551
|
Kim HJ, Cho H, Werring DJ, Jang YK, Kim YJ, Lee JS, Lee J, Jun S, Park S, Ryu YH, Choi JY, Cho YS, Moon SH, Na DL, Lyoo CH, Seo SW. 18F-AV-1451 PET Imaging in Three Patients with Probable Cerebral Amyloid Angiopathy. J Alzheimers Dis 2017; 57:711-716. [PMID: 28282808 DOI: 10.3233/jad-161139] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cerebrovascular deposition of amyloid-β, known as cerebral amyloid angiopathy (CAA), is associated with MRI findings of lobar hemorrhage, cerebral microbleeds, and cortical superficial siderosis. Although pathological studies suggest that tau may co-localize with vascular amyloid, this has not yet been investigated in CAA in vivo. Three patients with probable CAA underwent 11C-Pittsburgh Compound B (PiB) PET or 18F-florbetaben PET to evaluate amyloid burden, and 18F-AV-1451 PET to evaluate paired helical filament tau burden. Regions that had cerebral microbleeds or cortical superficial siderosis largely overlapped with those showing increased 18F-AV-1451. Our preliminary study raised the possibility that lobar cerebral microbleeds, and cortical superficial siderosis, which are characteristic markers of vascular amyloid, may be associated with local production of paired helical filament tau.
Collapse
Affiliation(s)
- Hee Jin Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - David J Werring
- Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, Queen Square, London, UK
| | - Young Kyoung Jang
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Yeo Jin Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Korea
| | - Jin San Lee
- Department of Neurology, Kyung Hee University Hospital, Seoul, Korea
| | - Juyoun Lee
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Department of Neurology, School of Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - Soomin Jun
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Seongbeom Park
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Yong Choi
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Seok Cho
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Duk L Na
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Won Seo
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
552
|
Ling H. Untangling the tauopathies: Current concepts of tau pathology and neurodegeneration. Parkinsonism Relat Disord 2017; 46 Suppl 1:S34-S38. [PMID: 28789904 DOI: 10.1016/j.parkreldis.2017.07.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 07/29/2017] [Indexed: 12/18/2022]
Abstract
Tau is the most common misfolded protein responsible for human neurodegenerative diseases. The identification of mutations in MAPT, the gene that encodes tau, causing dementia and parkinsonism established the notion that tau aggregation is responsible for the development of disease. An increased understanding of the pathway leading from conformational changes in tau protein and tau propagation to neuronal dysfunction, cell death and clinical manifestation will be the key for the development mechanism-based therapeutic strategies for tauopathies.
Collapse
Affiliation(s)
- Helen Ling
- Queen Square Brain Bank for Neurological Disorders, UCL Institute of Neurology, University College London, London, UK; Reta Lila Weston Institute for Neurological Studies, UCL Institute of Neurology, London, UK; Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, UK.
| |
Collapse
|
553
|
Le Guennec K, Quenez O, Nicolas G, Wallon D, Rousseau S, Richard AC, Alexander J, Paschou P, Charbonnier C, Bellenguez C, Grenier-Boley B, Lechner D, Bihoreau MT, Olaso R, Boland A, Meyer V, Deleuze JF, Amouyel P, Munter HM, Bourque G, Lathrop M, Frebourg T, Redon R, Letenneur L, Dartigues JF, Martinaud O, Kalev O, Mehrabian S, Traykov L, Ströbel T, Le Ber I, Caroppo P, Epelbaum S, Jonveaux T, Pasquier F, Rollin-Sillaire A, Génin E, Guyant-Maréchal L, Kovacs GG, Lambert JC, Hannequin D, Campion D, Rovelet-Lecrux A, Rovelet-Lecrux A. 17q21.31 duplication causes prominent tau-related dementia with increased MAPT expression. Mol Psychiatry 2017; 22:1119-1125. [PMID: 27956742 DOI: 10.1038/mp.2016.226] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/26/2016] [Accepted: 10/27/2016] [Indexed: 01/07/2023]
Abstract
To assess the role of rare copy number variations in Alzheimer's disease (AD), we conducted a case-control study using whole-exome sequencing data from 522 early-onset cases and 584 controls. The most recurrent rearrangement was a 17q21.31 microduplication, overlapping the CRHR1, MAPT, STH and KANSL1 genes that was found in four cases, including one de novo rearrangement, and was absent in controls. The increased MAPT gene dosage led to a 1.6-1.9-fold expression of the MAPT messenger RNA. Clinical signs, neuroimaging and cerebrospinal fluid biomarker profiles were consistent with an AD diagnosis in MAPT duplication carriers. However, amyloid positon emission tomography (PET) imaging, performed in three patients, was negative. Analysis of an additional case with neuropathological examination confirmed that the MAPT duplication causes a complex tauopathy, including prominent neurofibrillary tangle pathology in the medial temporal lobe without amyloid-β deposits. 17q21.31 duplication is the genetic basis of a novel entity marked by prominent tauopathy, leading to early-onset dementia with an AD clinical phenotype. This entity could account for a proportion of probable AD cases with negative amyloid PET imaging recently identified in large clinical series.
Collapse
Affiliation(s)
- K Le Guennec
- Inserm, U1079, faculté de médecine, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Centre for Genomic Medicine and Personalized Medicine, Rouen, France
| | - O Quenez
- Inserm, U1079, faculté de médecine, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Centre for Genomic Medicine and Personalized Medicine, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France
| | - G Nicolas
- Inserm, U1079, faculté de médecine, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Centre for Genomic Medicine and Personalized Medicine, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France
| | - D Wallon
- Inserm, U1079, faculté de médecine, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Centre for Genomic Medicine and Personalized Medicine, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France.,Department of Neurology, Rouen University Hospital, Rouen, France
| | - S Rousseau
- Inserm, U1079, faculté de médecine, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Centre for Genomic Medicine and Personalized Medicine, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France
| | - A-C Richard
- Inserm, U1079, faculté de médecine, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Centre for Genomic Medicine and Personalized Medicine, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France
| | - J Alexander
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupoli, Greece
| | - P Paschou
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupoli, Greece
| | - C Charbonnier
- Inserm, U1079, faculté de médecine, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Centre for Genomic Medicine and Personalized Medicine, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France
| | - C Bellenguez
- Inserm, U1167, Lille, France.,Institut Pasteur de Lille, Lille, France.,Université Lille-Nord de France, Lille, France
| | - B Grenier-Boley
- Inserm, U1167, Lille, France.,Institut Pasteur de Lille, Lille, France.,Université Lille-Nord de France, Lille, France
| | - D Lechner
- Centre National de Génotypage, Institut de Génomique, CEA, Evry, France
| | - M-T Bihoreau
- Centre National de Génotypage, Institut de Génomique, CEA, Evry, France
| | - R Olaso
- Centre National de Génotypage, Institut de Génomique, CEA, Evry, France
| | - A Boland
- Centre National de Génotypage, Institut de Génomique, CEA, Evry, France
| | - V Meyer
- Centre National de Génotypage, Institut de Génomique, CEA, Evry, France
| | - J-F Deleuze
- Centre National de Génotypage, Institut de Génomique, CEA, Evry, France.,Fondation Jean Dausset, Centre d'études du Polymorphisme Humain, Paris, France
| | - P Amouyel
- Inserm, U1167, Lille, France.,Institut Pasteur de Lille, Lille, France.,Université Lille-Nord de France, Lille, France
| | - H M Munter
- McGill University and Génome Québec Innovation Centre, Montréal, QC, Canada
| | - G Bourque
- McGill University and Génome Québec Innovation Centre, Montréal, QC, Canada
| | - M Lathrop
- McGill University and Génome Québec Innovation Centre, Montréal, QC, Canada
| | - T Frebourg
- Inserm, U1079, faculté de médecine, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Centre for Genomic Medicine and Personalized Medicine, Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France
| | - R Redon
- Inserm, UMR 1087, l'institut du thorax, CHU Nantes, Nantes, France.,CNRS, UMR 6291, Université de Nantes, Nantes, France
| | - L Letenneur
- INSERM, U1219, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - J-F Dartigues
- INSERM, U1219, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - O Martinaud
- CNR-MAJ, Rouen University Hospital, Rouen, France.,Department of Neurology, Rouen University Hospital, Rouen, France
| | - O Kalev
- Institute of Pathology and Neuropathology, Kepler University Hospital, Linz, Austria
| | - S Mehrabian
- Department of Neurology, Alexandrovska University Hospital, Medical University-Sofia, Sofia, Bulgaria
| | - L Traykov
- Department of Neurology, Alexandrovska University Hospital, Medical University-Sofia, Sofia, Bulgaria
| | - T Ströbel
- Institute of Neurology, Medical University Vienna, Vienna, Austria
| | - I Le Ber
- Sorbonne Universités, Inserm, CNRS, UPMC Univ Paris 06, UMR S 1127, Paris, France.,CNR-MAJ, IMMA, département des maladies du système nerveux, Hôpital Pitié-Salpêtrière, Paris, France
| | - P Caroppo
- Sorbonne Universités, Inserm, CNRS, UPMC Univ Paris 06, UMR S 1127, Paris, France.,CNR-MAJ, IMMA, département des maladies du système nerveux, Hôpital Pitié-Salpêtrière, Paris, France
| | - S Epelbaum
- Sorbonne Universités, Inserm, CNRS, UPMC Univ Paris 06, UMR S 1127, Paris, France.,CNR-MAJ, IMMA, département des maladies du système nerveux, Hôpital Pitié-Salpêtrière, Paris, France
| | - T Jonveaux
- Centre Mémoire de Ressources et de Recherche de Lorraine, CHRU Nancy Service de Gériatrie, Hôpital de Brabois, Vandoeuvre les Nancy, France.,Laboratoire INTERPSY, EA 4432, Groupe de recherche sur les Communications (GRC), Université de Lorraine, Psychologie, Nancy, France
| | - F Pasquier
- CNR-MAJ Inserm U1171, Univ Lille, CHU, Lille, France
| | | | - E Génin
- Inserm, UMR1078, CHU Brest, Université Bretagne Occidentale, Brest, France
| | - L Guyant-Maréchal
- Department of Neurology, Rouen University Hospital, Rouen, France.,Department of Neurophysiology, Rouen University Hospital, Rouen, France
| | - G G Kovacs
- Institute of Neurology, Medical University Vienna, Vienna, Austria
| | - J-C Lambert
- Inserm, U1167, Lille, France.,Institut Pasteur de Lille, Lille, France.,Université Lille-Nord de France, Lille, France
| | - D Hannequin
- Inserm, U1079, faculté de médecine, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Centre for Genomic Medicine and Personalized Medicine, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France.,Department of Neurology, Rouen University Hospital, Rouen, France
| | - D Campion
- Inserm, U1079, faculté de médecine, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Centre for Genomic Medicine and Personalized Medicine, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France.,Department of Research, Rouvray Psychiatric Hospital, Sotteville-lès-Rouen, France
| | - A Rovelet-Lecrux
- Inserm, U1079, faculté de médecine, Rouen University, IRIB, Normandy University, Rouen, France.,Normandy Centre for Genomic Medicine and Personalized Medicine, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France
| | | |
Collapse
|
554
|
Smith R, Schöll M, Widner H, van Westen D, Svenningsson P, Hägerström D, Ohlsson T, Jögi J, Nilsson C, Hansson O. In vivo retention of 18F-AV-1451 in corticobasal syndrome. Neurology 2017; 89:845-853. [PMID: 28754841 PMCID: PMC5580862 DOI: 10.1212/wnl.0000000000004264] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 05/30/2017] [Indexed: 12/24/2022] Open
Abstract
Objective: To study the usefulness of 18F-AV-1451 PET in patients with corticobasal syndrome (CBS). Methods: We recruited 8 patients with CBS, 17 controls, 31 patients with Alzheimer disease (AD), and 11 patients with progressive supranuclear palsy (PSP) from the Swedish BioFINDER study. All patients underwent clinical assessment, 18F-AV-1451 PET, MRI, and quantification of β-amyloid pathology. A subset of participants also underwent 18F-FDG-PET. Results: In the 8 patients with CBS, 6 had imaging findings compatible with the corticobasal degeneration pathology and 2 with typical AD pathology. In the 6 patients with CBS without typical AD pathology, there were substantial retentions of 18F-AV-1451 in the motor cortex, corticospinal tract, and basal ganglia contralateral to the most affected body side. These patients could be clearly distinguished from patients with AD dementia or PSP using 18F-AV-1451. However, cortical atrophy was more widespread than the cortical retention of 18F-AV1451 in these CBS cases, and cortical AV-1451 uptake did not correlate with cortical thickness or glucose hypometabolism. These results are in sharp contrast to AD dementia, where 18F-AV-1451 retention was more widespread than cortical atrophy, and correlated well with cortical thickness and hypometabolism. Conclusions: Patients with CBS without typical AD pathology exhibited AV-1451 retention in the motor cortex, corticospinal tract, and basal ganglia contralateral to the affected body side, clearly different from controls and patients with AD dementia or PSP. However, cortical atrophy measured with MRI and decreased 18F-fluorodeoxyglucose uptake were more widespread than 18F-AV-1451 uptake and probably represent earlier, yet less specific, markers of CBS. Classification of evidence: This study provides Class III evidence that 18F-AV-1451 PET distinguishes between CBS and AD or PSP.
Collapse
Affiliation(s)
- Ruben Smith
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden.
| | - Michael Schöll
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Håkan Widner
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Danielle van Westen
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Per Svenningsson
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Douglas Hägerström
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Tomas Ohlsson
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Jonas Jögi
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Christer Nilsson
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
555
|
Tau Isoforms Imbalance Impairs the Axonal Transport of the Amyloid Precursor Protein in Human Neurons. J Neurosci 2017; 37:58-69. [PMID: 28053030 DOI: 10.1523/jneurosci.2305-16.2016] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/21/2016] [Accepted: 11/04/2016] [Indexed: 01/11/2023] Open
Abstract
Tau, as a microtubule (MT)-associated protein, participates in key neuronal functions such as the regulation of MT dynamics, axonal transport, and neurite outgrowth. Alternative splicing of exon 10 in the tau primary transcript gives rise to protein isoforms with three (3R) or four (4R) MT binding repeats. Although tau isoforms are balanced in the normal adult human brain, imbalances in 3R:4R ratio have been tightly associated with the pathogenesis of several neurodegenerative disorders, yet the underlying molecular mechanisms remain elusive. Several studies exploiting tau overexpression and/or mutations suggested that perturbations in tau metabolism impair axonal transport. Nevertheless, no physiological model has yet demonstrated the consequences of altering the endogenous relative content of tau isoforms over axonal transport regulation. Here, we addressed this issue using a trans-splicing strategy that allows modulating tau exon 10 inclusion/exclusion in differentiated human-derived neurons. Upon changes in 3R:4R tau relative content, neurons showed no morphological changes, but live imaging studies revealed that the dynamics of the amyloid precursor protein (APP) were significantly impaired. Single trajectory analyses of the moving vesicles showed that predominance of 3R tau favored the anterograde movement of APP vesicles, increasing anterograde run lengths and reducing retrograde runs and segmental velocities. Conversely, the imbalance toward the 4R isoform promoted a retrograde bias by a significant reduction of anterograde velocities. These findings suggest that changes in 3R:4R tau ratio has an impact on the regulation of axonal transport and specifically in APP dynamics, which might link tau isoform imbalances with APP abnormal metabolism in neurodegenerative processes. SIGNIFICANCE STATEMENT The tau protein has a relevant role in the transport of cargos throughout neurons. Dysfunction in tau metabolism underlies several neurological disorders leading to dementia. In the adult human brain, two tau isoforms are found in equal amounts, whereas changes in such equilibrium have been associated with neurodegenerative diseases. We investigated the role of tau in human neurons in culture and found that perturbations in the endogenous balance of tau isoforms were sufficient to impair the transport of the Alzheimer's disease-related amyloid precursor protein (APP), although neuronal morphology was normal. Our results provide evidence of a direct relationship between tau isoform imbalance and defects in axonal transport, which induce an abnormal APP metabolism with important implications in neurodegeneration.
Collapse
|
556
|
Interaction of misfolded proteins and mitochondria in neurodegenerative disorders. Biochem Soc Trans 2017; 45:1025-1033. [PMID: 28733489 DOI: 10.1042/bst20170024] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/17/2022]
Abstract
The number of the people affected by neurodegenerative disorders is growing dramatically due to the ageing of population. The major neurodegenerative diseases share some common pathological features including the involvement of mitochondria in the mechanism of pathology and misfolding and the accumulation of abnormally aggregated proteins. Neurotoxicity of aggregated β-amyloid, tau, α-synuclein and huntingtin is linked to the effects of these proteins on mitochondria. All these misfolded aggregates affect mitochondrial energy metabolism by inhibiting diverse mitochondrial complexes and limit ATP availability in neurones. β-Amyloid, tau, α-synuclein and huntingtin are shown to be involved in increased production of reactive oxygen species, which can be generated in mitochondria or can target this organelle. Most of these aggregated proteins are capable of deregulating mitochondrial calcium handling that, in combination with oxidative stress, lead to opening of the mitochondrial permeability transition pore. Despite some of the common features, aggregated β-amyloid, tau, α-synuclein and huntingtin have diverse targets in mitochondria that can partially explain neurotoxic effect of these proteins in different brain regions.
Collapse
|
557
|
Tamagnini F, Walsh DA, Brown JT, Bondulich MK, Hanger DP, Randall AD. Hippocampal neurophysiology is modified by a disease-associated C-terminal fragment of tau protein. Neurobiol Aging 2017; 60:44-56. [PMID: 28917666 PMCID: PMC5654728 DOI: 10.1016/j.neurobiolaging.2017.07.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/26/2017] [Accepted: 07/11/2017] [Indexed: 01/09/2023]
Abstract
The accumulation of cleaved tau fragments in the brain is associated with several tauopathies. For this reason, we recently developed a transgenic mouse that selectively accumulates a C-Terminal 35 kDa human tau fragment (Tau35). These animals develop progressive motor and spatial memory impairment, paralleled by increased hippocampal glycogen synthase kinase 3β activity. In this neurophysiological study, we focused on the CA1 subfield of the hippocampus, a brain area involved in memory encoding. The accumulation of Tau35 results in a significant increase of short-term facilitation of the synaptic response in the theta frequency range (10 Hz), without affecting basal synaptic transmission and long-term synaptic plasticity. Tau35 expression also alters the intrinsic excitability of CA1 pyramidal neurons. Thus, Tau35 presence is associated with increased and decreased excitability at hyperpolarized and depolarized potentials, respectively. These observations are paralleled by a hyperpolarization of the voltage-sensitivity of noninactivating K+ currents. Further investigation is needed to assess the causal link between such functional alterations and the cognitive and motor impairments previously observed in this model.
Collapse
Affiliation(s)
- Francesco Tamagnini
- Institute of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK.
| | - Darren A Walsh
- Institute of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Jon T Brown
- Institute of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Marie K Bondulich
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Diane P Hanger
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Andrew D Randall
- Institute of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| |
Collapse
|
558
|
Yang Y, Qin M, Bao P, Xu W, Xu J. Secretory carrier membrane protein 5 is an autophagy inhibitor that promotes the secretion of α-synuclein via exosome. PLoS One 2017; 12:e0180892. [PMID: 28700687 PMCID: PMC5507457 DOI: 10.1371/journal.pone.0180892] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/22/2017] [Indexed: 01/07/2023] Open
Abstract
Autophagy-lysosomal pathway is a cellular protective system to remove aggregated proteins and damaged organelles. Meanwhile, exosome secretion has emerged as a mode to selectively clear the neurotoxic proteins, such as α-synuclein. Mounting evidence suggests that these two cellular processes are coordinated to facilitate the clearance of toxic cellular waste; however the regulators for the transition between these two processes are unclear. Here we show that SCAMP5, a secretory carrier membrane protein significantly induced in the brains of Huntington's disease patients, is quickly and transiently induced by protein stress and autophagic stimulation, and is regulated by the master autophagy transcriptional regulator TFEB. Ironically, SCAMP5 inhibits autophagy flux by blocking the fusion of autophagosomes and lysosomes. Although autophagy is blocked, SCAMP5 does not cause significant protein aggregation in cells. Instead, it promotes the Golgi fragmentation and stimulates the unconventional secretion of the co-localizing α-synuclein via exosome as an exosome component. Therefore, we have identified SCAMP5 as a novel coordinator of autophagy and exosome secretion, which is induced upon protein stress to channel the efficient clearance of toxic proteins via the exosomes rather than autophagy-lysosomal pathway.
Collapse
Affiliation(s)
- Yi Yang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Meiling Qin
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Puhua Bao
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Wangchao Xu
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Jin Xu
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
- * E-mail:
| |
Collapse
|
559
|
Dugger BN, Dickson DW. Pathology of Neurodegenerative Diseases. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a028035. [PMID: 28062563 DOI: 10.1101/cshperspect.a028035] [Citation(s) in RCA: 840] [Impact Index Per Article: 120.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative disorders are characterized by progressive loss of selectively vulnerable populations of neurons, which contrasts with select static neuronal loss because of metabolic or toxic disorders. Neurodegenerative diseases can be classified according to primary clinical features (e.g., dementia, parkinsonism, or motor neuron disease), anatomic distribution of neurodegeneration (e.g., frontotemporal degenerations, extrapyramidal disorders, or spinocerebellar degenerations), or principal molecular abnormality. The most common neurodegenerative disorders are amyloidoses, tauopathies, α-synucleinopathies, and TDP-43 proteinopathies. The protein abnormalities in these disorders have abnormal conformational properties. Growing experimental evidence suggests that abnormal protein conformers may spread from cell to cell along anatomically connected pathways, which may in part explain the specific anatomical patterns observed at autopsy. In this review, we detail the human pathology of select neurodegenerative disorders, focusing on their main protein aggregates.
Collapse
Affiliation(s)
- Brittany N Dugger
- Institute for Neurodegenerative Diseases, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143
| | | |
Collapse
|
560
|
Hall B, Mak E, Cervenka S, Aigbirhio FI, Rowe JB, O’Brien JT. In vivo tau PET imaging in dementia: Pathophysiology, radiotracer quantification, and a systematic review of clinical findings. Ageing Res Rev 2017; 36:50-63. [PMID: 28315409 DOI: 10.1016/j.arr.2017.03.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/15/2017] [Accepted: 03/06/2017] [Indexed: 12/14/2022]
Abstract
In addition to the deposition of β-amyloid plaques, neurofibrillary tangles composed of aggregated hyperphosphorylated tau are one of the pathological hallmarks of Alzheimer's disease and other neurodegenerative disorders. Until now, our understanding about the natural history and topography of tau deposition has only been based on post-mortem and cerebrospinal fluid studies, and evidence continues to implicate tau as a central driver of downstream neurodegenerative processes and cognitive decline. Recently, it has become possible to assess the regional distribution and severity of tau burden in vivo with the development of novel radiotracers for positron emission tomography (PET) imaging. In this article, we provide a comprehensive discussion of tau pathophysiology, its quantification with novel PET radiotracers, as well as a systematic review of tau PET imaging in normal aging and various dementia conditions: mild cognitive impairment, Alzheimer's disease, frontotemporal dementia, progressive supranuclear palsy, and Lewy body dementia. We discuss the main findings in relation to group differences, clinical-cognitive correlations of tau PET, and multi-modal relationships among tau PET and other pathological markers. Collectively, the small but growing literature of tau PET has yielded consistent anatomical patterns of tau accumulation that recapitulate post-mortem distribution of neurofibrillary tangles which correlate with cognitive functions and other markers of pathology. In general, AD is characterised by increased tracer retention in the inferior temporal lobe, extending into the frontal and parietal regions in more severe cases. It is also noted that the spatial topography of tau accumulation is markedly distinct to that of amyloid burden in aging and AD. Tau PET imaging has also revealed characteristic spatial patterns among various non-AD tauopathies, supporting its potential role for differential diagnosis. Finally, we propose novel directions for future tau research, including (a) longitudinal imaging in preclinical dementia, (b) multi-modal mapping of tau pathology onto other pathological processes such as neuroinflammation, and (c) the need for more validation studies against post-mortem samples of the same subjects.
Collapse
|
561
|
Dani M, Brooks D, Edison P. Suspected non-Alzheimer's pathology - Is it non-Alzheimer's or non-amyloid? Ageing Res Rev 2017; 36:20-31. [PMID: 28235659 DOI: 10.1016/j.arr.2017.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/04/2017] [Accepted: 02/16/2017] [Indexed: 01/10/2023]
Abstract
Neurodegeneration, the progressive loss of neurons, is a major process involved in dementia and age-related cognitive impairment. It can be detected clinically using currently available biomarker tests. Suspected Non-Alzheimer Pathology (SNAP) is a biomarker-based concept that encompasses a group of individuals with neurodegeneration, but no evidence of amyloid deposition (thereby distinguishing it from Alzheimer's disease (AD)). These individuals may often have a clinical diagnosis of AD, but their clinical features, genetic susceptibility and progression can differ significantly, carrying crucial implications for precise diagnostics, clinical management, and efficacy of clinical drug trials. SNAP has caused wide interest in the dementia research community, because it is still unclear whether it represents distinct pathology separate from AD, or whether in some individuals, it could represent the earliest stage of AD. This debate has raised pertinent questions about the pathways to AD, the need for biomarkers, and the sensitivity of current biomarker tests. In this review, we discuss the biomarker and imaging trials that first recognized SNAP. We describe the pathological correlates of SNAP and comment on the different causes of neurodegeneration. Finally, we discuss the debate around the concept of SNAP, and further unanswered questions that are emerging.
Collapse
|
562
|
Leyns CEG, Holtzman DM. Glial contributions to neurodegeneration in tauopathies. Mol Neurodegener 2017; 12:50. [PMID: 28662669 PMCID: PMC5492997 DOI: 10.1186/s13024-017-0192-x] [Citation(s) in RCA: 270] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/20/2017] [Indexed: 01/09/2023] Open
Abstract
Tauopathies are a broad set of neurodegenerative dementias characterized by aggregation of the tau protein into filamentous inclusions that can be found in neurons and glial cells. Activated microglia, astrocytes and elevated levels of proinflammatory molecules are also pathological hallmarks that are found in brain regions affected by tau pathology. There has been abundant research in recent years to understand the role of gliosis and neuroinflammation in neurodegenerative diseases, particularly in Alzheimer's disease (AD) which is the most common form of dementia. AD is a tauopathy characterized by both extracellular amyloid-β plaques in addition to intracellular neurofibrillary tangles and neuropil threads containing aggregated tau protein. Accumulating evidence suggests that neuroinflammation offers a possible mechanistic link between these pathologies. Additionally, there appears to be a role for neuroinflammation in aggravating tau pathology and neurodegeneration in tauopathies featuring tau deposits as the predominant pathological signature. In this review, we survey the literature regarding inflammatory mechanisms that may impact neurodegeneration in AD and related tauopathies. We consider a physical role for microglia in the spread of tau pathology as well as the non-cell autonomous effects of secreted proinflammatory cytokines, specifically interleukin 1 beta, interleukin 6, tumor necrosis factor alpha and complement proteins. These molecules appear to have direct effects on tau pathophysiology and overall neuronal health. They also indirectly impact neuronal homeostasis by altering glial function. We conclude by proposing a complex role for gliosis and neuroinflammation in accelerating the progression of AD and other tauopathies.
Collapse
Affiliation(s)
- Cheryl E. G. Leyns
- Department of Neurology, Washington University, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, 660 S. Euclid Ave, St. Louis, MO 63110 USA
| | - David M. Holtzman
- Department of Neurology, Washington University, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, 660 S. Euclid Ave, St. Louis, MO 63110 USA
| |
Collapse
|
563
|
Coakeley S, Strafella AP. Imaging tau pathology in Parkinsonisms. NPJ Parkinsons Dis 2017; 3:22. [PMID: 28685158 PMCID: PMC5491530 DOI: 10.1038/s41531-017-0023-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 12/23/2022] Open
Abstract
The recent development of positron emission tomography radiotracers targeting pathological tau in vivo has led to numerous human trials. While investigations have primarily focused on the most common tauopathy, Alzheimer's disease, it is imperative that testing also be performed in parkinsonian tauopathies, such as progressive supranuclear palsy, corticobasal degeneration, and frontotemporal dementia and parkinsonism linked to chromosome 17. Tau aggregates differ in isoforms and conformations across disorders, and as a result one radiotracer may not be appropriate for all tauopathies. In this review, we evaluate the preclinical and clinical reports of current tau radiotracers in parkinsonian disorders. These radiotracers include [18F]FDDNP, [11C]PBB3, [18F]THK-5317, [18F]THK-5351, and [18F]AV-1451 ([18F]T807). There are concerns of off-target binding with [18F]FDDNP and [11C]PBB3, which may increase the signal to noise ratio and thereby decrease the efficacy of these radiotracers. Testing in [18F]THK-5317, [18F]THK-5351, and [18F]AV-1451 has been performed in progressive supranuclear palsy, while [18F]THK-5317 and [18F]AV-1451 have also been tested in corticobasal degeneration patients. [18F]THK-5317 and [18F]THK-5351 have demonstrated binding in brain regions known to be afflicted with pathological tau; however, due to small sample sizes these studies should be replicated before concluding their appropriateness in parkinsonian tauopathies. [18F]AV-1451 has demonstrated mixed results in progressive supranuclear palsy patients and post-mortem analysis shows minimal to no binding to non-Alzheimer's disease tauopathies brain slices.
Collapse
Affiliation(s)
- Sarah Coakeley
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON Canada
- Division of Brain, Imaging and Behaviour—Systems Neuroscience, Krembil Research Institute, UHN, University of Toronto, Toronto, ON Canada
| | - Antonio P. Strafella
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON Canada
- Division of Brain, Imaging and Behaviour—Systems Neuroscience, Krembil Research Institute, UHN, University of Toronto, Toronto, ON Canada
- Morton and Gloria Shulman Movement Disorder Unit and E.J. Safra Parkinson Disease Program, Neurology Division, Dept. of Medicine, Toronto Western Hospital, UHN, University of Toronto, Toronto, ON Canada
| |
Collapse
|
564
|
Chemerovski‐Glikman M, Frenkel‐Pinter M, Mdah R, Abu‐Mokh A, Gazit E, Segal D. Inhibition of the Aggregation and Toxicity of the Minimal Amyloidogenic Fragment of Tau by Its Pro‐Substituted Analogues. Chemistry 2017; 23:9618-9624. [DOI: 10.1002/chem.201701218] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Marina Chemerovski‐Glikman
- Department of Molecular Microbiology & BiotechnologySagol Interdisciplinary School of NeurosciencesGeorge S. Wise Faculty of Life SciencesTel-Aviv University Tel-Aviv 69978 Israel
| | - Moran Frenkel‐Pinter
- Department of Molecular Microbiology & BiotechnologySagol Interdisciplinary School of NeurosciencesGeorge S. Wise Faculty of Life SciencesTel-Aviv University Tel-Aviv 69978 Israel
| | - Ragad Mdah
- Department of Molecular Microbiology & BiotechnologySagol Interdisciplinary School of NeurosciencesGeorge S. Wise Faculty of Life SciencesTel-Aviv University Tel-Aviv 69978 Israel
| | - Amjaad Abu‐Mokh
- Department of Molecular Microbiology & BiotechnologySagol Interdisciplinary School of NeurosciencesGeorge S. Wise Faculty of Life SciencesTel-Aviv University Tel-Aviv 69978 Israel
| | - Ehud Gazit
- Department of Molecular Microbiology & BiotechnologySagol Interdisciplinary School of NeurosciencesGeorge S. Wise Faculty of Life SciencesTel-Aviv University Tel-Aviv 69978 Israel
- Department of Materials Science and EngineeringIby and Aladar Fleischman Faculty of EngineeringTel Aviv University Tel Aviv 6997801 Israel
| | - Daniel Segal
- Department of Molecular Microbiology & BiotechnologySagol Interdisciplinary School of NeurosciencesGeorge S. Wise Faculty of Life SciencesTel-Aviv University Tel-Aviv 69978 Israel
| |
Collapse
|
565
|
Abstract
Intracellular accumulation of abnormally phosphorylated tau in different types of neurons is a pathological characteristic of Alzheimer's disease (AD). While tau modification and associated neuronal loss and hypometabolism start in the entorhinal cortex (EC) in early AD patients, the mechanism by which mutant P301L hTau leads to dementia is not fully elucidated. Here, we studied the effects of P301L hTau transduction in the medial EC (MEC) of mice on tau phosphorylation and accumulation, and cognitive deficit. We found that the exogenous mutant tau protein was restricted in MEC without spreading to other brain regions at one month after transduction. Interestingly, expression of the mutant tau in MEC induces endogenous tau hyperphosphorylation and accumulation in hippocampus and cortex, and inhibits neuronal activity with attenuated PP-DG synapse plasticity, leading to hippocampus-dependent memory deficit with intact olfactory function. These findings suggest a novel neuropathological mechanism of early AD, which is initiated by tau accumulation in MEC, and demonstrate a tau pathological model of early stage AD.
Collapse
|
566
|
Chiti F, Dobson CM. Protein Misfolding, Amyloid Formation, and Human Disease: A Summary of Progress Over the Last Decade. Annu Rev Biochem 2017; 86:27-68. [DOI: 10.1146/annurev-biochem-061516-045115] [Citation(s) in RCA: 1709] [Impact Index Per Article: 244.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Peptides and proteins have been found to possess an inherent tendency to convert from their native functional states into intractable amyloid aggregates. This phenomenon is associated with a range of increasingly common human disorders, including Alzheimer and Parkinson diseases, type II diabetes, and a number of systemic amyloidoses. In this review, we describe this field of science with particular reference to the advances that have been made over the last decade in our understanding of its fundamental nature and consequences. We list the proteins that are known to be deposited as amyloid or other types of aggregates in human tissues and the disorders with which they are associated, as well as the proteins that exploit the amyloid motif to play specific functional roles in humans. In addition, we summarize the genetic factors that have provided insight into the mechanisms of disease onset. We describe recent advances in our knowledge of the structures of amyloid fibrils and their oligomeric precursors and of the mechanisms by which they are formed and proliferate to generate cellular dysfunction. We show evidence that a complex proteostasis network actively combats protein aggregation and that such an efficient system can fail in some circumstances and give rise to disease. Finally, we anticipate the development of novel therapeutic strategies with which to prevent or treat these highly debilitating and currently incurable conditions.
Collapse
Affiliation(s)
- Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” Section of Biochemistry, Università di Firenze, 50134 Firenze, Italy
| | - Christopher M. Dobson
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
567
|
Hock EM, Polymenidou M. Prion-like propagation as a pathogenic principle in frontotemporal dementia. J Neurochem 2017; 138 Suppl 1:163-83. [PMID: 27502124 PMCID: PMC6680357 DOI: 10.1111/jnc.13668] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/22/2016] [Accepted: 05/11/2016] [Indexed: 12/12/2022]
Abstract
Frontotemporal dementia is a devastating neurodegenerative disease causing stark alterations in personality and language. Characterized by severe atrophy of the frontal and temporal brain lobes, frontotemporal dementia (FTD) shows extreme heterogeneity in clinical presentation, genetic causes, and pathological findings. Like most neurodegenerative diseases, the initial symptoms of FTD are subtle, but increase in severity over time, as the disease progresses. Clinical progression is paralleled by exacerbation of pathological findings and the involvement of broader brain regions, which currently lack mechanistic explanation. Yet, a flurry of studies indicate that protein aggregates accumulating in neurodegenerative diseases can act as propagating entities, amplifying their pathogenic conformation, in a way similar to infectious prions. In this prion‐centric view, FTD can be divided into three subtypes, TDP‐43 or FUS proteinopathy and tauopathy. Here, we review the current evidence that FTD‐linked pathology propagates in a prion‐like manner and discuss the implications of these findings for disease progression and heterogeneity.
Frontotemporal dementia (FTD) is a progressive neurodegenerative disease causing severe personality dysfunctions, characterized by profound heterogeneity. Accumulation of tau, TDP‐43 or FUS cytoplasmic aggregates characterize molecularly distinct and non‐overlapping FTD subtypes. Here, we discuss the current evidence suggesting that prion‐like propagation and cell‐to‐cell spread of each of these cytoplasmic aggregates may underlie disease progression and heterogeneity.
This article is part of the Frontotemporal Dementia special issue.
Collapse
Affiliation(s)
- Eva-Maria Hock
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
568
|
Choi Y, Ha S, Lee YS, Kim YK, Lee DS, Kim DJ. Development of tau PET Imaging Ligands and their Utility in Preclinical and Clinical Studies. Nucl Med Mol Imaging 2017; 52:24-30. [PMID: 29391909 DOI: 10.1007/s13139-017-0484-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/10/2017] [Accepted: 05/22/2017] [Indexed: 12/16/2022] Open
Abstract
The pathological features of Alzheimer's disease are senile plaques which are aggregates of β-amyloid peptides and neurofibrillary tangles in the brain. Neurofibrillary tangles are aggregates of hyperphosphorylated tau proteins, and these induce various other neurodegenerative diseases, such as progressive supranuclear palsy, corticobasal degeneration, frontotemporal lobar degeneration, frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17), and chronic traumatic encephalopathy. In the case of Alzheimer's disease, the measurement of neurofibrillary tangles associated with cognitive decline is suitable for differential diagnosis, disease progression assessment, and to monitor the effects of therapeutic treatment. This review discusses considerations for the development of tau ligands for imaging and summarizes the results of the first-in-human and preclinical studies of the tau tracers that have been developed thus far. The development of tau ligands for imaging studies will be helpful for differential diagnosis and for the development of therapeutic treatments for tauopathies including Alzheimer's disease.
Collapse
Affiliation(s)
- Yoori Choi
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,2Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744 South Korea
| | - Seunggyun Ha
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,3Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, 03080, 103 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Yun-Sang Lee
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,3Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, 03080, 103 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Yun Kyung Kim
- 4Institute of Brain Science, Korean Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 136-791 South Korea
| | - Dong Soo Lee
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,2Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744 South Korea.,3Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, 03080, 103 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Dong Jin Kim
- 4Institute of Brain Science, Korean Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 136-791 South Korea
| |
Collapse
|
569
|
De Conti L, Borroni B, Baralle M. New routes in frontotemporal dementia drug discovery. Expert Opin Drug Discov 2017; 12:659-671. [DOI: 10.1080/17460441.2017.1329294] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Laura De Conti
- Biotechnology Development Group, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders - Neurology Unit, University of Brescia, Brescia, Italy
| | - Marco Baralle
- Biotechnology Development Group, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| |
Collapse
|
570
|
Functional screening of Alzheimer risk loci identifies PTK2B as an in vivo modulator and early marker of Tau pathology. Mol Psychiatry 2017; 22:874-883. [PMID: 27113998 PMCID: PMC5444024 DOI: 10.1038/mp.2016.59] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/12/2015] [Accepted: 01/20/2016] [Indexed: 01/02/2023]
Abstract
A recent genome-wide association meta-analysis for Alzheimer's disease (AD) identified 19 risk loci (in addition to APOE) in which the functional genes are unknown. Using Drosophila, we screened 296 constructs targeting orthologs of 54 candidate risk genes within these loci for their ability to modify Tau neurotoxicity by quantifying the size of >6000 eyes. Besides Drosophila Amph (ortholog of BIN1), which we previously implicated in Tau pathology, we identified p130CAS (CASS4), Eph (EPHA1), Fak (PTK2B) and Rab3-GEF (MADD) as Tau toxicity modulators. Of these, the focal adhesion kinase Fak behaved as a strong Tau toxicity suppressor in both the eye and an independent focal adhesion-related wing blister assay. Accordingly, the human Tau and PTK2B proteins biochemically interacted in vitro and PTK2B co-localized with hyperphosphorylated and oligomeric Tau in progressive pathological stages in the brains of AD patients and transgenic Tau mice. These data indicate that PTK2B acts as an early marker and in vivo modulator of Tau toxicity.
Collapse
|
571
|
Hastings NB, Wang X, Song L, Butts BD, Grotz D, Hargreaves R, Fred Hess J, Hong KLK, Huang CRR, Hyde L, Laverty M, Lee J, Levitan D, Lu SX, Maguire M, Mahadomrongkul V, McEachern EJ, Ouyang X, Rosahl TW, Selnick H, Stanton M, Terracina G, Vocadlo DJ, Wang G, Duffy JL, Parker EM, Zhang L. Inhibition of O-GlcNAcase leads to elevation of O-GlcNAc tau and reduction of tauopathy and cerebrospinal fluid tau in rTg4510 mice. Mol Neurodegener 2017; 12:39. [PMID: 28521765 PMCID: PMC5437664 DOI: 10.1186/s13024-017-0181-0] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 05/09/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Hyperphosphorylation of microtubule-associated protein tau is a distinct feature of neurofibrillary tangles (NFTs) that are the hallmark of neurodegenerative tauopathies. O-GlcNAcylation is a lesser known post-translational modification of tau that involves the addition of N-acetylglucosamine onto serine and threonine residues. Inhibition of O-GlcNAcase (OGA), the enzyme responsible for the removal of O-GlcNAc modification, has been shown to reduce tau pathology in several transgenic models. Clarifying the underlying mechanism by which OGA inhibition leads to the reduction of pathological tau and identifying translatable measures to guide human dosing and efficacy determination would significantly facilitate the clinical development of OGA inhibitors for the treatment of tauopathies. METHODS Genetic and pharmacological approaches are used to evaluate the pharmacodynamic response of OGA inhibition. A panel of quantitative biochemical assays is established to assess the effect of OGA inhibition on pathological tau reduction. A "click" chemistry labeling method is developed for the detection of O-GlcNAcylated tau. RESULTS Substantial (>80%) OGA inhibition is required to observe a measurable increase in O-GlcNAcylated proteins in the brain. Sustained and substantial OGA inhibition via chronic treatment with Thiamet G leads to a significant reduction of aggregated tau and several phosphorylated tau species in the insoluble fraction of rTg4510 mouse brain and total tau in cerebrospinal fluid (CSF). O-GlcNAcylated tau is elevated by Thiamet G treatment and is found primarily in the soluble 55 kD tau species, but not in the insoluble 64 kD tau species thought as the pathological entity. CONCLUSION The present study demonstrates that chronic inhibition of OGA reduces pathological tau in the brain and total tau in the CSF of rTg4510 mice, most likely by directly increasing O-GlcNAcylation of tau and thereby maintaining tau in the soluble, non-toxic form by reducing tau aggregation and the accompanying panoply of deleterious post-translational modifications. These results clarify some conflicting observations regarding the effects and mechanism of OGA inhibition on tau pathology, provide pharmacodynamic tools to guide human dosing and identify CSF total tau as a potential translational biomarker. Therefore, this study provides additional support to develop OGA inhibitors as a treatment for Alzheimer's disease and other neurodegenerative tauopathies.
Collapse
Affiliation(s)
| | - Xiaohai Wang
- Department of In Vivo Pharmacology, West Point, PA USA
| | - Lixin Song
- Department of Neuroscience, Kenilworth, NJ USA
| | | | - Diane Grotz
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Kenilworth, NJ USA
| | | | | | | | | | - Lynn Hyde
- Department of Neuroscience, Kenilworth, NJ USA
| | | | - Julie Lee
- Department of Neuroscience, Kenilworth, NJ USA
| | - Diane Levitan
- Department of Molecular Biomarkers, Kenilworth, NJ USA
| | | | | | | | | | | | | | - Harold Selnick
- Discovery Chemistry, Merck Research Laboratories, West Point, PA USA
| | | | | | | | | | - Joseph L. Duffy
- Discovery Chemistry Merck Research Laboratories, Kenilworth, NJ USA
| | | | - Lili Zhang
- Department of Neuroscience, Kenilworth, NJ USA
| |
Collapse
|
572
|
Ultrasensitive and selective detection of 3-repeat tau seeding activity in Pick disease brain and cerebrospinal fluid. Acta Neuropathol 2017; 133:751-765. [PMID: 28293793 DOI: 10.1007/s00401-017-1692-z] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 10/20/2022]
Abstract
The diagnosis and treatment of diseases involving tau-based pathology such as Alzheimer disease and certain frontotemporal dementias is hampered by the inability to detect pathological forms of tau with sufficient sensitivity, specificity and practicality. In these neurodegenerative diseases, tau accumulates in self-seeding filaments. For example, Pick disease (PiD) is associated with frontotemporal degeneration and accumulation of 3-repeat (3R) tau isoforms in filaments constituting Pick bodies. Exploiting the self-seeding activity of tau deposits, and using a 3R tau fragment as a substrate, we have developed an assay (tau RT-QuIC) that can detect tau seeds in 2 µl aliquots of PiD brain dilutions down to 10-7-10-9. PiD seeding activities were 100-fold higher in frontal and temporal lobes compared to cerebellar cortex. Strikingly, this test was 103- to 105-fold less responsive when seeded with brain containing predominant 4-repeat (4R) tau aggregates from cases of corticobasal degeneration, argyrophilic grain disease, and progressive supranuclear palsy. Alzheimer disease brain, with 3R + 4R tau deposits, also gave much weaker responses than PiD brain. When applied to cerebrospinal fluid samples (5 µl), tau RT-QuIC analyses discriminated PiD from non-PiD cases. These findings demonstrate that abnormal tau aggregates can be detected with high sensitivity and disease-specificity in crude tissue and fluid samples. Accordingly, this tau RT-QuIC assay exemplifies a new approach to diagnosing tauopathies and monitoring therapeutic trials using aggregated tau itself as a biomarker.
Collapse
|
573
|
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the most common cause of dementia among the elderly population. The good correlation between the density and neocortical spread of neurofibrillary tangles (NFTs) and the severity of cognitive impairment offers an opportunity to use a noninvasive imaging technique such as positron emission tomography (PET) for early diagnosis and staging of the disease. PET imaging of NFTs holds promise not only as a diagnostic tool but also because it may enable the development of disease-modifying therapeutics for AD. In this review, we focus on the structural diversity of tau PET tracers, the challenges related to identifying high-affinity and highly selective NFT ligands, and recent progress in the clinical development of tau PET radioligands.
Collapse
Affiliation(s)
- Hartmuth C Kolb
- Janssen Research and Development, Neuroscience Biomarkers, San Diego, California 92121
| | - José Ignacio Andrés
- Janssen Research and Development, Discovery Sciences, Janssen-Cilag S.A., 45007 Toledo, Spain
| |
Collapse
|
574
|
Abstract
Most age-related neurodegenerative diseases are associated with the misfolding and aberrant accumulation of specific proteins in the nervous system. The proteins self-assemble and spread by a prion-like process of corruptive molecular templating, whereby abnormally folded proteins induce the misfolding and aggregation of like proteins into characteristic lesions. Despite the apparent simplicity of this process at the molecular level, diseases such as Alzheimer's, Parkinson's, Creutzfeldt-Jakob, and others display remarkable phenotypic heterogeneity, both clinically and pathologically. Evidence is growing that this variability is mediated, at least in part, by the acquisition of diverse molecular architectures by the misfolded proteins, variants referred to as proteopathic strains. The structural and functional diversity of the assemblies is influenced by genetic, epigenetic, and local contextual factors. Insights into proteopathic strains gleaned from the classical prion diseases can be profitably incorporated into research on other neurodegenerative diseases. Their potentially wide-ranging influence on disease phenotype also suggests that proteopathic strains should be considered in the design and interpretation of diagnostic and therapeutic approaches to these disorders.
Collapse
Affiliation(s)
- Lary C Walker
- Department of Neurology and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30322;
| |
Collapse
|
575
|
Brady ST, Morfini GA. Regulation of motor proteins, axonal transport deficits and adult-onset neurodegenerative diseases. Neurobiol Dis 2017; 105:273-282. [PMID: 28411118 DOI: 10.1016/j.nbd.2017.04.010] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/17/2017] [Accepted: 04/10/2017] [Indexed: 01/07/2023] Open
Abstract
Neurons affected in a wide variety of unrelated adult-onset neurodegenerative diseases (AONDs) typically exhibit a "dying back" pattern of degeneration, which is characterized by early deficits in synaptic function and neuritic pathology long before neuronal cell death. Consistent with this observation, multiple unrelated AONDs including Alzheimer's disease, Parkinson's disease, Huntington's disease, and several motor neuron diseases feature early alterations in kinase-based signaling pathways associated with deficits in axonal transport (AT), a complex cellular process involving multiple intracellular trafficking events powered by microtubule-based motor proteins. These pathogenic events have important therapeutic implications, suggesting that a focus on preservation of neuronal connections may be more effective to treat AONDs than addressing neuronal cell death. While the molecular mechanisms underlying AT abnormalities in AONDs are still being analyzed, evidence has accumulated linking those to a well-established pathological hallmark of multiple AONDs: altered patterns of neuronal protein phosphorylation. Here, we present a short overview on the biochemical heterogeneity of major motor proteins for AT, their regulation by protein kinases, and evidence revealing cell type-specific AT specializations. When considered together, these findings may help explain how independent pathogenic pathways can affect AT differentially in the context of each AOND.
Collapse
Affiliation(s)
- Scott T Brady
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA; Marine Biological Laboratory, Woods Hole, MA 02543, USA.
| | - Gerardo A Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA; Marine Biological Laboratory, Woods Hole, MA 02543, USA.
| |
Collapse
|
576
|
Hinz FI, Geschwind DH. Molecular Genetics of Neurodegenerative Dementias. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a023705. [PMID: 27940516 DOI: 10.1101/cshperspect.a023705] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative dementias are clinically heterogeneous, progressive diseases with frequently overlapping symptoms, such as cognitive impairments and behavior and movement deficits. Although a majority of cases appear to be sporadic, there is a large genetic component that has yet to be fully explained. Here, we review the recent genetic and genomic findings pertaining to Alzheimer's disease, frontotemporal dementia, Lewy body dementia, and prion dementia. In this review, we describe causal and susceptibility genes identified for these dementias and discuss recent research pertaining to the molecular function of these genes. Of particular interest, there is a large overlap in clinical phenotypes, genes, and/or aggregating protein products involved in these diseases, as well as frequent comorbid presentation, indicating that these dementias may represent a continuum of syndromes rather than individual diseases.
Collapse
Affiliation(s)
- Flora I Hinz
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095.,Center for Autism Research and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California 90024
| |
Collapse
|
577
|
Smith VD, Bachstetter AD, Ighodaro E, Roberts K, Abner EL, Fardo DW, Nelson PT. Overlapping but distinct TDP-43 and tau pathologic patterns in aged hippocampi. Brain Pathol 2017; 28:264-273. [PMID: 28281308 DOI: 10.1111/bpa.12505] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/06/2017] [Indexed: 01/06/2023] Open
Abstract
Intracellular proteinaceous aggregates (inclusion bodies) are almost always detectable at autopsy in brains of elderly individuals. Inclusion bodies composed of TDP-43 and tau proteins often coexist in the same brain, and each of these pathologic biomarkers is associated independently with cognitive impairment. However, uncertainties remain about how the presence and neuroanatomical distribution of inclusion bodies correlate with underlying diseases including Alzheimer's disease (AD). To address this knowledge gap, we analyzed data from the University of Kentucky AD Center autopsy series (n = 247); none of the brains had frontotemporal lobar degeneration. A specific question for this study was whether neurofibrillary tangle (NFT) pathology outside of the Braak NFT staging scheme is characteristic of brains with TDP-43 pathology but lacking AD, that is those with cerebral age-related TDP-43 with sclerosis (CARTS). We also tested whether TDP-43 pathology is associated with comorbid AD pathology, and whether argyrophilic grains are relatively likely to be present in cases with, vs. without, TDP-43 pathology. Consistent with prior studies, hippocampal TDP-43 pathology was associated with advanced AD - Braak NFT stages V/VI. However, argyrophilic grain pathology was not more common in cases with TDP-43 pathology in this data set. In brains with CARTS (TDP-43[+]/AD[-] cases), there were more NFTs in dentate granule neurons than were seen in TDP-43[-]/AD[-] cases. These dentate granule cell NFTs could provide a proxy indicator of CARTS pathology in cases lacking substantial AD pathology. Immunofluorescent experiments in a subsample of cases found that, in both advanced AD and CARTS, approximately 1% of dentate granule neurons were PHF-1 immunopositive, whereas ∼25% of TDP-43 positive cells showed colocalized PHF-1 immunoreactivity. We conclude that NFTs in hippocampal dentate granule neurons are often present in CARTS, and TDP-43 pathology may be secondary to or occurring in parallel with tauopathy.
Collapse
Affiliation(s)
- Vanessa D Smith
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY
| | - Adam D Bachstetter
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY.,Department of Neuroscience, University of Kentucky, Lexington, KY
| | - Eseosa Ighodaro
- Department of Neuroscience, University of Kentucky, Lexington, KY.,Sanders Brown Center on Aging, University of Kentucky, Lexington, KY
| | - Kelly Roberts
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY
| | - Erin L Abner
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY.,Department of Epidemiology, University of Kentucky, Lexington, KY
| | - David W Fardo
- Department of Biostatistics, University of Kentucky, Lexington, KY
| | - Peter T Nelson
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY.,Department of Neuroscience, University of Kentucky, Lexington, KY.,Sanders Brown Center on Aging, University of Kentucky, Lexington, KY
| |
Collapse
|
578
|
Lucato CM, Lupton CJ, Halls ML, Ellisdon AM. Amyloidogenicity at a Distance: How Distal Protein Regions Modulate Aggregation in Disease. J Mol Biol 2017; 429:1289-1304. [PMID: 28342736 DOI: 10.1016/j.jmb.2017.03.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/12/2017] [Accepted: 03/14/2017] [Indexed: 12/14/2022]
Abstract
The misfolding of proteins to form amyloid is a key pathological feature of several progressive, and currently incurable, diseases. A mechanistic understanding of the pathway from soluble, native protein to insoluble amyloid is crucial for therapeutic design, and recent efforts have helped to elucidate the key molecular events that trigger protein misfolding. Generally, either global or local structural perturbations occur early in amyloidogenesis to expose aggregation-prone regions of the protein that can then self-associate to form toxic oligomers. Surprisingly, these initiating structural changes are often caused or influenced by protein regions distal to the classically amyloidogenic sequences. Understanding the importance of these distal regions in the pathogenic process has highlighted many remaining knowledge gaps regarding the precise molecular events that occur in classic aggregation pathways. In this review, we discuss how these distal regions can influence aggregation in disease and the recent technical and conceptual advances that have allowed this insight.
Collapse
Affiliation(s)
- Christina M Lucato
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Christopher J Lupton
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Andrew M Ellisdon
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
579
|
Millan MJ. Linking deregulation of non-coding RNA to the core pathophysiology of Alzheimer's disease: An integrative review. Prog Neurobiol 2017; 156:1-68. [PMID: 28322921 DOI: 10.1016/j.pneurobio.2017.03.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 02/06/2023]
Abstract
The human genome encodes a vast repertoire of protein non-coding RNAs (ncRNA), some specific to the brain. MicroRNAs, which interfere with the translation of target mRNAs, are of particular interest since their deregulation has been implicated in neurodegenerative disorders like Alzheimer's disease (AD). However, it remains challenging to link the complex body of observations on miRNAs and AD into a coherent framework. Using extensive graphical support, this article discusses how a diverse panoply of miRNAs convergently and divergently impact (and are impacted by) core pathophysiological processes underlying AD: neuroinflammation and oxidative stress; aberrant generation of β-amyloid-42 (Aβ42); anomalies in the production, cleavage and post-translational marking of Tau; impaired clearance of Aβ42 and Tau; perturbation of axonal organisation; disruption of synaptic plasticity; endoplasmic reticulum stress and the unfolded protein response; mitochondrial dysfunction; aberrant induction of cell cycle re-entry; and apoptotic loss of neurons. Intriguingly, some classes of miRNA provoke these cellular anomalies, whereas others act in a counter-regulatory, protective mode. Moreover, changes in levels of certain species of miRNA are a consequence of the above-mentioned anomalies. In addition to miRNAs, circular RNAs, piRNAs, long non-coding RNAs and other types of ncRNA are being increasingly implicated in AD. Overall, a complex mesh of deregulated and multi-tasking ncRNAs reciprocally interacts with core pathophysiological mechanisms underlying AD. Alterations in ncRNAs can be detected in CSF and the circulation as well as the brain and are showing promise as biomarkers, with the ultimate goal clinical exploitation as targets for novel modes of symptomatic and course-altering therapy.
Collapse
Affiliation(s)
- Mark J Millan
- Centre for Therapeutic Innovation in Neuropsychiatry, institut de recherche Servier, 125 chemin de ronde, 78290 Croissy sur Seine, France.
| |
Collapse
|
580
|
The bridging integrator 1 Gene rs7561528 polymorphism contributes to Alzheimer's disease susceptibility in East Asian and Caucasian populations. Clin Chim Acta 2017; 469:13-21. [PMID: 28302384 DOI: 10.1016/j.cca.2017.03.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/10/2017] [Accepted: 03/11/2017] [Indexed: 01/16/2023]
Abstract
Genetic variants of the bridging integrator 1 (BIN1) at the rs7561528 single nucleotide polymorphism were implicated in increased risk of Alzheimer's disease in several case-control association studies. However, the studies have reported apparently conflicting results. Here, we searched the PubMed and Google Scholar databases. In total, 17,179 AD patients and 17,448 healthy controls (HCs) from 18 studies are included in the current study to examine the association between this polymorphism and AD risk. Significant associations of the SNP rs242557 with AD are found under allelic [A vs. G: odds ratio (OR)=0.86, 95% confidence interval (CI)=0.78, 0.96, P=0.006], dominant (AA+AG vs. GG: OR=0.87, 95% CI=0.77, 0.97, P=0.01), recessive (AA vs. AG+GG: OR=0.86, 95% CI=0.76, 0.98, P=0.21), homozygous (AA vs. GG: OR=0.86, 95% CI=0.76, 0.99, P=0.03) and heterozygous (AG vs. GG: OR=0.87, 95% CI=0.83, 0.92, P<0.00001) models in the pooled populations, under allelic (OR=0.77, 95% CI=0.65, 0.91, P=0.002), dominant (OR=0.75, 95% CI=0.63, 0.90, P=0.001) and heterozygous (OR =0.79, 95% CI=0.70, 0.88, P<0.0001) models in East Asian population, under heterozygous (OR=0.89, 95% CI=0.84, 0.94, P<0.0001) model in Caucasian population. The results of the current meta-analysis suggest that the rs7561528 A allele carriers may be a protective factor against susceptibility to AD under all the genetic models in the pooled populations and under allelic and dominant model in East Asian population, and individuals with A/G heterozygous genotype are not prone to suffer from AD in both Asians and Caucasians.
Collapse
|
581
|
Esteras N, Rohrer JD, Hardy J, Wray S, Abramov AY. Mitochondrial hyperpolarization in iPSC-derived neurons from patients of FTDP-17 with 10+16 MAPT mutation leads to oxidative stress and neurodegeneration. Redox Biol 2017; 12:410-422. [PMID: 28319892 PMCID: PMC5357682 DOI: 10.1016/j.redox.2017.03.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/03/2017] [Accepted: 03/07/2017] [Indexed: 12/25/2022] Open
Abstract
Tau protein inclusions are a frequent hallmark of a variety of neurodegenerative disorders. The 10+16 intronic mutation in MAPT gene, encoding tau, causes frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17), by altering the splicing of the gene and inducing an increase in the production of 4R tau isoforms, which are more prone to aggregation. However, the molecular mechanisms linking increased 4R tau to neurodegeneration are not well understood. Here, we have used iPSC-derived neurons from patients of FTDP-17 carrying the 10+16 mutation to study the molecular mechanisms underlying neurodegeneration. We show that mitochondrial function is altered in the neurons of the patients. We found that FTDP-17 neurons present an increased mitochondrial membrane potential, which is partially maintained by the F1Fo ATPase working in reverse mode. The 10+16 MAPT mutation is also associated with lower mitochondrial NADH levels, partially supressed complex I-driven respiration, and lower ATP production by oxidative phosphorylation, with cells relying on glycolysis to maintain ATP levels. Increased mitochondrial membrane potential in FTDP-17 neurons leads to overproduction of the ROS in mitochondria, which in turn causes oxidative stress and cell death. Mitochondrial ROS overproduction in these cells is a major trigger for neuronal cell death and can be prevented by mitochondrial antioxidants.
Collapse
Affiliation(s)
- Noemí Esteras
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK.
| | | | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Selina Wray
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Andrey Y Abramov
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK.
| |
Collapse
|
582
|
|
583
|
Niemelä V, Landtblom AM, Blennow K, Sundblom J. Tau or neurofilament light-Which is the more suitable biomarker for Huntington's disease? PLoS One 2017; 12:e0172762. [PMID: 28241046 PMCID: PMC5328385 DOI: 10.1371/journal.pone.0172762] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/09/2017] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Previous studies have suggested cerebrospinal fluid (CSF) levels of neurofilament light (NFL) and total tau are elevated in Huntington's disease (HD) and may be used as markers of disease stage. Biomarkers are needed due to the slow disease progression and the limitations of clinical assessment. This study aims to validate the role of NFL and tau as biomarkers in HD. METHODS CSF was obtained from a cohort of HD patients and premanifest HD-mutation carriers. Unified Huntington's Disease Rating Scale (UHDRS) testing was performed on all subjects at the time of sampling. NFL and tau concentrations were determined by ELISA. Spearman correlations were calculated with R version 3.2.3. RESULTS 11 premanifest HD and 12 manifest HD subjects were enrolled. NFL and tau levels were correlated. NFL showed strong correlations with all items included in the clinical assessment (for example the total functional capacity (TFC) (r = - 0.70 p < 0.01) and total motor score (TMS) (r = 0.83p < 0.01). Tau showed slightly weaker correlations (eg. TMS (r = 0.67 p < 0.01); TFC (r = - 0.59 p < 0.01)). NFL was significantly correlated with 5-year probability of disease onset, whereas tau was not. CONCLUSION This study strengthens the case for NFL as a useful biomarker of disease stage. NFL was strongly correlated to all evaluated items in the UHDRS assessment. Tau also has a potential for use as a biomarker but correlations to clinical tests are weaker in this study. We suggest that NFL and possibly tau be used in clinical drug trials as biomarkers of disease progression that are potentially influenced by future disease-modifying therapies.
Collapse
Affiliation(s)
- Valter Niemelä
- Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden
| | | | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal Campus, Mölndal, Sweden
| | - Jimmy Sundblom
- Department of Neuroscience, Neurosurgery, Uppsala University, Uppsala, Sweden
| |
Collapse
|
584
|
Hallmarks of Alzheimer's Disease in Stem-Cell-Derived Human Neurons Transplanted into Mouse Brain. Neuron 2017; 93:1066-1081.e8. [PMID: 28238547 DOI: 10.1016/j.neuron.2017.02.001] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 12/12/2016] [Accepted: 01/31/2017] [Indexed: 01/11/2023]
Abstract
Human pluripotent stem cells (PSCs) provide a unique entry to study species-specific aspects of human disorders such as Alzheimer's disease (AD). However, in vitro culture of neurons deprives them of their natural environment. Here we transplanted human PSC-derived cortical neuronal precursors into the brain of a murine AD model. Human neurons differentiate and integrate into the brain, express 3R/4R Tau splice forms, show abnormal phosphorylation and conformational Tau changes, and undergo neurodegeneration. Remarkably, cell death was dissociated from tangle formation in this natural 3D model of AD. Using genome-wide expression analysis, we observed upregulation of genes involved in myelination and downregulation of genes related to memory and cognition, synaptic transmission, and neuron projection. This novel chimeric model for AD displays human-specific pathological features and allows the analysis of different genetic backgrounds and mutations during the course of the disease.
Collapse
|
585
|
Nucleobindin 1 binds to multiple types of pre-fibrillar amyloid and inhibits fibrillization. Sci Rep 2017; 7:42880. [PMID: 28220836 PMCID: PMC5318909 DOI: 10.1038/srep42880] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 01/16/2017] [Indexed: 11/08/2022] Open
Abstract
During amyloid fibril formation, amyloidogenic polypeptides misfold and self assemble into soluble pre-fibrillar aggregates, i.e., protofibrils, which elongate and mature into insoluble fibrillar aggregates. An emerging class of chaperones, chaperone-like amyloid binding proteins (CLABPs), has been shown to interfere with aggregation of particular misfolded amyloid peptides or proteins. We have discovered that the calcium-binding protein nuclebindin-1 (NUCB1) is a novel CLABP. We show that NUCB1 inhibits aggregation of islet-amyloid polypeptide associated with type 2 diabetes mellitus, a-synuclein associated with Parkinson’s disease, transthyretin V30M mutant associated with familial amyloid polyneuropathy, and Aβ42 associated with Alzheimer’s disease by stabilizing their respective protofibril intermediates. Kinetic studies employing the modeling software AmyloFit show that NUCB1 affects both primary nucleation and secondary nucleation. We hypothesize that NUCB1 binds to the common cross-β-sheet structure of protofibril aggregates to “cap” and stabilize soluble macromolecular complexes. Transmission electron microscopy and atomic force microscopy were employed to characterize the size, shape and volume distribution of multiple sources of NUCB1-capped protofibrils. Interestingly, NUCB1 prevents Aβ42 protofibril toxicity in a cellular assay. NUCB1-stabilized amyloid protofibrils could be used as immunogens to prepare conformation-specific antibodies and as novel tools to develop screens for anti-protofibril diagnostics and therapeutics.
Collapse
|
586
|
Buccarello L, Grignaschi G, Castaldo AM, Di Giancamillo A, Domeneghini C, Melcangi RC, Borsello T. Sex Impact on Tau-Aggregation and Postsynaptic Protein Levels in the P301L Mouse Model of Tauopathy. J Alzheimers Dis 2017; 56:1279-1292. [DOI: 10.3233/jad-161087] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Lucia Buccarello
- Department of Neuroscience, IRCCS-Mario Negri Institute for Pharmacological Research, Milan, Italy
- Department of Health, Animal Science and Food Safety, Universitá degli Studi di Milano, Italy
| | - Giuliano Grignaschi
- Department of Neuroscience, IRCCS-Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Anna Maria Castaldo
- Department of Neuroscience, IRCCS-Mario Negri Institute for Pharmacological Research, Milan, Italy
- Department of Biomedical Sciences for Health, Universitá degli Studi di Milano, Italy
| | - Alessia Di Giancamillo
- Department of Health, Animal Science and Food Safety, Universitá degli Studi di Milano, Italy
| | - Cinzia Domeneghini
- Department of Health, Animal Science and Food Safety, Universitá degli Studi di Milano, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Universitá degli Studi di Milano, Italy
| | - Tiziana Borsello
- Department of Neuroscience, IRCCS-Mario Negri Institute for Pharmacological Research, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, Universitá degli Studi di Milano, Italy
| |
Collapse
|
587
|
Rombouts FJR, Andrés JI, Ariza M, Alonso JM, Austin N, Bottelbergs A, Chen L, Chupakhin V, Cleiren E, Fierens K, Fontana A, Langlois X, Leenaerts JE, Mariën J, Martínez Lamenca C, Salter R, Schmidt ME, Te Riele P, Wintmolders C, Trabanco AA, Zhang W, Macdonald G, Moechars D. Discovery of N-(Pyridin-4-yl)-1,5-naphthyridin-2-amines as Potential Tau Pathology PET Tracers for Alzheimer's Disease. J Med Chem 2017; 60:1272-1291. [PMID: 28106992 DOI: 10.1021/acs.jmedchem.6b01173] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A mini-HTS on 4000 compounds selected using 2D fragment-based similarity and 3D pharmacophoric and shape similarity to known selective tau aggregate binders identified N-(6-methylpyridin-2-yl)quinolin-2-amine 10 as a novel potent binder to human AD aggregated tau with modest selectivity versus aggregated β-amyloid (Aβ). Initial medicinal chemistry efforts identified key elements for potency and selectivity, as well as suitable positions for radiofluorination, leading to a first generation of fluoroalkyl-substituted quinoline tau binding ligands with suboptimal physicochemical properties. Further optimization toward a more optimal pharmacokinetic profile led to the discovery of 1,5-naphthyridine 75, a potent and selective tau aggregate binder with potential as a tau PET tracer.
Collapse
Affiliation(s)
- Frederik J R Rombouts
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - José-Ignacio Andrés
- Discovery Sciences, Janssen Research & Development, Janssen-Cilag S. A. , C/Jarama 75A, 45007 Toledo, Spain
| | - Manuela Ariza
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - José Manuel Alonso
- Discovery Sciences, Janssen Research & Development, Janssen-Cilag S. A. , C/Jarama 75A, 45007 Toledo, Spain
| | - Nigel Austin
- Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Astrid Bottelbergs
- Discovery Sciences, Janssen Research & Development, Janssen-Cilag S. A. , C/Jarama 75A, 45007 Toledo, Spain
| | - Lu Chen
- Isotope Chemistry and Biotransformation, Janssen Research & Development , Welsh & McKean Roads, Spring House, Pennsylvania 19477, United States
| | - Vladimir Chupakhin
- Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Erna Cleiren
- Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Katleen Fierens
- Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Alberto Fontana
- Discovery Sciences, Janssen Research & Development, Janssen-Cilag S. A. , C/Jarama 75A, 45007 Toledo, Spain
| | - Xavier Langlois
- Neuroscience Biology, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Joseph E Leenaerts
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Jonas Mariën
- Neuroscience Biology, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Carolina Martínez Lamenca
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Rhys Salter
- Isotope Chemistry and Biotransformation, Janssen Research & Development , Welsh & McKean Roads, Spring House, Pennsylvania 19477, United States
| | - Mark E Schmidt
- Neuroscience Experimental Medicine, Janssen Early Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Paula Te Riele
- Neuroscience Biology, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Cindy Wintmolders
- Neuroscience Biology, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Andrés A Trabanco
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S. A. , C/Jarama 75A, 45007 Toledo, Spain
| | - Wei Zhang
- Neuroscience Biomarker Research, Janssen Research & Development , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Gregor Macdonald
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Dieder Moechars
- Neuroscience Biology, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
588
|
Wang Z, Xiong L, Wan W, Duan L, Bai X, Zu H. Intranasal BMP9 Ameliorates Alzheimer Disease-Like Pathology and Cognitive Deficits in APP/PS1 Transgenic Mice. Front Mol Neurosci 2017; 10:32. [PMID: 28228716 PMCID: PMC5296319 DOI: 10.3389/fnmol.2017.00032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 01/27/2017] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common type of dementia and has no effective therapies. Previous studies showed that bone morphogenetic protein 9 (BMP9), an important factor in the differentiation and phenotype maintenance of cholinergic neurons, ameliorated the cholinergic defects resulting from amyloid deposition. These findings suggest that BMP9 has potential as a therapeutic agent for AD. However, the effects of BMP9 on cognitive function in AD and its underlying mechanisms remain elusive. In the present study, BMP9 was delivered intranasally to 7-month-old APP/PS1 mice for 4 weeks. Our data showed that intranasal BMP9 administration significantly improved the spatial and associative learning and memory of APP/PS1 mice. We also found that intranasal BMP9 administration significantly reduced the amyloid β (Aβ) plaques overall, inhibited tau hyperphosphorylation, and suppressed neuroinflammation in the transgenic mouse brain. Furthermore, intranasal BMP9 administration significantly promoted the expression of low-density lipoprotein receptor-related protein 1 (LRP1), an important membrane receptor involved in the clearance of amyloid β via the blood-brain barrier (BBB), and elevated the phosphorylation levels of glycogen synthase kinase-3β (Ser9), which is considered the main kinase involved in tau hyperphosphorylation. Our results suggest that BMP9 may be a promising candidate for treating AD by targeting multiple key pathways in the disease pathogenesis.
Collapse
Affiliation(s)
- Zigao Wang
- Department of Neurology, Jinshan Hospital, Fudan University Shanghai, China
| | - Lu Xiong
- Department of Anesthesiology, Tinglin Hospital Shanghai, China
| | - Wenbin Wan
- Department of Neurology, Zhongshan Hospital, Fudan University Shanghai, China
| | - Lijie Duan
- Department of Neurology, Jinshan Hospital, Fudan University Shanghai, China
| | - Xiaojing Bai
- Department of Neurology, Jinshan Hospital, Fudan University Shanghai, China
| | - Hengbing Zu
- Department of Neurology, Jinshan Hospital, Fudan University Shanghai, China
| |
Collapse
|
589
|
Lemstra AW, de Beer MH, Teunissen CE, Schreuder C, Scheltens P, van der Flier WM, Sikkes SAM. Concomitant AD pathology affects clinical manifestation and survival in dementia with Lewy bodies. J Neurol Neurosurg Psychiatry 2017; 88:113-118. [PMID: 27794030 DOI: 10.1136/jnnp-2016-313775] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 09/14/2016] [Accepted: 09/27/2016] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To investigate whether concomitant Alzheimer's disease (AD) pathology, reflected by cerebrospinal fluid (CSF) biomarkers, has an impact on dementia with Lewy bodies (DLB) in terms of clinical presentation, cognitive decline, nursing home admittance and survival. PARTICIPANTS We selected 111 patients with probable DLB and CSF available from the Amsterdam Dementia Cohort. On the basis of the AD biomarker profile (CSF tau/amyloid-β 1-42 (Aβ42) ratio >0.52), we divided patients into a DLB/AD+ and DLB/AD- group. Of the 111 patients, 42 (38%) had an AD CSF biomarker profile. We investigated differences between groups in memory, attention, executive functions, language and visuospatial functions. Difference in global cognitive decline (repeated Mini-Mental State Examination (MMSE)) was investigated using linear mixed models. Cox proportional hazard analyses were used to investigate the effects of the AD biomarker profile on time to nursing home admittance and time to death. RESULTS Memory performance was worse in DLB/AD+ patients compared with DLB/AD- patients (p<0.01), also after correction for age and sex. Hallucinations were more frequent in DLB/AD+ (OR=3.34, 95% CI 1.22-9.18). There was no significant difference in the rate of cognitive decline. DLB/AD+ patients had a higher mortality risk (HR=3.13, 95% CI 1.57 to 6.24) and nursing home admittance risk (HR=11.70, 95% CI 3.74 to 36.55) compared with DLB/AD- patients. CONCLUSIONS DLB-patients with a CSF AD profile have a more severe manifestation of the disease and a higher risk of institutionalisation and mortality. In clinical practice, CSF biomarkers may aid in predicting prognosis in DLB. In addition, DLB-patients with positive AD biomarkers could benefit from future treatment targeting AD pathology.
Collapse
Affiliation(s)
- A W Lemstra
- Alzheimer Center & Department of Neurology, VU University Medical Center and Neuroscience Campus, Amsterdam, The Netherlands
| | - M H de Beer
- Alzheimer Center & Department of Neurology, VU University Medical Center and Neuroscience Campus, Amsterdam, The Netherlands.,HagaZiekenhuis, Haga Hospital, The Hague, The Netherlands
| | - C E Teunissen
- Department of Clinical Chemistry, VU University Medical Center & Alzheimer Center, Amsterdam, The Netherlands
| | - C Schreuder
- Department of Medical Psychology & Alzheimer center, VU University Medical Center & Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
| | - P Scheltens
- Alzheimer Center & Department of Neurology, VU University Medical Center and Neuroscience Campus, Amsterdam, The Netherlands
| | - W M van der Flier
- Alzheimer center & Department of Neurology and Department of Epidemiology and Biostatistics, VU University Medical Center and Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
| | - S A M Sikkes
- Alzheimer center & Department of Epidemiology and Biostatistics, VU University Medical Center and Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
590
|
Ferrari R, Wang Y, Vandrovcova J, Guelfi S, Witeolar A, Karch CM, Schork AJ, Fan CC, Brewer JB, Momeni P, Schellenberg GD, Dillon WP, Sugrue LP, Hess CP, Yokoyama JS, Bonham LW, Rabinovici GD, Miller BL, Andreassen OA, Dale AM, Hardy J, Desikan RS. Genetic architecture of sporadic frontotemporal dementia and overlap with Alzheimer's and Parkinson's diseases. J Neurol Neurosurg Psychiatry 2017; 88:152-164. [PMID: 27899424 PMCID: PMC5237405 DOI: 10.1136/jnnp-2016-314411] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 11/04/2022]
Abstract
BACKGROUND Clinical, pathological and genetic overlap between sporadic frontotemporal dementia (FTD), Alzheimer's disease (AD) and Parkinson's disease (PD) has been suggested; however, the relationship between these disorders is still not well understood. Here we evaluated genetic overlap between FTD, AD and PD to assess shared pathobiology and identify novel genetic variants associated with increased risk for FTD. METHODS Summary statistics were obtained from the International FTD Genomics Consortium, International PD Genetics Consortium and International Genomics of AD Project (n>75 000 cases and controls). We used conjunction false discovery rate (FDR) to evaluate genetic pleiotropy and conditional FDR to identify novel FTD-associated SNPs. Relevant variants were further evaluated for expression quantitative loci. RESULTS We observed SNPs within the HLA, MAPT and APOE regions jointly contributing to increased risk for FTD and AD or PD. By conditioning on polymorphisms associated with PD and AD, we found 11 loci associated with increased risk for FTD. Meta-analysis across two independent FTD cohorts revealed a genome-wide signal within the APOE region (rs6857, 3'-UTR=PVRL2, p=2.21×10-12), and a suggestive signal for rs1358071 within the MAPT region (intronic=CRHR1, p=4.91×10-7) with the effect allele tagging the H1 haplotype. Pleiotropic SNPs at the HLA and MAPT loci associated with expression changes in cis-genes supporting involvement of intracellular vesicular trafficking, immune response and endo/lysosomal processes. CONCLUSIONS Our findings demonstrate genetic pleiotropy in these neurodegenerative diseases and indicate that sporadic FTD is a polygenic disorder where multiple pleiotropic loci with small effects contribute to increased disease risk.
Collapse
Affiliation(s)
- Raffaele Ferrari
- Department of Molecular Neuroscience, Institute of Neurology, UCL, London, UK
| | - Yunpeng Wang
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Jana Vandrovcova
- Department of Molecular Neuroscience, Institute of Neurology, UCL, London, UK.,Department of Medical & Molecular Genetics, King's College London, Guy's Hospital, London, UK
| | - Sebastian Guelfi
- Department of Molecular Neuroscience, Institute of Neurology, UCL, London, UK.,Department of Medical & Molecular Genetics, King's College London, Guy's Hospital, London, UK
| | - Aree Witeolar
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Celeste M Karch
- Department of Psychiatry, Washington University, St. Louis, Missouri, USA
| | - Andrew J Schork
- Department of Cognitive Sciences, University of California, San Diego, La Jolla, California, USA
| | - Chun C Fan
- Department of Cognitive Sciences, University of California, San Diego, La Jolla, California, USA
| | - James B Brewer
- Department of Radiology, University of California, San Diego, La Jolla, California, USA.,Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | | | | | | | - Parastoo Momeni
- Laboratory of Neurogenetics, Department of Internal Medicine, Texas Tech University Health Science Center, Lubbock, Texas, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - William P Dillon
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Leo P Sugrue
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Christopher P Hess
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Jennifer S Yokoyama
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Luke W Bonham
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Gil D Rabinovici
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Bruce L Miller
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Ole A Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Anders M Dale
- Department of Cognitive Sciences, University of California, San Diego, La Jolla, California, USA.,Department of Radiology, University of California, San Diego, La Jolla, California, USA.,Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, UCL, London, UK
| | - Rahul S Desikan
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
591
|
Hensley K, Kursula P. Collapsin Response Mediator Protein-2 (CRMP2) is a Plausible Etiological Factor and Potential Therapeutic Target in Alzheimer's Disease: Comparison and Contrast with Microtubule-Associated Protein Tau. J Alzheimers Dis 2017; 53:1-14. [PMID: 27079722 PMCID: PMC4942723 DOI: 10.3233/jad-160076] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer’s disease (AD) has long been viewed as a pathology that must be caused either by aberrant amyloid-β protein precursor (AβPP) processing, dysfunctional tau protein processing, or a combination of these two factors. This is a reasonable assumption because amyloid-β peptide (Aβ) accumulation and tau hyperphosphorylation are the defining histological features in AD, and because AβPP and tau mutations can cause AD in humans or AD-like features in animal models. Nonetheless, other protein players are emerging that one can argue are significant etiological players in subsets of AD and potentially novel, druggable targets. In particular, the microtubule-associated protein CRMP2 (collapsin response mediator protein-2) bears striking analogies to tau and is similarly relevant to AD. Like tau, CRMP2 dynamically regulates microtubule stability; it is acted upon by the same kinases; collects similarly in neurofibrillary tangles (NFTs); and when sequestered in NFTs, complexes with critical synapse-stabilizing factors. Additionally, CRMP2 is becoming recognized as an important adaptor protein involved in vesicle trafficking, amyloidogenesis and autophagy, in ways that tau is not. This review systematically compares the biology of CRMP2 to that of tau in the context of AD and explores the hypothesis that CRMP2 is an etiologically significant protein in AD and participates in pathways that can be rationally engaged for therapeutic benefit.
Collapse
Affiliation(s)
- Kenneth Hensley
- Department of Pathology, University of Toledo Health Science Campus, Toledo, OH, USA
| | - Petri Kursula
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
592
|
Hoffman A, Taleski G, Sontag E. The protein serine/threonine phosphatases PP2A, PP1 and calcineurin: A triple threat in the regulation of the neuronal cytoskeleton. Mol Cell Neurosci 2017; 84:119-131. [PMID: 28126489 DOI: 10.1016/j.mcn.2017.01.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/16/2017] [Accepted: 01/21/2017] [Indexed: 01/08/2023] Open
Abstract
The microtubule, F-actin and neurofilament networks play a critical role in neuronal cell morphogenesis, polarity and synaptic plasticity. Significantly, the assembly/disassembly and stability of these cytoskeletal networks is crucially modulated by protein phosphorylation and dephosphorylation events. Herein, we aim to more closely examine the role played by three major neuronal Ser/Thr protein phosphatases, PP2A, PP1 and calcineurin, in the homeostasis of the neuronal cytoskeleton. There is strong evidence that these enzymes interact with and dephosphorylate a variety of cytoskeletal proteins, resulting in major regulation of neuronal cytoskeletal dynamics. Conversely, we also discuss how multi-protein cytoskeletal scaffolds can also influence the regulation of these phosphatases, with important implications for neuronal signalling and homeostasis. Not surprisingly, deregulation of these cytoskeletal scaffolds and phosphatase dysfunction are associated with many neurological diseases.
Collapse
Affiliation(s)
- Alexander Hoffman
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Goce Taleski
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Estelle Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
593
|
Developmental Expression of 4-Repeat-Tau Induces Neuronal Aneuploidy in Drosophila Tauopathy Models. Sci Rep 2017; 7:40764. [PMID: 28112163 PMCID: PMC5256094 DOI: 10.1038/srep40764] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 12/12/2016] [Indexed: 01/23/2023] Open
Abstract
Tau-mediated neurodegeneration in Alzheimer’s disease and tauopathies is generally assumed to start in a normally developed brain. However, several lines of evidence suggest that impaired Tau isoform expression during development could affect mitosis and ploidy in post-mitotic differentiated tissue. Interestingly, the relative expression levels of Tau isoforms containing either 3 (3R-Tau) or 4 repeats (4R-Tau) play an important role both during brain development and neurodegeneration. Here, we used genetic and cellular tools to study the link between 3R and 4R-Tau isoform expression, mitotic progression in neuronal progenitors and post-mitotic neuronal survival. Our results illustrated that the severity of Tau-induced adult phenotypes depends on 4R-Tau isoform expression during development. As recently described, we observed a mitotic delay in 4R-Tau expressing cells of larval eye discs and brains. Live imaging revealed that the spindle undergoes a cycle of collapse and recovery before proceeding to anaphase. Furthermore, we found a high level of aneuploidy in post-mitotic differentiated tissue. Finally, we showed that overexpression of wild type and mutant 4R-Tau isoform in neuroblastoma SH-SY5Y cell lines is sufficient to induce monopolar spindles. Taken together, our results suggested that neurodegeneration could be in part linked to neuronal aneuploidy caused by 4R-Tau expression during brain development.
Collapse
|
594
|
Li X, Wang Z, Tan L, Wang Y, Lu C, Chen R, Zhang S, Gao Y, Liu Y, Yin Y, Liu X, Liu E, Yang Y, Hu Y, Xu Z, Xu F, Wang J, Liu GP, Wang JZ. Correcting miR92a-vGAT-Mediated GABAergic Dysfunctions Rescues Human Tau-Induced Anxiety in Mice. Mol Ther 2017; 25:140-152. [PMID: 28129110 DOI: 10.1016/j.ymthe.2016.10.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 10/10/2016] [Accepted: 10/11/2016] [Indexed: 01/30/2023] Open
Abstract
Patients with Alzheimer's disease (AD) commonly show anxiety behaviors, but the molecular mechanisms are not clear and no efficient intervention exists. Here, we found that overexpression of human wild-type, full-length tau (termed htau) in hippocampus significantly decreased the extracellular γ-aminobutyric acid (GABA) level with inhibition of γ oscillation and the evoked inhibitory postsynaptic potential (eIPSP). With tau accumulation, the mice show age-dependent anxiety behaviors. Among the factors responsible for GABA synthesis, release, uptake, and transport, we found that accumulation of htau selectively suppressed expression of the intracellular vesicular GABA transporter (vGAT). Tau accumulation increased miR92a, which targeted vGAT mRNA 3' UTR and inhibited vGAT translation. Importantly, we found that upregulating GABA tones by intraperitoneal injection of midazolam (a GABA agonist), ChR2-mediated photostimulating and overexpressing vGAT, or blocking miR92a by using specific antagomir or inhibitor efficiently rescued the htau-induced GABAergic dysfunctions with attenuation of anxiety. Finally, we also demonstrated that vGAT level decreased while the miR92a increased in the AD brains. These findings demonstrate that the AD-like tau accumulation induces anxiety through disrupting miR92a-vGAT-GABA signaling, which reveals molecular mechanisms underlying the anxiety behavior in AD patients and potentially leads to the development of new therapeutics for tauopathies.
Collapse
Affiliation(s)
- Xiaoguang Li
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhihao Wang
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lu Tan
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Yali Wang
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Physiology Department, Henan Provincial Key Laboratory for Brain Research, Xinxiang Medical University, Xinxiang 453000, China
| | - Chengbiao Lu
- Physiology Department, Henan Provincial Key Laboratory for Brain Research, Xinxiang Medical University, Xinxiang 453000, China
| | - Rongxiang Chen
- State Key Laboratory for Magnet Resonance and Atom and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academia of Science, Wuhan 430071, China
| | - Shujuan Zhang
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuan Gao
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanchao Liu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yaling Yin
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinghua Liu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Enjie Liu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Hu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhipeng Xu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fuqiang Xu
- State Key Laboratory for Magnet Resonance and Atom and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academia of Science, Wuhan 430071, China
| | - Jie Wang
- State Key Laboratory for Magnet Resonance and Atom and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academia of Science, Wuhan 430071, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| |
Collapse
|
595
|
Abstract
Protein phosphorylation regulates brain development and neuronal activities; and dysregulation of phosphorylation contributes to neurobiological disorders. Phosphoproteomic analysis provides comprehensive modification maps for measuring protein activities in cellular pathways and biological processes. Here, we introduce a mass spectrometry (MS)-based protocol to quantitatively analyze the phosphoproteome of human postmortem brains of Alzheimer's disease. In this isobaric labeling protocol, up to ten brain samples are selected from control and diseased cases for comparison. Approximately 1 mg proteins per sample are extracted, digested, labeled, and then mixed at an equal ratio. To improve the coverage of phosphoproteome, the peptide mix is further fractionated by offline basic pH reversed-phase liquid chromatography (LC) with high-resolution power. Phosphopeptides in each fraction are then enriched by the titanium dioxide method and analyzed by online acidic pH reverse phase LC-MS/MS, leading to the analysis of tens of thousands of phosphorylation events. This protocol can also be adapted to profile phosphoproteome in other biological samples.
Collapse
|
596
|
Antioxidative and neuroprotective activities of peanut sprout extracts against oxidative stress in SK-N-SH cells. Asian Pac J Trop Biomed 2017. [DOI: 10.1016/j.apjtb.2016.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
597
|
Quinlan S, Kenny A, Medina M, Engel T, Jimenez-Mateos EM. MicroRNAs in Neurodegenerative Diseases. MIRNAS IN AGING AND CANCER 2017; 334:309-343. [DOI: 10.1016/bs.ircmb.2017.04.002] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
598
|
Xie C, Miyasaka T. The Role of the Carboxyl-Terminal Sequence of Tau and MAP2 in the Pathogenesis of Dementia. Front Mol Neurosci 2016; 9:158. [PMID: 28082867 PMCID: PMC5186789 DOI: 10.3389/fnmol.2016.00158] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/08/2016] [Indexed: 01/25/2023] Open
Abstract
Dementia includes several diseases characterized by acquired and irreversible brain dysfunctions that interfere with daily life. According to the etiology, dementia can be induced by poisoning or metabolic disorders, and other cases of dementia have a clear pathogenesis. However, half of neurodegenerative diseases have an unclear pathogenesis and etiology. Alzheimer’s disease (AD), Lewy body dementia and frontal-temporal dementia are the three most common types of dementia. These neurodegenerative diseases are characterized by the appearance of the following specific protein inclusions: amyloid beta and tau in AD; α-synuclein in Lewy body dementia; and tau, TDP-43, or FUS in frontal-temporal dementia. Thus far, studies on the pathogenesis of dementia mainly focus aberrant inclusions formed by the aforementioned proteins. As a historically heavily studied protein tau is likely to be associated with the pathogenesis of several neurodegenerative diseases that cause dementia. The role of tau in neurodegeneration has been unknown for many years. However, both pathological and genetic analyses have helped tau become gradually recognized as an important factor in the pathogenesis of tauopathy. Currently, especially in the field of AD, tau is attracting more attention and is being considered a potential target for drug development. In this review article, previously discovered biochemical and pathological features of tau are highlighted, and current opinions regarding the neurotoxicity of tau are summarized. Additionally, we introduce key amino acid sequences responsible for tau neurotoxicity from our studies using transgenic Caenorhabditis elegans. Finally, a new hypothesis regarding the roles of microtubule-associated protein 2 (MAP2) and tau in the pathogenesis of tauopathy is discussed.
Collapse
Affiliation(s)
- Ce Xie
- College of Basic Medical Sciences, Dalian Medical UniversityDalian, China; Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha UniversityKyotanabe, Japan
| | - Tomohiro Miyasaka
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University Kyotanabe, Japan
| |
Collapse
|
599
|
Safety and immunogenicity of the tau vaccine AADvac1 in patients with Alzheimer's disease: a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Neurol 2016; 16:123-134. [PMID: 27955995 DOI: 10.1016/s1474-4422(16)30331-3] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 10/20/2016] [Accepted: 10/20/2016] [Indexed: 01/06/2023]
Abstract
BACKGROUND Neurofibrillary pathology composed of tau protein is a main correlate of cognitive impairment in patients with Alzheimer's disease. Immunotherapy targeting pathological tau proteins is therefore a promising strategy for disease-modifying treatment of Alzheimer's disease. We have developed an active vaccine, AADvac1, against pathological tau proteins and assessed it in a phase 1 trial. METHODS We did a first-in-man, phase 1, 12 week, randomised, double-blind, placebo-controlled study of AADvac1 with a 12 week open-label extension in patients aged 50-85 years with mild-to-moderate Alzheimer's disease at four centres in Austria. We randomly assigned patients with a computer-generated sequence in a 4:1 ratio overall to receive AADvac1 or placebo. They received three subcutaneous doses of AADvac1 or placebo from masked vaccine kits at monthly intervals, and then entered the open-label phase, in which all patients were allocated to AADvac1 treatment and received another three doses at monthly intervals. Patients, carers, and all involved with the trial were masked to treatment allocation. The primary endpoint was all-cause treatment-emergent adverse events, with separate analyses for injection site reactions and other adverse events. We include all patients who received at least one dose of AADvac1 in the safety assessment. Patients who had a positive IgG titre against the tau peptide component of AADvac1 at least once during the study were classified as responders. The first-in-man study is registered with EU Clinical Trials Register, number EudraCT 2012-003916-29, and ClinicalTrials.gov, number NCT01850238; the follow-up study, which is ongoing, is registered with EU Clinical Trials Register, number EudraCT 2013-004499-36, and ClinicalTrials.gov, number NCT02031198. FINDINGS This study was done between June 9, 2013, and March 26, 2015. 30 patients were randomly assigned in the double-blind phase: 24 patients to the AADvac1 group and six to the placebo group. A total of 30 patients received AADvac1. Two patients withdrew because of serious adverse events. The most common adverse events were injection site reactions after administration (reported in 16 [53%] vaccinated patients [92 individual events]). No cases of meningoencephalitis or vasogenic oedema occurred after administration. One patient with pre-existing microhaemorrhages had newly occurring microhaemorrhages. Of 30 patients given AADvac1, 29 developed an IgG immune response. A geometric mean IgG antibody titre of 1:31415 was achieved. Baseline values of CD3+ CD4+ lymphocytes correlated with achieved antibody titres. INTERPRETATION AADvac1 had a favourable safety profile and excellent immunogenicity in this first-in-man study. Further trials are needed to corroborate the safety assessment and to establish proof of clinical efficacy of AADvac1. FUNDING AXON Neuroscience SE.
Collapse
|
600
|
Adams HHH, Hibar DP, Chouraki V, Stein JL, Nyquist PA, Rentería ME, Trompet S, Arias-Vasquez A, Seshadri S, Desrivières S, Beecham AH, Jahanshad N, Wittfeld K, Van der Lee SJ, Abramovic L, Alhusaini S, Amin N, Andersson M, Arfanakis K, Aribisala BS, Armstrong NJ, Athanasiu L, Axelsson T, Beiser A, Bernard M, Bis JC, Blanken LME, Blanton SH, Bohlken MM, Boks MP, Bralten J, Brickman AM, Carmichael O, Chakravarty MM, Chauhan G, Chen Q, Ching CRK, Cuellar-Partida G, Braber AD, Doan NT, Ehrlich S, Filippi I, Ge T, Giddaluru S, Goldman AL, Gottesman RF, Greven CU, Grimm O, Griswold ME, Guadalupe T, Hass J, Haukvik UK, Hilal S, Hofer E, Hoehn D, Holmes AJ, Hoogman M, Janowitz D, Jia T, Kasperaviciute D, Kim S, Klein M, Kraemer B, Lee PH, Liao J, Liewald DCM, Lopez LM, Luciano M, Macare C, Marquand A, Matarin M, Mather KA, Mattheisen M, Mazoyer B, McKay DR, McWhirter R, Milaneschi Y, Mirza-Schreiber N, Muetzel RL, Maniega SM, Nho K, Nugent AC, Loohuis LMO, Oosterlaan J, Papmeyer M, Pappa I, Pirpamer L, Pudas S, Pütz B, Rajan KB, Ramasamy A, Richards JS, Risacher SL, Roiz-Santiañez R, Rommelse N, Rose EJ, Royle NA, Rundek T, Sämann PG, Satizabal CL, Schmaal L, Schork AJ, Shen L, Shin J, Shumskaya E, Smith AV, Sprooten E, Strike LT, Teumer A, Thomson R, Tordesillas-Gutierrez D, Toro R, Trabzuni D, Vaidya D, Van der Grond J, Van der Meer D, Van Donkelaar MMJ, Van Eijk KR, Van Erp TGM, Van Rooij D, Walton E, Westlye LT, Whelan CD, Windham BG, Winkler AM, Woldehawariat G, Wolf C, Wolfers T, Xu B, Yanek LR, Yang J, Zijdenbos A, Zwiers MP, Agartz I, Aggarwal NT, Almasy L, Ames D, Amouyel P, Andreassen OA, Arepalli S, Assareh AA, Barral S, Bastin ME, Becker DM, Becker JT, Bennett DA, Blangero J, van Bokhoven H, Boomsma DI, Brodaty H, Brouwer RM, Brunner HG, Buckner RL, Buitelaar JK, Bulayeva KB, Cahn W, Calhoun VD, Cannon DM, Cavalleri GL, Chen C, Cheng CY, Cichon S, Cookson MR, Corvin A, Crespo-Facorro B, Curran JE, Czisch M, Dale AM, Davies GE, De Geus EJC, De Jager PL, de Zubicaray GI, Delanty N, Depondt C, DeStefano AL, Dillman A, Djurovic S, Donohoe G, Drevets WC, Duggirala R, Dyer TD, Erk S, Espeseth T, Evans DA, Fedko IO, Fernández G, Ferrucci L, Fisher SE, Fleischman DA, Ford I, Foroud TM, Fox PT, Francks C, Fukunaga M, Gibbs JR, Glahn DC, Gollub RL, Göring HHH, Grabe HJ, Green RC, Gruber O, Gudnason V, Guelfi S, Hansell NK, Hardy J, Hartman CA, Hashimoto R, Hegenscheid K, Heinz A, Le Hellard S, Hernandez DG, Heslenfeld DJ, Ho BC, Hoekstra PJ, Hoffmann W, Hofman A, Holsboer F, Homuth G, Hosten N, Hottenga JJ, Hulshoff Pol HE, Ikeda M, Ikram MK, Jack CR, Jenkinson M, Johnson R, Jönsson EG, Jukema JW, Kahn RS, Kanai R, Kloszewska I, Knopman DS, Kochunov P, Kwok JB, Lawrie SM, Lemaître H, Liu X, Longo DL, Longstreth WT, Lopez OL, Lovestone S, Martinez O, Martinot JL, Mattay VS, McDonald C, McIntosh AM, McMahon KL, McMahon FJ, Mecocci P, Melle I, Meyer-Lindenberg A, Mohnke S, Montgomery GW, Morris DW, Mosley TH, Mühleisen TW, Müller-Myhsok B, Nalls MA, Nauck M, Nichols TE, Niessen WJ, Nöthen MM, Nyberg L, Ohi K, Olvera RL, Ophoff RA, Pandolfo M, Paus T, Pausova Z, Penninx BWJH, Pike GB, Potkin SG, Psaty BM, Reppermund S, Rietschel M, Roffman JL, Romanczuk-Seiferth N, Rotter JI, Ryten M, Sacco RL, Sachdev PS, Saykin AJ, Schmidt R, Schofield PR, Sigurdsson S, Simmons A, Singleton A, Sisodiya SM, Smith C, Smoller JW, Soininen H, Srikanth V, Steen VM, Stott DJ, Sussmann JE, Thalamuthu A, Tiemeier H, Toga AW, Traynor BJ, Troncoso J, Turner JA, Tzourio C, Uitterlinden AG, Hernández MCV, Van der Brug M, Van der Lugt A, Van der Wee NJA, Van Duijn CM, Van Haren NEM, Van T Ent D, Van Tol MJ, Vardarajan BN, Veltman DJ, Vernooij MW, Völzke H, Walter H, Wardlaw JM, Wassink TH, Weale ME, Weinberger DR, Weiner MW, Wen W, Westman E, White T, Wong TY, Wright CB, Zielke HR, Zonderman AB, Deary IJ, DeCarli C, Schmidt H, Martin NG, De Craen AJM, Wright MJ, Launer LJ, Schumann G, Fornage M, Franke B, Debette S, Medland SE, Ikram MA, Thompson PM. Novel genetic loci underlying human intracranial volume identified through genome-wide association. Nat Neurosci 2016; 19:1569-1582. [PMID: 27694991 PMCID: PMC5227112 DOI: 10.1038/nn.4398] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/31/2016] [Indexed: 02/08/2023]
Abstract
Intracranial volume reflects the maximally attained brain size during development, and remains stable with loss of tissue in late life. It is highly heritable, but the underlying genes remain largely undetermined. In a genome-wide association study of 32,438 adults, we discovered five previously unknown loci for intracranial volume and confirmed two known signals. Four of the loci were also associated with adult human stature, but these remained associated with intracranial volume after adjusting for height. We found a high genetic correlation with child head circumference (ρgenetic = 0.748), which indicates a similar genetic background and allowed us to identify four additional loci through meta-analysis (Ncombined = 37,345). Variants for intracranial volume were also related to childhood and adult cognitive function, and Parkinson's disease, and were enriched near genes involved in growth pathways, including PI3K-AKT signaling. These findings identify the biological underpinnings of intracranial volume and their link to physiological and pathological traits.
Collapse
Affiliation(s)
- Hieab H H Adams
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Derrek P Hibar
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Vincent Chouraki
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
- Lille University, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk factors and molecular determinants of aging-related diseases, Lille, France
- Framingham Heart Study, Framingham, Massachusetts, USA
| | - Jason L Stein
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- Department of Genetics and UNC Neuroscience Center, University of North Carolina (UNC), Chapel Hill, North Carolina, USA
| | - Paul A Nyquist
- Department of Neurology, Department of Anesthesia/Critical Care Medicine, Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Alejandro Arias-Vasquez
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
- Framingham Heart Study, Framingham, Massachusetts, USA
| | - Sylvane Desrivières
- MRC-SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ashley H Beecham
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, Florida, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Neda Jahanshad
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Katharina Wittfeld
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Greifswald, Germany
- Department of Psychiatry, University Medicine Greifswald, Greifswald, Germany
| | | | - Lucija Abramovic
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht, the Netherlands
| | - Saud Alhusaini
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada
- The Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Micael Andersson
- Department of Integrative Medical Biology and Umeå center for Functional Brain Imaging, Umeå University, Umeå, Sweden
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Benjamin S Aribisala
- Brain Research Imaging Centre, University of Edinburgh, Edinburgh, UK
- Department of Computer Science, Lagos State University, Lagos, Nigeria
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Department of Neuroimaging Sciences, University of Edinburgh, Edinburgh, UK
| | - Nicola J Armstrong
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
- Mathematics and Statistics, Murdoch University, Perth, Australia
| | - Lavinia Athanasiu
- NORMENT - KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- NORMENT - KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Tomas Axelsson
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Alexa Beiser
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
- Framingham Heart Study, Framingham, Massachusetts, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Manon Bernard
- Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Laura M E Blanken
- Generation R Study Group, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Susan H Blanton
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, Florida, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Marc M Bohlken
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht, the Netherlands
| | - Marco P Boks
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht, the Netherlands
| | - Janita Bralten
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
- G.H. Sergievsky Center, Columbia University Medical Center, New York, New York, USA
- Department of Neurology, Columbia University Medical Center, New York, New York, USA
| | - Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - M Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Canada
- Department of Psychiatry and Biomedical Engineering, McGill University, Montreal, Canada
| | | | - Qiang Chen
- Lieber Institute for Brain Development, Baltimore, Maryland, USA
| | - Christopher R K Ching
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- Interdepartmental Neuroscience Graduate Program, UCLA School of Medicine, Los Angeles, California, USA
| | | | - Anouk Den Braber
- Biological Psychology, Neuroscience Campus Amsterdam, Vrije Universiteit University and Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | - Nhat Trung Doan
- NORMENT - KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Stefan Ehrlich
- Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Germany
- Department of Psychiatry, Massachusetts General Hospital, Boston, Masschusetts, USA
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, North Carolina, USA
| | - Irina Filippi
- NSERM Unit 1000 ″Neuroimaging and Psychiatry″, University Paris Sud, University Paris Descartes, Paris, France
- Maison de Solenn, Adolescent Psychopathology and Medicine Department, APHP Hospital Cochin, Paris, France
| | - Tian Ge
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, North Carolina, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Sudheer Giddaluru
- NORMENT - KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Aaron L Goldman
- Lieber Institute for Brain Development, Baltimore, Maryland, USA
| | - Rebecca F Gottesman
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Corina U Greven
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Karakter Child and Adolescent Psychiatry University Center, Nijmegen, the Netherlands
- King's College London, Medical Research Council Social, Genetic and Developmental Psychiatry Centre, Institute of Psychology, Psychiatry and Neurosciene, London, UK
| | - Oliver Grimm
- Central Institute of Mental Health, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Michael E Griswold
- Center of Biostatistics and Bioinformatics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Tulio Guadalupe
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
- International Max Planck Research School for Language Sciences, Nijmegen, the Netherlands
| | - Johanna Hass
- Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Unn K Haukvik
- NORMENT - KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Research and Development, Diakonhjemmet Hospital, Oslo, Norway
| | - Saima Hilal
- Department of Pharmacology, National University of Singapore, Singapore
- Memory Aging and Cognition Centre (MACC), National University Health System, Singapore
| | - Edith Hofer
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Austria, Graz, Austria
- Institute of Medical Informatics, Statistics and Documentation, Medical University Graz, Austria, Graz, Austria
| | - David Hoehn
- Max Planck Institute of Psychiatry, Department of Translational Research in Psychiatry, Munich, Germany
| | - Avram J Holmes
- Department of Psychiatry, Massachusetts General Hospital, Boston, Masschusetts, USA
- Department of Psychology, Yale University, New Haven, Connecticut, USA
| | - Martine Hoogman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Deborah Janowitz
- Department of Psychiatry, University Medicine Greifswald, Greifswald, Germany
| | - Tianye Jia
- MRC-SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Dalia Kasperaviciute
- UCL Institute of Neurology, London, United Kingdom and Epilepsy Society, Bucks, UK
- Department of Medicine, Imperial College London, London, UK
| | - Sungeun Kim
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Marieke Klein
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Bernd Kraemer
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University, Heidelberg, Germany
| | - Phil H Lee
- Department of Psychiatry, Massachusetts General Hospital, Boston, Masschusetts, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
- Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Lexington, Massachusetts, USA
| | - Jiemin Liao
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - David C M Liewald
- Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh, UK
| | - Lorna M Lopez
- Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh, UK
| | - Michelle Luciano
- Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh, UK
| | - Christine Macare
- MRC-SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Andre Marquand
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Donders Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, The Netherlands
| | - Mar Matarin
- UCL Institute of Neurology, London, United Kingdom and Epilepsy Society, Bucks, UK
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Manuel Mattheisen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus and Copenhagen, Denmark
- Center for integrated Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
| | | | - David R McKay
- Department of Psychiatry, Yale University, New Haven, Connecticut, USA
- Olin Neuropsychiatric Research Center, Hartford, Connecticut, USA
| | - Rebekah McWhirter
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Yuri Milaneschi
- Department of Psychiatry, EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ inGeest, Amsterdam, The Netherlands
| | - Nazanin Mirza-Schreiber
- Max Planck Institute of Psychiatry, Department of Translational Research in Psychiatry, Munich, Germany
| | - Ryan L Muetzel
- Generation R Study Group, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Susana Muñoz Maniega
- Brain Research Imaging Centre, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Department of Neuroimaging Sciences, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh, UK
| | - Kwangsik Nho
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health Intramural Research Program, National Institutes of Health, Bethesda, Maryland, USA
| | - Loes M Olde Loohuis
- Center for Neurobehavioral Genetics, University of California, Los Angeles, California, USA
| | - Jaap Oosterlaan
- Department of Clinical Neuropsychology, VU University Amsterdam, Amsterdam, the Netherlands
| | - Martina Papmeyer
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
- Division of Systems Neuroscience of Psychopathology, Translational Research Center, University Hospital of Psychiatry, University of Bern, Switzerland
| | - Irene Pappa
- Generation R Study Group, Erasmus Medical Center, Rotterdam, the Netherlands
- School of Pedagogical and Educational Sciences, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - Lukas Pirpamer
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Austria, Graz, Austria
| | - Sara Pudas
- Department of Integrative Medical Biology and Umeå center for Functional Brain Imaging, Umeå University, Umeå, Sweden
| | - Benno Pütz
- Max Planck Institute of Psychiatry, Department of Translational Research in Psychiatry, Munich, Germany
| | - Kumar B Rajan
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, Illinois, USA
| | - Adaikalavan Ramasamy
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
- The Jenner Institute Laboratories, University of Oxford, Oxford, UK
| | - Jennifer S Richards
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Karakter Child and Adolescent Psychiatry University Center, Nijmegen, the Netherlands
| | - Shannon L Risacher
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Roberto Roiz-Santiañez
- Department of Medicine and Psychiatry, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria-IDIVAL, Santander, Spain
- CIBERSAM (Centro Investigación Biomédica en Red Salud Mental), Santander, Spain
| | - Nanda Rommelse
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Karakter Child and Adolescent Psychiatry University Center, Nijmegen, the Netherlands
| | - Emma J Rose
- Psychosis Research Group, Department of Psychiatry and Trinity Translational Medicine Institute, Trinity College Dublin
| | - Natalie A Royle
- Brain Research Imaging Centre, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Department of Neuroimaging Sciences, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Tatjana Rundek
- Department of Neurology, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Department of Epidemiology and Public Health Sciences, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Philipp G Sämann
- Max Planck Institute of Psychiatry, Department of Translational Research in Psychiatry, Munich, Germany
| | - Claudia L Satizabal
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
- Framingham Heart Study, Framingham, Massachusetts, USA
| | - Lianne Schmaal
- Orygen, The National Centre of Excellence in Youth Mental Health, Melbourne, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
- Department of Psychiatry, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Andrew J Schork
- Multimodal Imaging Laboratory, Department of Neurosciences, University of California, San Diego, USA
- Department of Cognitive Sciences, University of California, San Diego, USA
| | - Li Shen
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jean Shin
- Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Elena Shumskaya
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Donders Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, The Netherlands
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Emma Sprooten
- Department of Psychiatry, Yale University, New Haven, Connecticut, USA
- Olin Neuropsychiatric Research Center, Hartford, Connecticut, USA
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lachlan T Strike
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Russell Thomson
- School of Computing Engineering and Mathematics, Western Sydney University, Parramatta, Australia
| | - Diana Tordesillas-Gutierrez
- CIBERSAM (Centro Investigación Biomédica en Red Salud Mental), Santander, Spain
- Neuroimaging Unit,Technological Facilities. Valdecilla Biomedical Research Institute IDIVAL, Santander, Cantabria, Spain
| | | | - Daniah Trabzuni
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Dhananjay Vaidya
- GeneSTAR Research Center, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeroen Van der Grond
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dennis Van der Meer
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marjolein M J Van Donkelaar
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Kristel R Van Eijk
- Brain Center Rudolf Magnus, Human Neurogenetics Unit, UMC Utrecht, Utrecht, the Netherlands
| | - Theo G M Van Erp
- Department of Psychiatry and Human Behavior, University of California-Irvine, Irvine, California, USA
| | - Daan Van Rooij
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Esther Walton
- Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Lars T Westlye
- NORMENT - KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- NORMENT - KG Jebsen Centre, Department of Psychology, University of Oslo, Oslo, Norway
| | - Christopher D Whelan
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- The Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Beverly G Windham
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Anderson M Winkler
- Department of Psychiatry, Yale University, New Haven, Connecticut, USA
- FMRIB Centre, University of Oxford, Oxford, UK
| | - Girma Woldehawariat
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health Intramural Research Program, National Institutes of Health, Bethesda, Maryland, USA
| | - Christiane Wolf
- University of Wuerzburg, Department of Psychiatry, Psychosomatics and Psychotherapy, Wuerzburg, Germany
| | - Thomas Wolfers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Bing Xu
- MRC-SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Lisa R Yanek
- GeneSTAR Research Center, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jingyun Yang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Alex Zijdenbos
- Biospective Inc, Montreal, Quebec, Canada, Montréal, Québec, Canada
| | - Marcel P Zwiers
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Donders Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, The Netherlands
| | - Ingrid Agartz
- NORMENT - KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Research and Development, Diakonhjemmet Hospital, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Neelum T Aggarwal
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, Illinois, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Laura Almasy
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine Brownsville/Edinburg/San Antonio, Texas, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - David Ames
- National Ageing Research Institute, Royal Melbourne Hospital, Melbourne, Australia
- Academic Unit for Psychiatry of Old Age, University of Melbourne, Melbourne, Australia
| | - Philippe Amouyel
- Lille University, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk factors and molecular determinants of aging-related diseases, Lille, France
| | - Ole A Andreassen
- NORMENT - KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- NORMENT - KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Sampath Arepalli
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Amelia A Assareh
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Sandra Barral
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| | - Mark E Bastin
- Brain Research Imaging Centre, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Department of Neuroimaging Sciences, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Diane M Becker
- GeneSTAR Research Center, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James T Becker
- Departments of Psychiatry, Neurology, and Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine Brownsville/Edinburg/San Antonio, Texas, USA
| | - Hans van Bokhoven
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Dorret I Boomsma
- Biological Psychology, Neuroscience Campus Amsterdam, Vrije Universiteit University and Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | - Henry Brodaty
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
- Dementia Collaborative Research Centre - Assessment and Better Care, UNSW, Sydney, Australia
| | - Rachel M Brouwer
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht, the Netherlands
| | - Han G Brunner
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Randy L Buckner
- Department of Psychiatry, Massachusetts General Hospital, Boston, Masschusetts, USA
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Karakter Child and Adolescent Psychiatry University Center, Nijmegen, the Netherlands
| | - Kazima B Bulayeva
- Department of Evolution and Genetics, Dagestan State University, Makhachkala, Dagestan, Russia
| | - Wiepke Cahn
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht, the Netherlands
| | - Vince D Calhoun
- The Mind Research Network and LBERI, Albuquerque, New Mexico, USA
- Department of ECE, University of New Mexico, Albuquerque, New Mexico, USA
| | - Dara M Cannon
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health Intramural Research Program, National Institutes of Health, Bethesda, Maryland, USA
- Centre for Neuroimaging and Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | | | - Christopher Chen
- Department of Pharmacology, National University of Singapore, Singapore
- Memory Aging and Cognition Centre (MACC), National University Health System, Singapore
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Academic Medicine Research Institute, Duke-NUS Graduate Medical School, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sven Cichon
- Division of Medical Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Aiden Corvin
- Psychosis Research Group, Department of Psychiatry and Trinity Translational Medicine Institute, Trinity College Dublin
| | - Benedicto Crespo-Facorro
- Department of Medicine and Psychiatry, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria-IDIVAL, Santander, Spain
- CIBERSAM (Centro Investigación Biomédica en Red Salud Mental), Santander, Spain
| | - Joanne E Curran
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine Brownsville/Edinburg/San Antonio, Texas, USA
| | - Michael Czisch
- Max Planck Institute of Psychiatry, Department of Translational Research in Psychiatry, Munich, Germany
| | - Anders M Dale
- Center for Multimodal Imaging and Genetics, University of California, San Diego, California, USA
- Department of Neurosciences, University of California, San Diego, California, USA
- Department of Radiology, University of California, San Diego, California, USA
- Department of Psychiatry, University of California, San Diego, California, USA
- Department of Cognitive Science, University of California, San Diego, California, USA
| | - Gareth E Davies
- Avera Institute for Human Genetics, Sioux Falls, South Dakota, USA
- Program in Translational NeuroPsychiatric Genomics, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Eco J C De Geus
- Biological Psychology, Neuroscience Campus Amsterdam, Vrije Universiteit University and Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | - Philip L De Jager
- Harvard Medical School, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Greig I de Zubicaray
- Faculty of Health and Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia
| | - Norman Delanty
- The Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Neurology Division, Beaumont Hospital, Dublin, 9, Ireland
| | - Chantal Depondt
- Department of Neurology, Hopital Erasme, Universite Libre de Bruxelles, Brussels, Belgium
| | - Anita L DeStefano
- Framingham Heart Study, Framingham, Massachusetts, USA
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Allissa Dillman
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Srdjan Djurovic
- NORMENT - KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Gary Donohoe
- Cognitive Genetics and Cognitive Therapy Group, Neuroimaging, Cognition and Genomics Centre (NICOG) and NCBES Galway Neuroscience Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, Ireland
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Trinity College Institute of Psychiatry, Trinity College Dublin, Dublin 8, Ireland
| | - Wayne C Drevets
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health Intramural Research Program, National Institutes of Health, Bethesda, Maryland, USA
- Janssen Research and Development, LLC, Titusville, New Jersey, USA
| | - Ravi Duggirala
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine Brownsville/Edinburg/San Antonio, Texas, USA
| | - Thomas D Dyer
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine Brownsville/Edinburg/San Antonio, Texas, USA
| | - Susanne Erk
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, CCM, Berlin, Germany
| | - Thomas Espeseth
- NORMENT - KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- NORMENT - KG Jebsen Centre, Department of Psychology, University of Oslo, Oslo, Norway
| | - Denis A Evans
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, Illinois, USA
| | - Iryna O Fedko
- Biological Psychology, Neuroscience Campus Amsterdam, Vrije Universiteit University and Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | - Guillén Fernández
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Luigi Ferrucci
- Intramural Research Program of the National Institute on Aging, Baltimore, Maryland, USA
| | - Simon E Fisher
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Debra A Fleischman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Ian Ford
- Robertson Center for Biostatistics, University of Glasgow, Glasgow, UK
| | - Tatiana M Foroud
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Peter T Fox
- University of Texas Health Science Center, San Antonio, Texas, USA
| | - Clyde Francks
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Masaki Fukunaga
- Division of Cerebral Integration, National Institute for Physiological Sciences, Aichi, Japan
| | - J Raphael Gibbs
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - David C Glahn
- Department of Psychiatry, Yale University, New Haven, Connecticut, USA
- Olin Neuropsychiatric Research Center, Hartford, Connecticut, USA
| | - Randy L Gollub
- Department of Psychiatry, Massachusetts General Hospital, Boston, Masschusetts, USA
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, North Carolina, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Harald H H Göring
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine Brownsville/Edinburg/San Antonio, Texas, USA
| | - Hans J Grabe
- Department of Psychiatry, University Medicine Greifswald, Greifswald, Germany
| | - Robert C Green
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Oliver Gruber
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University, Heidelberg, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Sebastian Guelfi
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Narelle K Hansell
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - John Hardy
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Catharina A Hartman
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ryota Hashimoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Katrin Hegenscheid
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, CCM, Berlin, Germany
| | - Stephanie Le Hellard
- NORMENT - KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Dena G Hernandez
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Dirk J Heslenfeld
- Department of Psychology, VU University Amsterdam, Amsterdam, the Netherlands
| | - Beng-Choon Ho
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
| | - Pieter J Hoekstra
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Wolfgang Hoffmann
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Florian Holsboer
- Max Planck Institute of Psychiatry, Department of Translational Research in Psychiatry, Munich, Germany
- HMNC Brain Health, Munich, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Norbert Hosten
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Jouke-Jan Hottenga
- Biological Psychology, Neuroscience Campus Amsterdam, Vrije Universiteit University and Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | - Hilleke E Hulshoff Pol
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht, the Netherlands
| | - Masashi Ikeda
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - M Kamran Ikram
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Pharmacology, National University of Singapore, Singapore
- Memory Aging and Cognition Centre (MACC), National University Health System, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Academic Medicine Research Institute, Duke-NUS Graduate Medical School, Singapore
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Robert Johnson
- NICHD Brain and Tissue Bank for Developmental Disorders, University of Maryland Medical School, Baltimore, Maryland, USA
| | - Erik G Jönsson
- NORMENT - KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- FMRIB Centre, University of Oxford, Oxford, UK
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - René S Kahn
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht, the Netherlands
| | - Ryota Kanai
- School of Psychology, University of Sussex, Brighton, UK
- Institute of Cognitive Neuroscience, University College London, London, UK
- Department of Neuroinformatics, Araya Brain Imaging, Tokyo, Japan
| | | | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - John B Kwok
- Neuroscience Research Australia, Sydney, Australia
- School of Medical Sciences, UNSW, Sydney, Australia
| | - Stephen M Lawrie
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Hervé Lemaître
- NSERM Unit 1000 ″Neuroimaging and Psychiatry″, University Paris Sud, University Paris Descartes, Paris, France
- Maison de Solenn, Adolescent Psychopathology and Medicine Department, APHP Hospital Cochin, Paris, France
| | - Xinmin Liu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health Intramural Research Program, National Institutes of Health, Bethesda, Maryland, USA
- Columbia University Medical Center, New York, New York, USA
| | - Dan L Longo
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - W T Longstreth
- Department of Neurology, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, UK
- NIHR Dementia Biomedical Research Unit, King's College London, London, UK
| | - Oliver Martinez
- Imaging of Dementia and Aging (IDeA) Laboratory, Department of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, California, USA
| | - Jean-Luc Martinot
- NSERM Unit 1000 ″Neuroimaging and Psychiatry″, University Paris Sud, University Paris Descartes, Paris, France
- Maison de Solenn, Adolescent Psychopathology and Medicine Department, APHP Hospital Cochin, Paris, France
| | - Venkata S Mattay
- Lieber Institute for Brain Development, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Colm McDonald
- Centre for Neuroimaging and Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Andrew M McIntosh
- Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh, UK
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Katie L McMahon
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | - Francis J McMahon
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health Intramural Research Program, National Institutes of Health, Bethesda, Maryland, USA
| | - Patrizia Mecocci
- Section of Gerontology and Geriatrics, Department of Medicine, University of Perugia, Perugia, Italy
| | - Ingrid Melle
- NORMENT - KG Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- NORMENT - KG Jebsen Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Andreas Meyer-Lindenberg
- Central Institute of Mental Health, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Sebastian Mohnke
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, CCM, Berlin, Germany
| | | | - Derek W Morris
- Cognitive Genetics and Cognitive Therapy Group, Neuroimaging, Cognition and Genomics Centre (NICOG) and NCBES Galway Neuroscience Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, Ireland
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Trinity College Institute of Psychiatry, Trinity College Dublin, Dublin 8, Ireland
| | - Thomas H Mosley
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Thomas W Mühleisen
- Centre for Neuroimaging and Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Department of Translational Research in Psychiatry, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- University of Liverpool, Institute of Translational Medicine, Liverpool, UK
| | - Michael A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (DZHK e.V.), partner site Greifswald, Germany
| | - Thomas E Nichols
- FMRIB Centre, University of Oxford, Oxford, UK
- Department of Statistics and Warwick Manufacturing Group, University of Warwick, Coventry, UK
| | - Wiro J Niessen
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
- Department of Medical Informatics Erasmus MC, Rotterdam, the Netherlands
- Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
| | - Lars Nyberg
- Department of Integrative Medical Biology and Umeå center for Functional Brain Imaging, Umeå University, Umeå, Sweden
| | - Kazutaka Ohi
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Rene L Olvera
- University of Texas Health Science Center, San Antonio, Texas, USA
| | - Roel A Ophoff
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht, the Netherlands
- Center for Neurobehavioral Genetics, University of California, Los Angeles, California, USA
| | - Massimo Pandolfo
- Department of Neurology, Hopital Erasme, Universite Libre de Bruxelles, Brussels, Belgium
| | - Tomas Paus
- Rotman Research Institute, University of Toronto, Toronto, Canada
- Departments of Psychology and Psychiatry, University of Toronto, Canada
- Child Mind Institute, New York, New York, USA
| | - Zdenka Pausova
- Hospital for Sick Children, University of Toronto, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada
| | - Brenda W J H Penninx
- Department of Psychiatry, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - G Bruce Pike
- Department of Radiology, University of Calgary, Calgary, Canada
- Department of Clinical Neuroscience, University of Calgary, Calgary, Canada
| | - Steven G Potkin
- Department of Psychiatry and Human Behavior, University of California-Irvine, Irvine, California, USA
| | - Bruce M Psaty
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Health Services, University of Washington, Seattle, Washington, USA
- Group Health Research Institute, Group Health, Seattle, Washington, USA
| | - Simone Reppermund
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
- Department of Developmental Disability Neuropsychiatry, School of Psychiatry, UNSW Medicine, Australia
| | - Marcella Rietschel
- Central Institute of Mental Health, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Joshua L Roffman
- Department of Psychiatry, Massachusetts General Hospital, Boston, Masschusetts, USA
| | - Nina Romanczuk-Seiferth
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, CCM, Berlin, Germany
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Pediatrics at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Mina Ryten
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Ralph L Sacco
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Department of Neurology, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Department of Epidemiology and Public Health Sciences, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Evelyn F. McKnight Brain Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, Australia
| | - Andrew J Saykin
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Reinhold Schmidt
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Austria, Graz, Austria
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, Australia
- School of Medical Sciences, UNSW, Sydney, Australia
| | | | - Andy Simmons
- Department of Neuroimaging, Institute of Psychiatry, King's College London, London, UK
- Biomedical Research Centre for Mental Health, King's College London, London, UK
- Biomedical Research Unit for Dementia, King's College London, London, UK
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Sanjay M Sisodiya
- UCL Institute of Neurology, London, United Kingdom and Epilepsy Society, Bucks, UK
| | - Colin Smith
- MRC Edinburgh Brain Bank, University of Edinburgh, Academic Department of Neuropathology, Centre for Clinical Brain Sciences, Edinburgh, UK
| | - Jordan W Smoller
- Department of Psychiatry, Massachusetts General Hospital, Boston, Masschusetts, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Hilkka Soininen
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
- Neurocentre Neurology, Kuopio University Hospital, Finland
| | - Velandai Srikanth
- Department of Medicine, Peninsula Health and Monash University, Melbourne, Australia
| | - Vidar M Steen
- NORMENT - KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - David J Stott
- Institute of Cardiovascular and Medical Sciences, Faculty of Medicine, University of Glasgow, Glasgow, UK
| | - Jessika E Sussmann
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Henning Tiemeier
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Arthur W Toga
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Bryan J Traynor
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Juan Troncoso
- Brain Resource Center, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Christophe Tzourio
- Institute for Neurodegenerative Disorders, UMR 5293, CEA, CNRS, Université de Bordeaux, France
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Maria C Valdés Hernández
- Brain Research Imaging Centre, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Department of Neuroimaging Sciences, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | | | - Aad Van der Lugt
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Nic J A Van der Wee
- Department of Psychiatry and Leiden Institute for Brain and Cognition, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Neeltje E M Van Haren
- Brain Center Rudolf Magnus, Department of Psychiatry, UMC Utrecht, Utrecht, the Netherlands
| | - Dennis Van T Ent
- Biological Psychology, Neuroscience Campus Amsterdam, Vrije Universiteit University and Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | - Marie-Jose Van Tol
- Neuroimaging Centre, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Badri N Vardarajan
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| | - Dick J Veltman
- Department of Psychiatry, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, CCM, Berlin, Germany
| | - Joanna M Wardlaw
- Brain Research Imaging Centre, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Department of Neuroimaging Sciences, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Thomas H Wassink
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Michael E Weale
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Baltimore, Maryland, USA
- Departments of Psychiatry, Neurology, Neuroscience and the Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael W Weiner
- Center for Imaging of Neurodegenerative Disease, San Francisco VA Medical Center, University of California, San Francisco, California, USA
| | - Wei Wen
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Eric Westman
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Tonya White
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Tien Y Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Department of Evolution and Genetics, Dagestan State University, Makhachkala, Dagestan, Russia
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Clinton B Wright
- Department of Neurology, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Department of Epidemiology and Public Health Sciences, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Evelyn F. McKnight Brain Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - H Ronald Zielke
- NICHD Brain and Tissue Bank for Developmental Disorders, University of Maryland Medical School, Baltimore, Maryland, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, Psychology, University of Edinburgh, Edinburgh, UK
| | - Charles DeCarli
- Imaging of Dementia and Aging (IDeA) Laboratory, Department of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, California, USA
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry, Medical University Graz, Graz, Austria
| | | | - Anton J M De Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | - Lenore J Launer
- Intramural Research Program, NIA, NIH, Bethesda, Maryland, USA
| | - Gunter Schumann
- MRC-SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Myriam Fornage
- Institute of Molecular Medicine and Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Barbara Franke
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Stéphanie Debette
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
- Lieber Institute for Brain Development, Baltimore, Maryland, USA
- Department of Neurology, Bordeaux University Hospital, Bordeaux, France
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
- Department of Neurology, Erasmus MC, Rotterdam, the Netherlands
| | - Paul M Thompson
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| |
Collapse
|