Balmus IM, Luca IS, Ciobica A, Kazis D. YKL-40 as a Potential Biomarker for the Differential Diagnosis of Alzheimer's Disease. MEDICINA (KAUNAS, LITHUANIA) 2021; 58:medicina58010060. [PMID: 35056368 PMCID: PMC8777884 DOI: 10.3390/medicina58010060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder, associated with extensive neuronal loss, dendritic and synaptic changes resulting in significant cognitive impairment. An increased number of studies have given rise to the neuroinflammatory hypothesis in AD. It is widely accepted that AD brains show chronic inflammation, probably triggered by the presence of insoluble amyloid beta deposits and neurofibrillary tangles (NFT) and is also related to the activation of neuronal death cascade. In the present study we aimed to investigate the role of YKL-40 levels in the cerebrospinal fluid (CSF) in the diagnosis of AD, and to discuss whether there are further potential roles of this protein in the management and treatment of AD. We conducted an online search on PubMed, Web of Science, and the Cochrane library databases from 1990 to 2021. The quantitative analysis showed that the levels of YKL-40 were significantly higher in Alzheimer’s disease compared to controls, to mild cognitive impairment (MCI) AD (MCI-AD) and to stable MCI. They were also increased in MCI-AD compared to stable MCI. The present study shows that the CSF levels of YKL-40 could be potentially used as a biomarker for the prognosis of mild cognitive impairment and the likelihood of progression to AD, as well as for the differential diagnosis between AD and MCI.
Collapse
Affiliation(s)
- Ioannis Mavroudis
- Department of Neurology, Leeds Teaching Hospitals, NHS Trust, Leeds LS2 9JT, UK; (I.M.); (R.C.)
| | - Rumana Chowdhury
- Department of Neurology, Leeds Teaching Hospitals, NHS Trust, Leeds LS2 9JT, UK; (I.M.); (R.C.)
| | - Foivos Petridis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| | - Eleni Karantali
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| | - Symela Chatzikonstantinou
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| | - Ioana Miruna Balmus
- Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, ”Alexandru Ioan Cuza” University of Iasi, Alexandru Lapsuneanu Street, No. 26, 700057 Iasi, Romania;
| | - Iuliana Simona Luca
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, No. 20A, 700505 Iasi, Romania
- Correspondence: (I.S.L.); (A.C.)
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, No. 20A, 700505 Iasi, Romania
- Correspondence: (I.S.L.); (A.C.)
| | - Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| |
Collapse
|
42
|
Agnello L, Lo Sasso B, Vidali M, Scazzone C, Piccoli T, Gambino CM, Bivona G, Giglio RV, Ciaccio AM, La Bella V, Ciaccio M. Neurogranin as a Reliable Biomarker for Synaptic Dysfunction in Alzheimer's Disease. Diagnostics (Basel) 2021; 11:diagnostics11122339. [PMID: 34943576 PMCID: PMC8700711 DOI: 10.3390/diagnostics11122339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 01/02/2023] Open
Abstract
(1) Background: Neurogranin is a post-synaptic protein expressed in the neurons of the hippocampus and cerebral cortex. It has been recently proposed as a promising biomarker of synaptic dysfunction, especially in Alzheimer's disease (AD). However, more efforts are needed before introducing it in clinical practice, including the definition of its reference interval (RI). The aim of the study was to establish the RI of cerebrospinal fluid (CSF) neurogranin levels in controls and individuals with non-neurodegenerative neurological diseases; (2) We included a total of 136 individuals that were sub-grouped as follows: AD patients (n = 33), patients with non-neurodegenerative neurological diseases (n = 70) and controls (33). We measured CSF neurogranin levels by a commercial ELISA kit. CSF RI of neurogranin was calculated by a robust method; (3) Results: AD patients showed increased levels of neurogranin. We also found that neurogranin was significantly correlated with T-tau, P-tau and mini mental state examination in AD patients. The lower and upper reference limits of the RI were 2.9 (90%CI 0.1-10.8) and 679 (90%CI 595-779), respectively; (4) Conclusion: This is the first study establishing the RI of CSF neurogranin.
Collapse
Affiliation(s)
- Luisa Agnello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
| | - Bruna Lo Sasso
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
- Department of Laboratory Medicine, Azienda Ospedaliera Universitaria Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Matteo Vidali
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Concetta Scazzone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
| | - Tommaso Piccoli
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy;
| | - Caterina Maria Gambino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
| | - Giulia Bivona
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
| | - Rosaria Vincenza Giglio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
| | - Anna Maria Ciaccio
- Unit of Clinical Biochemistry, University of Palermo, 90127 Palermo, Italy;
| | - Vincenzo La Bella
- ALS Clinical Research Center, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy;
| | - Marcello Ciaccio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
- Department of Laboratory Medicine, Azienda Ospedaliera Universitaria Policlinico “P. Giaccone”, 90127 Palermo, Italy
- Correspondence: ; Tel.: +39-0916553296
| |
Collapse
|
43
|
Hao Y, Liu X, Zhu R. Neurodegeneration and Glial Activation Related CSF Biomarker as the Diagnosis of Alzheimer's Disease: A Systematic Review and an Updated Meta-analysis. Curr Alzheimer Res 2021; 19:32-46. [PMID: 34879804 DOI: 10.2174/1567205018666211208142702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/29/2021] [Accepted: 10/21/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Recently, neuron specific enolase (NSE), Visinin-like protein-1 (VLP-1), neurogranin (Ng), and YKL-40 have been identified as candidates for neuronal degeneration and glial activation biomarkers. Therefore, we perform a comprehensive meta-analysis to assess the diagnostic value of CSF NSE, VLP-1, Ng and YKL-40 in Alzheimer's disease (AD). METHODS We searched Pubmed, MEDLINE, EMBASE databases for research about the levels of CSF NSE, VLP-1, Ng and YKL-40 in AD patients compared with controls or other dementia diseases until Dec 2020. RESULTS The present meta-analysis contained a total of 51 studies comprising 6248 patients with dementia disorders and 3861 controls. Among them, there were 3262 patients with AD, 2456 patients with mild cognitive impairment (MCI), 173 patients with vascular dementia (VaD), 221 patients with frontotemporal dementia (FTD), and 136 with Lewy bodies dementia (DLB). Our study demonstrated that CSF NSE, VLP-1, Ng and YKL-40 levels were increased in AD as compared to healthy controls. We also observed that the CSF NSE level was higher in AD than VaD, suggesting CSF NSE might act as a key role in distinguishing between AD and VaD. Interestingly, there was a higher VLP-1 expression in AD, and a lower expression in DLB patients. Moreover, we found the CSF Ng level was increased in AD than MCI, implying CSF Ng might be a biomarker for identifying the progression of AD. Additionally, a significantly higher CSF YKL-40 level was detected not only in AD, but also in FTD, DLB, VaD, signifying YKL-40 was not sensitive in the diagnosis of AD. CONCLUSION Our study confirmed that CSF levels of NSE, VLP-1, and Ng could be valuable biomarkers for identifying patients who are more susceptible to AD and distinguishing AD from other neurodegenerative dementia disorders.
Collapse
Affiliation(s)
- Yuehan Hao
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110001. China
| | - Xu Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110001. China
| | - Ruixia Zhu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110001. China
| |
Collapse
|
44
|
Yoong SQ, Lu J, Xing H, Gyanwali B, Tan YQ, Wu XV. The prognostic utility of CSF neurogranin in predicting future cognitive decline in the Alzheimer's disease continuum: A systematic review and meta-analysis with narrative synthesis. Ageing Res Rev 2021; 72:101491. [PMID: 34688925 DOI: 10.1016/j.arr.2021.101491] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/13/2021] [Accepted: 10/15/2021] [Indexed: 01/08/2023]
Abstract
Core cerebrospinal fluid (CSF) biomarkers (Aβ42, T-tau, P-tau) were included as supporting diagnostic criteria for Alzheimer's Disease (AD), but they lack the power to predict AD progression. On the other hand, a new biomarker CSF Neurogranin (Ng) has been shown to predict cognitive decline. This systematic review aims to synthesise the prognostic utility of CSF Ng in predicting cognitive decline in the AD continuum. Seven databases were searched systematically from inception to 30 September 2020. Participants were 55 years or older, who had baseline and at least one follow-up cognitive assessments. Risk of bias was assessed using the Quality in Prognosis Studies tool. Meta-analysis was conducted by pooling standardised beta coefficients and adjusted hazard ratios. Thirteen studies were included and high-quality evidence suggests that CSF Ng predicts Mini-Mental State Examination (MMSE) decline in Aβ+ mild cognitive impairment (MCI). Moderate quality evidence showed that CSF Ng could predict the decline of memory and executive function in MCI. Narrative synthesis found that CSF Ng/Aβ42 was also likely to predict cognitive decline. More studies are required to validate the use of CSF Ng as an AD prognostic marker and its application in future development of drug treatment and diagnosis.
Collapse
|
45
|
Milà-Alomà M, Brinkmalm A, Ashton NJ, Kvartsberg H, Shekari M, Operto G, Salvadó G, Falcon C, Gispert JD, Vilor-Tejedor N, Arenaza-Urquijo EM, Grau-Rivera O, Sala-Vila A, Sanchez-Benavides G, González-de-Echávarri JM, Minguillon C, Fauria K, Niñerola-Baizán A, Perissinotti A, Kollmorgen G, Suridjan I, Zetterberg H, Molinuevo JL, Blennow K, Suárez-Calvet M. CSF Synaptic Biomarkers in the Preclinical Stage of Alzheimer Disease and Their Association With MRI and PET: A Cross-sectional Study. Neurology 2021; 97:e2065-e2078. [PMID: 34556565 PMCID: PMC8610620 DOI: 10.1212/wnl.0000000000012853] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 09/08/2021] [Indexed: 12/05/2022] Open
Abstract
Background and Objectives To determine whether CSF synaptic biomarkers are altered in the early preclinical stage of the Alzheimer continuum and associated with Alzheimer disease (AD) risk factors, primary pathology, and neurodegeneration markers. Methods This cross-sectional study was performed in the Alzheimer's and Families (ALFA+) cohort, comprising middle-aged cognitively unimpaired participants. CSF neurogranin and growth-associated protein-43 (GAP-43) were measured with immunoassays, and synaptosomal-associated protein-25 (SNAP-25) and synaptotagmin-1 were measured with immunoprecipitation mass spectrometry. AD CSF biomarkers β-amyloid (Aβ)42/40, phosphorylated tau (p-tau), and total tau and the neurodegeneration biomarker neurofilament light chain (NfL) were also measured. Participants underwent structural MRI and fluorodeoxyglucose and Aβ PET imaging. General linear modeling was used to test the associations between CSF synaptic biomarkers and risk factors, Aβ pathology, tau pathology, and neurodegeneration markers. Results All CSF synaptic biomarkers increased with age. CSF neurogranin was higher in females, while CSF SNAP-25 was higher in APOE ε4 carriers. All CSF synaptic biomarkers increased with higher Aβ load (as measured by CSF Aβ42/40 and Aβ PET Centiloid values), and it is important to note that the synaptic biomarkers were increased even in individuals in the earliest stages of Aβ deposition. Higher CSF synaptic biomarkers were also associated with higher CSF p-tau and NfL. Higher CSF neurogranin and GAP-43 were significantly associated with higher brain metabolism but lower cortical thickness in AD-related brain regions. Discussion CSF synaptic biomarkers increase in the early preclinical stages of the Alzheimer continuum even when a low burden of Aβ pathology is present, and they differ in their association with age, sex, APOE ε4, and markers of neurodegeneration. Trial Registration Information ClinicalTrials.gov Identifier NCT02485730.
Collapse
Affiliation(s)
- Marta Milà-Alomà
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Ann Brinkmalm
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Nicholas J Ashton
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Hlin Kvartsberg
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Mahnaz Shekari
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Grégory Operto
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Gemma Salvadó
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Carles Falcon
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Juan Domingo Gispert
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Natalia Vilor-Tejedor
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Eider M Arenaza-Urquijo
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Oriol Grau-Rivera
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Aleix Sala-Vila
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Gonzalo Sanchez-Benavides
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - José María González-de-Echávarri
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Carolina Minguillon
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Karine Fauria
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Aida Niñerola-Baizán
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Andrés Perissinotti
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Gwendlyn Kollmorgen
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Ivonne Suridjan
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Henrik Zetterberg
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - José Luis Molinuevo
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Kaj Blennow
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark
| | - Marc Suárez-Calvet
- From the Barcelonaβeta Brain Research Center (BBRC) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., N.V.-T., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., K.F., J.L.M., M.S.-C.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.M.-A., M.S., G.O., G.S., C.F., J.D.G., E.M.A.-U., O.G.-R., A.S.-V., G.S.-B., J.M.G.-d-E., C.M., M.S.-C.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES) (M.M.-A., G.O., E.M.A.-U., O.G.-R., G.S.-B., C.M., K.F., M.S.-C.), Madrid; Universitat Pompeu Fabra (M.M.-A., M.S.), Barcelona, Spain; Department of Psychiatry and Neurochemistry (A.B., N.J.A., H.K., H.Z., K.B.), Institute of Neuroscience and Physiology, University of Gothenburg; Clinical Neurochemistry Laboratory (A.B., H.K., H.Z., K.B.), Sahlgrenska University Hospital, Mölndal; Wallenberg Centre for Molecular and Translational Medicine (A.B., N.J.A., H.K.), Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation (N.J.A.), London, UK; Centro de Investigación Biomédica en Red de Bioingeniería (C.F., J.D.G., A.N.-B., A.P.), Biomateriales y Nanomedicina (CIBER-BBN), Madrid; Centre for Genomic Regulation (CRG) (N.V.-T.), Barcelona Institute for Science and Technology; Department of Clinical Genetics (N.V.-T.), Erasmus MC, University Medical Center Rotterdam, the Netherlands; Servei de Neurologia (O.G.-R., M.S.-C.), Hospital del Mar; Servei de Medicina Nuclear (A.N.-B., A.P.), Hospital Clínic, Barcelona, Spain; Roche Diagnostics GmbH (G.K.), Penzberg, Germany; Roche Diagnostics International Ltd (I.S.), Rotkreuz, Switzerland; UK Dementia Research Institute at UCL (H.Z.), London; Department of Neurodegenerative Disease (H.Z.), UCL Queen Square Institute of Neurology, London, UK; and H. Lundbeck A/S (J.L.M.), Copenhagen, Denmark.
| | | |
Collapse
|
46
|
Willemse EAJ, Sieben A, Somers C, Vermeiren Y, De Roeck N, Timmers M, Van Broeckhoven C, De Vil B, Cras P, De Deyn PP, Martin JJ, Teunissen CE, Engelborghs S, Bjerke M. Neurogranin as biomarker in CSF is non-specific to Alzheimer's disease dementia. Neurobiol Aging 2021; 108:99-109. [PMID: 34551375 DOI: 10.1016/j.neurobiolaging.2021.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 07/10/2021] [Accepted: 08/01/2021] [Indexed: 11/19/2022]
Abstract
We aimed to evaluate the specificity of neurogranin (Ng) for Alzheimer's disease (AD) in a dementia cohort. Cerebrospinal fluid (CSF) Ng was measured (ELISA) in two independent cohorts: (1) clinical (n = 116; age 72±11 years): AD, non-AD (+high T-tau), and controls; and (2) autopsy-confirmed (n = 97; age 71±11 years): AD and non-AD, and 50 controls (age 60±6 years). In 16 autopsy-confirmed AD and 8 control subjects, Ng was measured in tissue (BA6+BA22). Ng was compared across diagnostic groups or neuropathological staging using multilinear regression models. Median[IQR] Ng concentrations were elevated in AD (414[315-499]pg/mL) and non-AD (464[319-699]pg/mL) compared to controls (260[193-306]pg/mL), but highest in AD-high-T-tau (874[716, 1148] pg/mL) and Creutzfeldt-Jakob disease (CJD; 828[703-1373]pg/mL) in cohort 1 (p < 0.01), but not in cohort 2: AD: 358[249-470]pg/mL; non-AD:245[137-416]pg/mL; controls: 259[193-370]pg/mL. Ng and tau biomarkers strongly correlated (r = 0.4-0.9, p < 0.05), except in CJD. CSF Ng concentrations were not associated with neuropathological AD hallmarks, nor with tissue Ng concentrations. CSF Ng is a general biomarker for synaptic degeneration, strongly correlating with CSF tau, but without added value for AD differential diagnosis.
Collapse
Affiliation(s)
- Eline A J Willemse
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Neurochemistry laboratory. Dept. of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands; Alzheimer Center, Dept. of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Anne Sieben
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Charisse Somers
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Yannick Vermeiren
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Naomi De Roeck
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Maarten Timmers
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Janssen Research and Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Christine Van Broeckhoven
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | - Bart De Vil
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Patrick Cras
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Department of Neurology, University Hospital Antwerp, Antwerp, Belgium
| | - Peter P De Deyn
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Jean-Jacques Martin
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Charlotte E Teunissen
- Neurochemistry laboratory. Dept. of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Department of Neurology and Center for Neurosciences (C4N), Universitair Ziekenhuis Brussel (UZ Brussel) and Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Maria Bjerke
- Reference Center for Biological Markers of Dementia (BIODEM) and Laboratory of Neurochemistry and Behavior, Laboratory of Neurobiology, Laboratory of Neurogenetics, and Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Neurochemistry laboratory, Department of Clinical Biology and Center for Neurosciences (C4N), Universitair Ziekenhuis Brussel (UZ Brussel) and Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| |
Collapse
|
47
|
Enache D, Pereira JB, Jelic V, Winblad B, Nilsson P, Aarsland D, Bereczki E. Increased Cerebrospinal Fluid Concentration of ZnT3 Is Associated with Cognitive Impairment in Alzheimer's Disease. J Alzheimers Dis 2021; 77:1143-1155. [PMID: 32925049 DOI: 10.3233/jad-200498] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cognitive deficits arising in the course of Alzheimer's disease (AD), dementia with Lewy bodies (DLB), and Parkinson's disease with dementia (PDD) are directly linked to synaptic loss. Postmortem studies suggest that zinc transporter protein 3 (ZnT3), AMPA glutamate receptor 3 (GluA3), and Dynamin1 are associated with cognitive decline in AD and Lewy body dementia patients. OBJECTIVE We aimed to evaluate the diagnostic value of ZnT3, GluA3, and Dynamin 1 in the cerebrospinal fluid (CSF) of patients with dementia due to AD, DLB, and PDD compared to cognitively normal subjective cognitive decline (SCD) patients in a retrospective study. In addition, we assessed the relationship between synaptic markers and age, sex, cognitive impairment, and depressive symptoms as well as CSF amyloid, phosphorylated tau (p-tau), and total tau (T-tau). METHODS Commercially available ELISA immunoassay was used to measure the levels of proteins in a total of 97 CSF samples from AD (N = 24), PDD (N = 18), DLB (N = 27), and SCD (N = 28) patients. Cognitive impairment was assessed using the Mini-Mental State Examination (MMSE). RESULTS We found a significant increase in the concentrations of ZnT3, GluA3, and Dynamin1 in AD (p = 0.002) and of ZnT3 and Dynamin 1 in DLB (p = 0.001, p = 0.002) when compared to SCD patients. Changes in ZnT3 concentrations correlated with MMSE scores in AD (p = 0.011), and with depressive symptoms in SCD (p = 0.041). CONCLUSION We found alteration of CSF levels of synaptic proteins in AD, PDD, and DLB. Our results reveal distinct changes in CSF concentrations of ZnT3 that could reflect cognitive impairment in AD with implications for future prognostic and diagnostic marker development.
Collapse
Affiliation(s)
- Daniela Enache
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Joana B Pereira
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Vesna Jelic
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Dag Aarsland
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden.,Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Erika Bereczki
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
48
|
Thordardottir S, Almkvist O, Johansson C, Zetterberg H, Blennow K, Graff C. Cerebrospinal Fluid YKL-40 and Neurogranin in Familial Alzheimer's Disease: A Pilot Study. J Alzheimers Dis 2021; 76:941-953. [PMID: 32568193 PMCID: PMC7505010 DOI: 10.3233/jad-191261] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND YKL-40 and neurogranin are promising additional cerebrospinal fluid (CSF) biomarkers for Alzheimer's disease (AD) which reflect different underlying disease mechanisms. OBJECTIVE To compare the levels of CSF YKL-40 and neurogranin between asymptomatic carriers of familial AD (FAD) mutations (MC) and non-carriers (NC) from the same families. Another objective was to assess changes in YKL-40 and neurogranin, from the presymptomatic to clinical phase of FAD. METHODS YKL-40 and neurogranin, as well as Aβ42, total tau-protein, and phospho-tau, were measured in the CSF of 14 individuals carrying one of three FAD mutations, APPswe (p.KM670/671NL), APParc (p.E693G), and PSEN1 (p.H163Y), as well as in 17 NC from the same families. Five of the MC developed mild cognitive impairment (MCI) during follow-up. RESULTS In this pilot study, there was no difference in either CSF YKL-40 or neurogranin when comparing the presymptomatic MC to the NC. YKL-40 correlated positively with expected years to symptom onset and to age in both the MC and the NC, while neurogranin had no correlation to either variable in either of the groups. A subgroup of the participants underwent more than one CSF sampling in which half of the MC developed MCI during follow-up. The longitudinal data showed an increase in YKL-40 levels in the MC as the expected symptom onset approached. Neurogranin remained stable over time in both the MC and the NC. CONCLUSION These findings support a positive correlation between progression from presymptomatic to symptomatic AD and levels of CSF YKL-40, but not neurogranin.
Collapse
Affiliation(s)
- Steinunn Thordardottir
- Department of NVS, Karolinska Institutet, Division of Neurogeriatrics, Center for Alzheimer Disease Research, Solna, Sweden.,Theme Aging, Karolinska University Hospital Huddinge, Unit for Hereditary Dementias, Solna, Sweden
| | - Ove Almkvist
- Department of NVS, Karolinska Institutet, Center for Alzheimer Research, Division of Clinical Geriatrics, Huddinge, Sweden
| | - Charlotte Johansson
- Department of NVS, Karolinska Institutet, Division of Neurogeriatrics, Center for Alzheimer Disease Research, Solna, Sweden.,Theme Aging, Karolinska University Hospital Huddinge, Unit for Hereditary Dementias, Solna, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,UCL Insitute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Caroline Graff
- Department of NVS, Karolinska Institutet, Division of Neurogeriatrics, Center for Alzheimer Disease Research, Solna, Sweden.,Theme Aging, Karolinska University Hospital Huddinge, Unit for Hereditary Dementias, Solna, Sweden
| |
Collapse
|
49
|
Fan Y, Gao Y, Therriault J, Luo J, Ba M, Zhang H. The Effects of CSF Neurogranin and APOE ε4 on Cognition and Neuropathology in Mild Cognitive Impairment and Alzheimer's Disease. Front Aging Neurosci 2021; 13:667899. [PMID: 33986657 PMCID: PMC8110906 DOI: 10.3389/fnagi.2021.667899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/30/2021] [Indexed: 01/08/2023] Open
Abstract
Cerebrospinal fluid (CSF) measurements of neurogranin (Ng) have emerged as a promising biomarker for cognitive decline in mild cognitive impairment (MCI) and Alzheimer’s disease (AD). The apolipoprotein E ε4 (APOE ε4) allele is by far the most consistent genetic risk factor for AD. However, it is not known whether the pathophysiological roles of Ng in MCI or AD are related to APOEε4. We stratified 250 participants from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database into cognitively normal (CN) ε4 negative (CN ε4−), CN ε4 positive (CN ε4+), MCI ε4 negative (MCI ε4−), MCI ε4 positive (MCI ε4+), AD ε4 negative (AD ε4−), and AD ε4 positive (AD ε4+). CSF Ng levels were significantly increased in APOE ε4 carriers compared to APOE ε4 non-carriers with MCI. In addition, CSF Ng identified MCI ε4+ versus CN ε4−, but not MCI ε4− versus CN ε4−. Similarly, CSF Ng negatively correlated with Mini-Mental State Examination (MMSE) scores at baseline in the MCI ε4+ group. Our findings support the use of CSF Ng as a biomarker of synaptic pathology for AD. We propose that the roles of CSF Ng in the pathophysiology of MCI may be related to APOE ε4.
Collapse
|
50
|
Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, Cummings J, van der Flier WM. Alzheimer's disease. Lancet 2021; 397:1577-1590. [PMID: 33667416 PMCID: PMC8354300 DOI: 10.1016/s0140-6736(20)32205-4] [Citation(s) in RCA: 1797] [Impact Index Per Article: 599.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 08/21/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022]
Abstract
In this Seminar, we highlight the main developments in the field of Alzheimer's disease. The most recent data indicate that, by 2050, the prevalence of dementia will double in Europe and triple worldwide, and that estimate is 3 times higher when based on a biological (rather than clinical) definition of Alzheimer's disease. The earliest phase of Alzheimer's disease (cellular phase) happens in parallel with accumulating amyloid β, inducing the spread of tau pathology. The risk of Alzheimer's disease is 60-80% dependent on heritable factors, with more than 40 Alzheimer's disease-associated genetic risk loci already identified, of which the APOE alleles have the strongest association with the disease. Novel biomarkers include PET scans and plasma assays for amyloid β and phosphorylated tau, which show great promise for clinical and research use. Multidomain lifestyle-based prevention trials suggest cognitive benefits in participants with increased risk of dementia. Lifestyle factors do not directly affect Alzheimer's disease pathology, but can still contribute to a positive outcome in individuals with Alzheimer's disease. Promising pharmacological treatments are poised at advanced stages of clinical trials and include anti-amyloid β, anti-tau, and anti-inflammatory strategies.
Collapse
Affiliation(s)
- Philip Scheltens
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Neurology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Life Science Partners, Amsterdam, Netherlands.
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven Department for Neurology, Leuven, Belgium; Dementia Research Institute, University College London, London, UK
| | - Miia Kivipelto
- Division of Clinical Geriatrics and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska University Hospital, Stockholm, Sweden; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Ageing and Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | - Henne Holstege
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Clinical Genetics, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Gael Chételat
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Groupement d'Intérêt Public Cyceron, Caen, France
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, University of Nevada, Las Vegas, NV, USA; Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Wiesje M van der Flier
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Epidemiology and Datascience, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|