3051
|
Replogle JM, Chan G, White CC, Raj T, Winn PA, Evans DA, Sperling RA, Chibnik LB, Bradshaw EM, Schneider JA, Bennett DA, De Jager PL. A TREM1 variant alters the accumulation of Alzheimer-related amyloid pathology. Ann Neurol 2015; 77:469-77. [PMID: 25545807 DOI: 10.1002/ana.24337] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 11/23/2014] [Accepted: 12/21/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Genome-wide association studies have linked variants in TREM2 (triggering receptor expressed on myeloid cells 2) and TREML2 with Alzheimer disease (AD) and AD endophenotypes. Here, we pursue a targeted analysis of the TREM locus in relation to cognitive decline and pathological features of AD. METHODS Clinical, cognitive, and neuropathological phenotypes were collected in 3 prospective cohorts on aging (n = 3,421 subjects). Our primary analysis was an association with neuritic plaque pathology. To functionally characterize the associated variants, we used flow cytometry to measure TREM1 expression on monocytes. RESULTS We provide evidence that an intronic variant, rs6910730(G) , in TREM1, is associated with an increased burden of neuritic plaques (p = 3.7 × 10(-4) ), diffuse plaques (p = 4.1 × 10(-3) ), and Aβ density (p = 2.6 × 10(-3) ) as well as an increased rate of cognitive decline (p = 5.3 × 10(-3) ). A variant upstream of TREM2, rs7759295(C) , is independently associated with an increased tau tangle density (p = 4.9 × 10(-4) ), an increased burden of neurofibrillary tangles (p = 9.1 × 10(-3) ), and an increased rate of cognitive decline (p = 2.3 × 10(-3) ). Finally, a cytometric analysis shows that the TREM1 rs6910730(G) allele is associated with decreased TREM1 expression on the surface of myeloid cells (p = 1.7 × 10(-3) ). INTERPRETATION We provide evidence that 2 common variants within the TREM locus are associated with pathological features of AD and aging-related cognitive decline. Our evidence suggests that these variants are likely to be independent of known AD variants and that they may work through an alteration of myeloid cell function.
Collapse
Affiliation(s)
- Joseph M Replogle
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, Boston, MA; Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA; Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3052
|
Chibnik LB, Yu L, Eaton ML, Srivastava G, Schneider JA, Kellis M, Bennett DA, De Jager PL. Alzheimer's loci: epigenetic associations and interaction with genetic factors. Ann Clin Transl Neurol 2015; 2:636-47. [PMID: 26125039 PMCID: PMC4479524 DOI: 10.1002/acn3.201] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/20/2015] [Accepted: 03/06/2015] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE We explore the role of DNA methylation in Alzheimer's disease (AD). To elucidate where DNA methylation falls along the causal pathway linking risk factors to disease, we examine causal models to assess its role in the pathology of AD. METHODS DNA methylation profiles were generated in 740 brain samples using the Illumina HumanMet450K beadset. We focused our analysis on CpG sites from 11 AD susceptibility gene regions. The primary outcome was a quantitative measure of neuritic amyloid plaque (NP), a key early element of AD pathology. We tested four causal models: (1) independent associations, (2) CpG mediating the association of a variant, (3) reverse causality, and (4) genetic variant by CpG interaction. RESULTS Six genes regions (17 CpGs) showed evidence of CpG associations with NP, independent of genetic variation - BIN1 (5), CLU (5), MS4A6A (3), ABCA7 (2), CD2AP (1), and APOE (1). Together they explained 16.8% of the variability in NP. An interaction effect was seen in the CR1 region for two CpGs, cg10021878 (P = 0.01) and cg05922028 (P = 0.001), in relation to NP. In both cases, subjects with the risk allele rs6656401(AT) (/) (AA) display more methylation being associated with more NP burden, whereas subjects with the rs6656401(TT) protective genotype have an inverse association with more methylation being associated with less NP. INTERPRETATION These observations suggest that, within known AD susceptibility loci, methylation is related to pathologic processes of AD and may play a largely independent role by influencing gene expression in AD susceptibility loci.
Collapse
Affiliation(s)
- Lori B Chibnik
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital Boston, Massachusetts, 02115 ; Department of Neurology, Harvard Medical School Boston, Massachusetts, 02115 ; Medical and Population Genetics, Broad Institute of MIT and Harvard Cambridge, Massachusetts, 02142 ; Department of Epidemiology, Harvard T.H. Chan School of Public Health Boston, Massachusetts, 02115
| | - Lei Yu
- Medical and Population Genetics, Broad Institute of MIT and Harvard Cambridge, Massachusetts, 02142 ; Rush Alzheimer's Disease Center, Rush University Medical Center Chicago, Illinois, 60612
| | - Matthew L Eaton
- Medical and Population Genetics, Broad Institute of MIT and Harvard Cambridge, Massachusetts, 02142 ; Computer Science and Artificial Intelligence Laboratory, MIT Cambridge, Massachusetts, 02124
| | - Gyan Srivastava
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital Boston, Massachusetts, 02115 ; Department of Neurology, Harvard Medical School Boston, Massachusetts, 02115 ; Medical and Population Genetics, Broad Institute of MIT and Harvard Cambridge, Massachusetts, 02142
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center Chicago, Illinois, 60612
| | - Manolis Kellis
- Medical and Population Genetics, Broad Institute of MIT and Harvard Cambridge, Massachusetts, 02142 ; Computer Science and Artificial Intelligence Laboratory, MIT Cambridge, Massachusetts, 02124
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center Chicago, Illinois, 60612
| | - Philip L De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital Boston, Massachusetts, 02115 ; Department of Neurology, Harvard Medical School Boston, Massachusetts, 02115 ; Medical and Population Genetics, Broad Institute of MIT and Harvard Cambridge, Massachusetts, 02142
| |
Collapse
|
3053
|
Genetic risk of neurodegenerative diseases is associated with mild cognitive impairment and conversion to dementia. Alzheimers Dement 2015; 11:1277-85. [PMID: 25916564 DOI: 10.1016/j.jalz.2014.12.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 10/09/2014] [Accepted: 12/18/2014] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Neurodegenerative diseases are a major cause of cognitive impairment and can ultimately lead to dementia. Genome-wide association studies have uncovered many genetic variants conferring risk of neurodegenerative diseases, but their role in cognitive impairment remains unexplored. METHODS In the prospective, population-based Rotterdam Study, 3605 nondemented persons aged ≥55 years were genotyped, screened for mild cognitive impairment (MCI) in 2002 to 2005 and underwent continuous follow-up for dementia until 2012. Weighted polygenic risk scores of genetic variants for Alzheimer's disease (AD), Parkinson's disease (PD), and the frontotemporal lobar degeneration/amyotrophic lateral sclerosis disease spectrum (FTLD/ALS) were constructed and investigated for association with MCI and the subsequent conversion to dementia. RESULTS In total, 360 (10.0%) persons had MCI, of whom 147 (4.1%) were amnestic and 213 (5.9%) nonamnestic. The AD risk score was associated with both MCI subtypes (odds ratio for all MCI 1.15 [95% CI, 1.03-1.28]), whereas PD and FTLD/ALS risk scores were associated only with nonamnestic MCI (odds ratios 1.15 [1.00-1.32] and 1.19 [1.03-1.37], respectively). The AD risk score, but not PD and FTLD/ALS risk scores, was associated with an increased risk of dementia (hazard ratio 1.55 [1.37-1.77]). DISCUSSION Genetic evidence supports the view that multiple neurodegenerative pathways lead to MCI and that the subsequent conversion to dementia, primarily of the AD subtype, is mainly due to the AD pathway(s).
Collapse
|
3054
|
Evans DM, Davey Smith G. Mendelian Randomization: New Applications in the Coming Age of Hypothesis-Free Causality. Annu Rev Genomics Hum Genet 2015; 16:327-50. [PMID: 25939054 DOI: 10.1146/annurev-genom-090314-050016] [Citation(s) in RCA: 338] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mendelian randomization (MR) is an approach that uses genetic variants associated with a modifiable exposure or biological intermediate to estimate the causal relationship between these variables and a medically relevant outcome. Although it was initially developed to examine the relationship between modifiable exposures/biomarkers and disease, its use has expanded to encompass applications in molecular epidemiology, systems biology, pharmacogenomics, and many other areas. The purpose of this review is to introduce MR, the principles behind the approach, and its limitations. We consider some of the new applications of the methodology, including informing drug development, and comment on some promising extensions, including two-step, two-sample, and bidirectional MR. We show how these new methods can be combined to efficiently examine causality in complex biological networks and provide a new framework to data mine high-dimensional studies as we transition into the age of hypothesis-free causality.
Collapse
Affiliation(s)
- David M Evans
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland 4102, Australia;
| | | |
Collapse
|
3055
|
Debette S, Ibrahim Verbaas CA, Bressler J, Schuur M, Smith A, Bis JC, Davies G, Wolf C, Gudnason V, Chibnik LB, Yang Q, deStefano AL, de Quervain DJF, Srikanth V, Lahti J, Grabe HJ, Smith JA, Priebe L, Yu L, Karbalai N, Hayward C, Wilson JF, Campbell H, Petrovic K, Fornage M, Chauhan G, Yeo R, Boxall R, Becker J, Stegle O, Mather KA, Chouraki V, Sun Q, Rose LM, Resnick S, Oldmeadow C, Kirin M, Wright AF, Jonsdottir MK, Au R, Becker A, Amin N, Nalls MA, Turner ST, Kardia SLR, Oostra B, Windham G, Coker LH, Zhao W, Knopman DS, Heiss G, Griswold ME, Gottesman RF, Vitart V, Hastie ND, Zgaga L, Rudan I, Polasek O, Holliday EG, Schofield P, Choi SH, Tanaka T, An Y, Perry RT, Kennedy RE, Sale MM, Wang J, Wadley VG, Liewald DC, Ridker PM, Gow AJ, Pattie A, Starr JM, Porteous D, Liu X, Thomson R, Armstrong NJ, Eiriksdottir G, Assareh AA, Kochan NA, Widen E, Palotie A, Hsieh YC, Eriksson JG, Vogler C, van Swieten JC, Shulman JM, Beiser A, Rotter J, Schmidt CO, Hoffmann W, Nöthen MM, Ferrucci L, Attia J, Uitterlinden AG, Amouyel P, Dartigues JF, Amieva H, Räikkönen K, Garcia M, et alDebette S, Ibrahim Verbaas CA, Bressler J, Schuur M, Smith A, Bis JC, Davies G, Wolf C, Gudnason V, Chibnik LB, Yang Q, deStefano AL, de Quervain DJF, Srikanth V, Lahti J, Grabe HJ, Smith JA, Priebe L, Yu L, Karbalai N, Hayward C, Wilson JF, Campbell H, Petrovic K, Fornage M, Chauhan G, Yeo R, Boxall R, Becker J, Stegle O, Mather KA, Chouraki V, Sun Q, Rose LM, Resnick S, Oldmeadow C, Kirin M, Wright AF, Jonsdottir MK, Au R, Becker A, Amin N, Nalls MA, Turner ST, Kardia SLR, Oostra B, Windham G, Coker LH, Zhao W, Knopman DS, Heiss G, Griswold ME, Gottesman RF, Vitart V, Hastie ND, Zgaga L, Rudan I, Polasek O, Holliday EG, Schofield P, Choi SH, Tanaka T, An Y, Perry RT, Kennedy RE, Sale MM, Wang J, Wadley VG, Liewald DC, Ridker PM, Gow AJ, Pattie A, Starr JM, Porteous D, Liu X, Thomson R, Armstrong NJ, Eiriksdottir G, Assareh AA, Kochan NA, Widen E, Palotie A, Hsieh YC, Eriksson JG, Vogler C, van Swieten JC, Shulman JM, Beiser A, Rotter J, Schmidt CO, Hoffmann W, Nöthen MM, Ferrucci L, Attia J, Uitterlinden AG, Amouyel P, Dartigues JF, Amieva H, Räikkönen K, Garcia M, Wolf PA, Hofman A, Longstreth WT, Psaty BM, Boerwinkle E, DeJager PL, Sachdev PS, Schmidt R, Breteler MMB, Teumer A, Lopez OL, Cichon S, Chasman DI, Grodstein F, Müller-Myhsok B, Tzourio C, Papassotiropoulos A, Bennett DA, Ikram MA, Deary IJ, van Duijn CM, Launer L, Fitzpatrick AL, Seshadri S, Mosley TH. Genome-wide studies of verbal declarative memory in nondemented older people: the Cohorts for Heart and Aging Research in Genomic Epidemiology consortium. Biol Psychiatry 2015; 77:749-63. [PMID: 25648963 PMCID: PMC4513651 DOI: 10.1016/j.biopsych.2014.08.027] [Show More Authors] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 08/23/2014] [Accepted: 08/25/2014] [Indexed: 01/06/2023]
Abstract
BACKGROUND Memory performance in older persons can reflect genetic influences on cognitive function and dementing processes. We aimed to identify genetic contributions to verbal declarative memory in a community setting. METHODS We conducted genome-wide association studies for paragraph or word list delayed recall in 19 cohorts from the Cohorts for Heart and Aging Research in Genomic Epidemiology consortium, comprising 29,076 dementia- and stroke-free individuals of European descent, aged ≥45 years. Replication of suggestive associations (p < 5 × 10(-6)) was sought in 10,617 participants of European descent, 3811 African-Americans, and 1561 young adults. RESULTS rs4420638, near APOE, was associated with poorer delayed recall performance in discovery (p = 5.57 × 10(-10)) and replication cohorts (p = 5.65 × 10(-8)). This association was stronger for paragraph than word list delayed recall and in the oldest persons. Two associations with specific tests, in subsets of the total sample, reached genome-wide significance in combined analyses of discovery and replication (rs11074779 [HS3ST4], p = 3.11 × 10(-8), and rs6813517 [SPOCK3], p = 2.58 × 10(-8)) near genes involved in immune response. A genetic score combining 58 independent suggestive memory risk variants was associated with increasing Alzheimer disease pathology in 725 autopsy samples. Association of memory risk loci with gene expression in 138 human hippocampus samples showed cis-associations with WDR48 and CLDN5, both related to ubiquitin metabolism. CONCLUSIONS This largest study to date exploring the genetics of memory function in ~40,000 older individuals revealed genome-wide associations and suggested an involvement of immune and ubiquitin pathways.
Collapse
Affiliation(s)
- Stéphanie Debette
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Institut National de la Santé et de la Recherche Médicale, Epidemiology, University of Bordeaux; Department of Neurology, University Hospital of Bordeaux, Bordeaux, France.
| | - Carla A Ibrahim Verbaas
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Department of Neurology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
| | - Maaike Schuur
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Department of Neurology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Albert Smith
- Icelandic Heart Association, Kopavogur; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington
| | - Gail Davies
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Department of Psychology, The University of Edinburgh; Medical Genetics Section, Molecular Medicine Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Lori B Chibnik
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Boston
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Anita L deStefano
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Dominique J F de Quervain
- Psychiatric University Clinics and Department of Psychology, Division of Cognitive Neuroscience, University of Basel, Basel, Switzerland
| | - Velandai Srikanth
- Stroke and Ageing Research Centre, Southern Clinical School, Department of Medicine, Monash University, Melbourne; Menzies Research Institute Tasmania, University of Tasmania, Hobart, Australia
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki; Folkhälsan Research Centre, Helsinki, Finland
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, HELIOS-Hospital Stralsund, Stralsund; German Center for Neurodegenerative Diseases, Site Rostock/ Greifswald, Rostock, Germany
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Lutz Priebe
- Institute of Human Genetics, Universitätsklinikum Bonn, Bonn, Germany
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | | | | | - James F Wilson
- Institute of Genetics and Molecular Medicine, University of Edinburgh, United Kingdom, Centre for Population Health Sciences, University of Edinburgh, United Kingdom
| | - Harry Campbell
- Division of General Neurology, Department of Neurology, Medical University and General Hospital of Graz, Austria
| | - Katja Petrovic
- Division of General Neurology, Department of Neurology, Medical University and General Hospital of Graz, Austria
| | - Myriam Fornage
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas; Institute of Molecular Medicine, University of Texas-Houston Health Science Center, Houston, Texas
| | - Ganesh Chauhan
- Institut National de la Santé et de la Recherche Médicale, Epidemiology, University of Bordeaux
| | - Robin Yeo
- Institut National de la Santé et de la Recherche Médicale, Epidemiology, University of Bordeaux
| | - Ruth Boxall
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh
| | - James Becker
- Departments of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Psychology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Oliver Stegle
- Max Planck Institute for Intelligent Systems, Tübingen, Germany; Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Medicine, University of New South Wales, Sydney, Australia
| | - Vincent Chouraki
- Institut National de la Santé et de la Recherche Médicale Unit 744, Institut Pasteur de Lille, and Université Lille Nord de France, Lille, France
| | - Qi Sun
- Department of Nutrition, Harvard School of Public Health; Channing Division of Network Medicine, Department of Medicine, Brigham and Women׳s Hospital and Harvard Medical School; Harvard Medical School, Brigham and Women׳s Hospital, Boston, Massachusetts
| | - Lynda M Rose
- Division of Preventive Medicine, Brigham and Women׳s Hospital, Boston, Massachusetts
| | - Susan Resnick
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Christopher Oldmeadow
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine, Newcastle, Australia; Public Health, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Mirna Kirin
- Institute of Genetics and Molecular Medicine, University of Edinburgh, United Kingdom, Centre for Population Health Sciences, University of Edinburgh, United Kingdom
| | - Alan F Wright
- Institute of Genetics and Molecular Medicine, University of Edinburgh, United Kingdom, Centre for Population Health Sciences, University of Edinburgh, United Kingdom
| | | | - Rhoda Au
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Albert Becker
- Institute of Neuropathology, Universitätsklinikum Bonn, Bonn, Germany
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Mike A Nalls
- Molecular Genetics Section , Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Stephen T Turner
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Ben Oostra
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Department of Clinical Genetics, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Gwen Windham
- Department of Medicine, Division of Geriatrics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Laura H Coker
- Division of Public Health Sciences and Neurology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | | | - Gerardo Heiss
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michael E Griswold
- Center of Biostatistics and Bioinformatics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Rebecca F Gottesman
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | - Lina Zgaga
- Medical Research Council Human Genetics Unit
| | - Igor Rudan
- Institute of Genetics and Molecular Medicine, University of Edinburgh, United Kingdom, Centre for Population Health Sciences, University of Edinburgh, United Kingdom
| | - Ozren Polasek
- Department of Public Health, Faculty of Medicine, University of Split, Split, Croatia
| | - Elizabeth G Holliday
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine, Newcastle, Australia
| | - Peter Schofield
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine, Newcastle, Australia; Department of Medicine, John Hunter Hospital, Newcastle, Australia
| | - Seung Hoan Choi
- Department of Biostatistics, Boston University School of Public Health, Boston
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Yang An
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Rodney T Perry
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Richard E Kennedy
- Division of Gerontology, Geriatrics, and Palliative Care, University of Alabama at Birmingham, Birmingham, Alabama
| | - Michèle M Sale
- Center for Public Health Genomics, Department of Medicine, Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - Jing Wang
- Department of Biostatistics, Boston University School of Public Health, Boston
| | - Virginia G Wadley
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David C Liewald
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Department of Psychology, The University of Edinburgh
| | - Paul M Ridker
- Harvard Medical School, Brigham and Women׳s Hospital, Boston, Massachusetts; Division of Preventive Medicine, Brigham and Women׳s Hospital, Boston, Massachusetts
| | - Alan J Gow
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Department of Psychology, The University of Edinburgh
| | - Alison Pattie
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - David Porteous
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Medical Genetics Section, Molecular Medicine Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Xuan Liu
- Department of Biostatistics, Boston University School of Public Health, Boston
| | - Russell Thomson
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Australia
| | - Nicola J Armstrong
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Medicine, University of New South Wales, Sydney, Australia; Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst; School of Mathematics & Statistics and Prince of Wales Clinical School, University of New South Wales, Sydney
| | | | - Arezoo A Assareh
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Medicine, University of New South Wales, Sydney, Australia; Neuroscience Research Australia and Primary Dementia Collaborative Research Centre-Assessment and Better Care, University of New South Wales, Sydney
| | - Nicole A Kochan
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Medicine, University of New South Wales, Sydney, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick New South Wales, Australia
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland, University of Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, Finland; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom; Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - Yi-Chen Hsieh
- Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Johan G Eriksson
- Folkhälsan Research Centre, Helsinki, Finland; National Institute for Health and Welfare, Helsinki, Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Helsinki University Central Hospital, Unit of General Practice, Helsinki, Vasa Central Hospital, Vasa, Finland
| | - Christian Vogler
- Psychiatric University Clinics and Department of Psychology, Division of Molecular Neuroscience, University of Basel, Basel, Switzerland
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Joshua M Shulman
- Departments of Neurology and Molecular and Human Genetics, Baylor College of Medicine and The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas
| | - Alexa Beiser
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Jerome Rotter
- Institute for Translational Genomics and Populaton Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California
| | | | - Wolfgang Hoffmann
- German Center for Neurodegenerative Diseases, Site Rostock/ Greifswald, Rostock, Germany; Section Epidemiology of Health Care and Community Health, Greifswald
| | - Markus M Nöthen
- Institute of Human Genetics, Department of Genomics, Life & Brain Research Center, University of Bonn, Bonn; German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - John Attia
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine, Newcastle, Australia; Public Health, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia; Department of Medicine, John Hunter Hospital, Newcastle, Australia
| | - Andre G Uitterlinden
- Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands; Department of Internal Medicine, Erasmus Medical Center University Medical Center, Rotterdam, the Netherlands
| | - Philippe Amouyel
- Institut National de la Santé et de la Recherche Médicale Unit 744, Institut Pasteur de Lille, and Université Lille Nord de France, Lille, France; Centre Hospitalier Régional Universitaire de Lille, Lille
| | - Jean-François Dartigues
- Institut National de la Santé et de la Recherche Médicale, Bordeaux University, Talence, France
| | - Hélène Amieva
- Institut National de la Santé et de la Recherche Médicale, Bordeaux University, Talence, France
| | | | - Melissa Garcia
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Bethesda, Maryland
| | - Philip A Wolf
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Albert Hofman
- Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands; Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - W T Longstreth
- Departments of Neurology, University of Washington; Epidemiology, University of Washington
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington; Epidemiology, University of Washington; Health Services, University of Washington; Group Health Research Institute, Group Health Cooperative, Seattle, Washington
| | - Eric Boerwinkle
- Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas
| | - Philip L DeJager
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Boston
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Medicine, University of New South Wales, Sydney, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick New South Wales, Australia
| | - Reinhold Schmidt
- Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Monique M B Breteler
- German Center for Neurodegenerative Diseases, Site Rostock/ Greifswald, Rostock, Germany; Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands; Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Population Health Sciences, University of Bonn, Bonn, Germany; Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, Massachusetts
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Oscar L Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; The Alzheimer׳s Disease Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sven Cichon
- Institute of Human Genetics, Universitätsklinikum Bonn, Bonn, Germany; Institute of Neuroscience and Medicine, Research Center Julich, Julich, Germany; Division of Medical Genetics, Department of Biomedicine, University of Basel, Switzerland
| | - Daniel I Chasman
- Harvard Medical School, Brigham and Women׳s Hospital, Boston, Massachusetts; Division of Preventive Medicine, Brigham and Women׳s Hospital, Boston, Massachusetts
| | - Francine Grodstein
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women׳s Hospital and Harvard Medical School; Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, Massachusetts
| | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christophe Tzourio
- Institut National de la Santé et de la Recherche Médicale, Epidemiology, University of Bordeaux; University Bordeaux Segalen, Bordeaux, France
| | - Andreas Papassotiropoulos
- Psychiatric University Clinics and Department of Psychology, Division of Molecular Neuroscience, University of Basel, Basel, Switzerland; Department Biozentrum, Life Sciences Training Facility, Basel, Switzerland
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - M Arfan Ikram
- Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands; Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Department of Radiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Department of Psychology, The University of Edinburgh
| | - Cornelia M van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands; Center for Medical Systems Biology, Netherlands Genomics Initiative, Leiden University Medical Center, Leiden, The Netherlands
| | - Lenore Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Bethesda, Maryland
| | | | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Thomas H Mosley
- Department of Medicine and Neurology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
3056
|
Improving Significance in Association Studies: a New Perspective for Association Studies Submitted to the Journal of Molecular Neuroscience. J Mol Neurosci 2015; 56:529-30. [DOI: 10.1007/s12031-015-0557-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/26/2015] [Indexed: 10/23/2022]
|
3057
|
Singleton AB, Traynor BJ. Genetics. For complex disease genetics, collaboration drives progress. Science 2015; 347:1422-3. [PMID: 25814571 DOI: 10.1126/science.aaa9838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA.
| | - Bryan J Traynor
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| |
Collapse
|
3058
|
Dienemann C, Coburger I, Mehmedbasic A, Andersen OM, Than ME. Mutants of Metal Binding Site M1 in APP E2 Show Metal Specific Differences in Binding of Heparin but Not of sorLA. Biochemistry 2015; 54:2490-9. [DOI: 10.1021/acs.biochem.5b00111] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Christian Dienemann
- Leibniz
Institute for Age Research, Fritz Lipmann Institute (FLI), Protein Crystallography Group, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Ina Coburger
- Leibniz
Institute for Age Research, Fritz Lipmann Institute (FLI), Protein Crystallography Group, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Arnela Mehmedbasic
- The
Lundbeck Foundation Research Center MIND, Danish Research Institute
of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership,
Department of Biomedicine, Aarhus University, Ole Worms Allé 3, DK-8000 Aarhus C, Denmark
| | - Olav M. Andersen
- The
Lundbeck Foundation Research Center MIND, Danish Research Institute
of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership,
Department of Biomedicine, Aarhus University, Ole Worms Allé 3, DK-8000 Aarhus C, Denmark
| | - Manuel E. Than
- Leibniz
Institute for Age Research, Fritz Lipmann Institute (FLI), Protein Crystallography Group, Beutenbergstrasse 11, 07745 Jena, Germany
| |
Collapse
|
3059
|
Calero M, Gómez-Ramos A, Calero O, Soriano E, Avila J, Medina M. Additional mechanisms conferring genetic susceptibility to Alzheimer's disease. Front Cell Neurosci 2015; 9:138. [PMID: 25914626 PMCID: PMC4391239 DOI: 10.3389/fncel.2015.00138] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/23/2015] [Indexed: 01/18/2023] Open
Abstract
Familial Alzheimer’s disease (AD), mostly associated with early onset, is caused by mutations in three genes (APP, PSEN1, and PSEN2) involved in the production of the amyloid β peptide. In contrast, the molecular mechanisms that trigger the most common late onset sporadic AD remain largely unknown. With the implementation of an increasing number of case-control studies and the upcoming of large-scale genome-wide association studies there is a mounting list of genetic risk factors associated with common genetic variants that have been associated with sporadic AD. Besides apolipoprotein E, that presents a strong association with the disease (OR∼4), the rest of these genes have moderate or low degrees of association, with OR ranging from 0.88 to 1.23. Taking together, these genes may account only for a fraction of the attributable AD risk and therefore, rare variants and epistastic gene interactions should be taken into account in order to get the full picture of the genetic risks associated with AD. Here, we review recent whole-exome studies looking for rare variants, somatic brain mutations with a strong association to the disease, and several studies dealing with epistasis as additional mechanisms conferring genetic susceptibility to AD. Altogether, recent evidence underlines the importance of defining molecular and genetic pathways, and networks rather than the contribution of specific genes.
Collapse
Affiliation(s)
- Miguel Calero
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Madrid, Spain ; Chronic Disease Programme, Instituto de Salud Carlos III Madrid, Spain ; Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center Madrid, Spain
| | - Alberto Gómez-Ramos
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Madrid, Spain ; Centro de Biología Molecular Severo Ochoa CSIC-UAM Madrid, Spain
| | - Olga Calero
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Madrid, Spain ; Chronic Disease Programme, Instituto de Salud Carlos III Madrid, Spain
| | - Eduardo Soriano
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Madrid, Spain ; University of Barcelona Barcelona, Spain
| | - Jesús Avila
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Madrid, Spain ; Centro de Biología Molecular Severo Ochoa CSIC-UAM Madrid, Spain
| | - Miguel Medina
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Madrid, Spain ; Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center Madrid, Spain
| |
Collapse
|
3060
|
Sun M, Chen L. Studying tauopathies in Drosophila: A fruitful model. Exp Neurol 2015; 274:52-7. [PMID: 25862286 DOI: 10.1016/j.expneurol.2015.03.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/06/2015] [Accepted: 03/31/2015] [Indexed: 11/26/2022]
Abstract
Tauopathies are a group of neurodegenerative disorders that include hereditary frontotemporal dementias (FTDs) such as FTD with parkinsonism linked to chromosome 17 (FTDP-17), as well as sporadic variants of FTDs like progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and Pick's disease. These diverse diseases all have in common the presence of abnormally phosphorylated tau aggregates. In this review, we will summarize key features of transgenic Drosophila models of tauopathies and a number of insights into disease mechanisms as well as therapeutic implications gained from the fruit fly models.
Collapse
Affiliation(s)
- Mingkuan Sun
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA; The Key Laboratory of Developmental Genes and Human Disease, Institute of Life Science, Southeast University, Nanjing 210009, China
| | - Liam Chen
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3061
|
Yang T, Wang J, Sun Q, Hibar DP, Jahanshad N, Liu L, Wang Y, Zhan L, Thompson PM, Ye J. Detecting Genetic Risk Factors for Alzheimer's Disease in Whole Genome Sequence Data via Lasso Screening. PROCEEDINGS. IEEE INTERNATIONAL SYMPOSIUM ON BIOMEDICAL IMAGING 2015; 2015:985-989. [PMID: 26413209 DOI: 10.1109/isbi.2015.7164036] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Genetic factors play a key role in Alzheimer's disease (AD). The Alzheimer's Disease Neuroimaging Initiative (ADNI) whole genome sequence (WGS) data offers new power to investigate mechanisms of AD by combining entire genome sequences with neuroimaging and clinical data. Here we explore the ADNI WGS SNP (single nucleotide polymorphism) data in depth and extract approximately six million valid SNP features. We investigate imaging genetics associations using Lasso regression-a widely used sparse learning technique. To solve the large-scale Lasso problem more efficiently, we employ a highly efficient screening rule for Lasso-called dual polytope projections (DPP)-to remove irrelevant features from the optimization problem. Experiments demonstrate that the DPP can effectively identify irrelevant features and leads to a 400× speedup. This allows us for the first time to run the compute-intensive model selection procedure called stability selection to rank SNPs that may affect the brain and AD risk.
Collapse
Affiliation(s)
- Tao Yang
- Dept. of Computer Science and Engineering, Arizona State Univ., Tempe, AZ, USA ; Center for Evolutionary Medicine and Informatics, The Biodesign Institute, Arizona State Univ., Tempe, AZ, USA
| | - Jie Wang
- Dept. of Computer Science and Engineering, Arizona State Univ., Tempe, AZ, USA ; Center for Evolutionary Medicine and Informatics, The Biodesign Institute, Arizona State Univ., Tempe, AZ, USA
| | - Qian Sun
- Dept. of Computer Science and Engineering, Arizona State Univ., Tempe, AZ, USA ; Center for Evolutionary Medicine and Informatics, The Biodesign Institute, Arizona State Univ., Tempe, AZ, USA
| | - Derrek P Hibar
- Imaging Genetics Center, Keck School of Medicine, Univ. of Southern California, Los Angeles, CA, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Keck School of Medicine, Univ. of Southern California, Los Angeles, CA, USA
| | - Li Liu
- Center for Evolutionary Medicine and Informatics, The Biodesign Institute, Arizona State Univ., Tempe, AZ, USA
| | - Yalin Wang
- Dept. of Computer Science and Engineering, Arizona State Univ., Tempe, AZ, USA
| | - Liang Zhan
- Imaging Genetics Center, Keck School of Medicine, Univ. of Southern California, Los Angeles, CA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Keck School of Medicine, Univ. of Southern California, Los Angeles, CA, USA
| | - Jieping Ye
- Dept. of Computational Medicine and Bioinformatics, Univ. of Michigan, Ann Arbor, MI, USA ; Dept. of Electrical Engineering and Computer Science, Univ. of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
3062
|
Sibener L, Zaganjor I, Snyder HM, Bain LJ, Egge R, Carrillo MC. Alzheimer's Disease prevalence, costs, and prevention for military personnel and veterans. Alzheimers Dement 2015; 10:S105-10. [PMID: 24924663 DOI: 10.1016/j.jalz.2014.04.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
By 2050, more than 13 million Americans of all ages are estimated to be living with Alzheimer's disease (AD), and the aggregate costs of care will swell to approximately $1.2 trillion. The rapidly climbing number of those affected with AD includes a growing population of aging military veterans affected who may have an added risk for the disease as a consequence of traumatic brain injury, posttraumatic stress disorder, and/or service-related injuries. The increasing number of individuals, the long duration of disability, and the rising cost of care for AD and other dementia to our society are important public health challenges facing many older adults. These challenges are further compounded by a burgeoning military veteran population that is much younger, with an increased risk of AD and other dementia, and who may experience decades-long periods of disability and care. This outlook underscores the critical need for investments in research at the federal and international levels to accelerate the pace of progress in developing breakthrough discoveries that will change the trajectory of AD and related dementia.
Collapse
Affiliation(s)
- Leslie Sibener
- Medical and Scientific Relations Division, Alzheimer's Association, Chicago, IL, USA
| | - Ibrahim Zaganjor
- Medical and Scientific Relations Division, Alzheimer's Association, Chicago, IL, USA
| | - Heather M Snyder
- Medical and Scientific Relations Division, Alzheimer's Association, Chicago, IL, USA
| | - Lisa J Bain
- Independent Science Writer, Philadelphia, PA, USA
| | - Robert Egge
- Public Policy and Advocacy Division, Alzheimer's Association, Washington, DC, USA
| | - Maria C Carrillo
- Medical and Scientific Relations Division, Alzheimer's Association, Chicago, IL, USA.
| |
Collapse
|
3063
|
Yokoyama JS, Bonham LW, Sears RL, Klein E, Karydas A, Kramer JH, Miller BL, Coppola G. Decision tree analysis of genetic risk for clinically heterogeneous Alzheimer's disease. BMC Neurol 2015; 15:47. [PMID: 25880661 PMCID: PMC4459447 DOI: 10.1186/s12883-015-0304-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 03/12/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Heritability of Alzheimer's disease (AD) is estimated at 74% and genetic contributors have been widely sought. The ε4 allele of apolipoprotein E (APOE) remains the strongest common risk factor for AD, with numerous other common variants contributing only modest risk for disease. Variability in clinical presentation of AD, which is typically amnestic (AmnAD) but can less commonly involve visuospatial, language and/or dysexecutive syndromes (atypical or AtAD), further complicates genetic analyses. Taking a multi-locus approach may increase the ability to identify individuals at highest risk for any AD syndrome. In this study, we sought to develop and investigate the utility of a multi-variant genetic risk assessment on a cohort of phenotypically heterogeneous patients with sporadic AD clinical diagnoses. METHODS We genotyped 75 variants in our cohort and, using a two-staged study design, we developed a 17-marker AD risk score in a Discovery cohort (n = 59 cases, n = 133 controls) then assessed its utility in a second Validation cohort (n = 126 cases, n = 150 controls). We also performed a data-driven decision tree analysis to identify genetic and/or demographic criteria that are most useful for accurately differentiating all AD cases from controls. RESULTS We confirmed APOE ε4 as a strong risk factor for AD. A 17-marker risk panel predicted AD significantly better than APOE genotype alone (P < 0.00001) in the Discovery cohort, but not in the Validation cohort. In decision tree analyses, we found that APOE best differentiated cases from controls only in AmnAD but not AtAD. In AtAD, HFE SNP rs1799945 was the strongest predictor of disease; variation in HFE has previously been implicated in AD risk in non-ε4 carriers. CONCLUSIONS Our study suggests that APOE ε4 remains the best predictor of broad AD risk when compared to multiple other genetic factors with modest effects, that phenotypic heterogeneity in broad AD can complicate simple polygenic risk modeling, and supports the association between HFE and AD risk in individuals without APOE ε4.
Collapse
Affiliation(s)
- Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94158, USA.
| | - Luke W Bonham
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94158, USA.
| | - Renee L Sears
- Semel Institute for Neuroscience and Human Behavior, Departments of Neurology and Psychiatry, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Eric Klein
- Semel Institute for Neuroscience and Human Behavior, Departments of Neurology and Psychiatry, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Anna Karydas
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94158, USA.
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94158, USA.
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94158, USA.
| | - Giovanni Coppola
- Semel Institute for Neuroscience and Human Behavior, Departments of Neurology and Psychiatry, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
3064
|
Yan J, Kim S, Nho K, Chen R, Risacher SL, Moore JH, Saykin AJ, Shen L. Hippocampal transcriptome-guided genetic analysis of correlated episodic memory phenotypes in Alzheimer's disease. Front Genet 2015; 6:117. [PMID: 25859259 PMCID: PMC4374536 DOI: 10.3389/fgene.2015.00117] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 03/09/2015] [Indexed: 01/18/2023] Open
Abstract
As the most common type of dementia, Alzheimer's disease (AD) is a neurodegenerative disorder initially manifested by impaired memory performances. While the diagnosis information indicates a dichotomous status of a patient, memory scores have the potential to capture the continuous nature of the disease progression and may provide more insights into the underlying mechanism. In this work, we performed a targeted genetic study of memory scores on an AD cohort to identify the associations between a set of genes highly expressed in the hippocampal region and seven cognitive scores related to episodic memory. Both main effects and interaction effects of the targeted genetic markers on these correlated memory scores were examined. In addition to well-known AD genetic markers APOE and TOMM40, our analysis identified a new risk gene NAV2 through the gene-level main effect analysis. NAV2 was found to be significantly and consistently associated with all seven episodic memory scores. Genetic interaction analysis also yielded a few promising hits warranting further investigation, especially for the RAVLT list B Score.
Collapse
Affiliation(s)
- Jingwen Yan
- BioHealth, Indiana University School of Informatics and Computing Indianapolis, IN, USA ; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA
| | - Sungeun Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine Indianapolis, IN, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine Indianapolis, IN, USA
| | - Rui Chen
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Computer Science, Dartmouth College Hanover, NH, USA
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine Indianapolis, IN, USA
| | - Jason H Moore
- Genetics, Community and Family Medicine, Geisel School of Medicine at Dartmouth Lebanon, NH, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine Indianapolis, IN, USA ; Medical and Molecular Genetics, Indiana University School of Medicine Indianapolis, IN, USA
| | - Li Shen
- BioHealth, Indiana University School of Informatics and Computing Indianapolis, IN, USA ; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine Indianapolis, IN, USA ; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine Indianapolis, IN, USA
| | | |
Collapse
|
3065
|
Steinberg S, Stefansson H, Jonsson T, Johannsdottir H, Ingason A, Helgason H, Sulem P, Magnusson OT, Gudjonsson SA, Unnsteinsdottir U, Kong A, Helisalmi S, Soininen H, Lah JJ, Aarsland D, Fladby T, Ulstein ID, Djurovic S, Sando SB, White LR, Knudsen GP, Westlye LT, Selbæk G, Giegling I, Hampel H, Hiltunen M, Levey AI, Andreassen OA, Rujescu D, Jonsson PV, Bjornsson S, Snaedal J, Stefansson K. Loss-of-function variants in ABCA7 confer risk of Alzheimer's disease. Nat Genet 2015; 47:445-7. [PMID: 25807283 DOI: 10.1038/ng.3246] [Citation(s) in RCA: 262] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 02/13/2015] [Indexed: 12/12/2022]
Abstract
We conducted a search for rare, functional variants altering susceptibility to Alzheimer's disease that exploited knowledge of common variants associated with the same disease. We found that loss-of-function variants in ABCA7 confer risk of Alzheimer's disease in Icelanders (odds ratio (OR) = 2.12, P = 2.2 × 10(-13)) and discovered that the association replicated in study groups from Europe and the United States (combined OR = 2.03, P = 6.8 × 10(-15)).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Seppo Helisalmi
- 1] Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland. [2] NeuroCenter, Kuopio University Hospital, Kuopio, Finland
| | - Hilkka Soininen
- 1] Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland. [2] NeuroCenter, Kuopio University Hospital, Kuopio, Finland
| | - James J Lah
- School of Medicine, Emory University, Atlanta, Georgia, USA
| | | | - Dag Aarsland
- 1] Alzheimer's Disease Research Centre, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden. [2] Center for Age-Related Diseases, Stavanger University Hospital, Stavanger, Norway. [3] Institute of Clinical Medicine, Division of Medicine and Laboratory Sciences, University of Oslo, Oslo, Norway
| | - Tormod Fladby
- 1] Institute of Clinical Medicine, Division of Medicine and Laboratory Sciences, University of Oslo, Oslo, Norway. [2] Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Ingun D Ulstein
- 1] Department of Psychiatry of Old Age, Oslo University Hospital, Oslo, Norway. [2] Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- 1] Department of Medical Genetics, Oslo University Hospital, Oslo, Norway. [2] NORMENT-K.G. Jebsen Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Sigrid B Sando
- 1] Department of Neuroscience, Norwegian University of Science and Technology, Trondheim, Norway. [2] Department of Neurology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Linda R White
- 1] Department of Neuroscience, Norwegian University of Science and Technology, Trondheim, Norway. [2] Department of Neurology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Gun-Peggy Knudsen
- Division of Mental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Lars T Westlye
- 1] NORMENT-K.G. Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway. [2] Department of Psychology, University of Oslo, Oslo, Norway
| | - Geir Selbæk
- Ageing and Health, Norwegian Centre for Research, Education and Service Development, Vestfold Hospital Trust, Tønsberg, Norway
| | - Ina Giegling
- Department of Psychiatry, University of Halle, Halle, Germany
| | - Harald Hampel
- Sorbonne Universités, Université Pierre et Marie Curie, Département de Neurologie, Hôpital Pitié Salpêtrière, Paris, France
| | - Mikko Hiltunen
- 1] Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland. [2] NeuroCenter, Kuopio University Hospital, Kuopio, Finland. [3] Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Allan I Levey
- School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Ole A Andreassen
- 1] NORMENT-K.G. Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway. [2] NORMENT-K.G. Jebsen Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dan Rujescu
- Department of Psychiatry, University of Halle, Halle, Germany
| | - Palmi V Jonsson
- 1] Faculty of Medicine, University of Iceland, Reykjavik, Iceland. [2] Department of Geriatric Medicine, Landspitali University Hospital, Reykjavik, Iceland
| | - Sigurbjorn Bjornsson
- Department of Geriatric Medicine, Landspitali University Hospital, Reykjavik, Iceland
| | - Jon Snaedal
- Department of Geriatric Medicine, Landspitali University Hospital, Reykjavik, Iceland
| | - Kari Stefansson
- 1] deCODE Genetics, Reykjavik, Iceland. [2] Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
3066
|
Shulman JM. Drosophila and experimental neurology in the post-genomic era. Exp Neurol 2015; 274:4-13. [PMID: 25814441 DOI: 10.1016/j.expneurol.2015.03.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/03/2015] [Accepted: 03/18/2015] [Indexed: 12/31/2022]
Abstract
For decades, the fruit fly, Drosophila melanogaster, has been among the premiere genetic model systems for probing fundamental neurobiology, including elucidation of mechanisms responsible for human neurologic disorders. Flies continue to offer virtually unparalleled versatility and speed for genetic manipulation, strong genomic conservation, and a nervous system that recapitulates a range of cellular and network properties relevant to human disease. I focus here on four critical challenges emerging from recent advances in our understanding of the genomic basis of human neurologic disorders where innovative experimental strategies are urgently needed: (1) pinpointing causal genes from associated genomic loci; (2) confirming the functional impact of allelic variants; (3) elucidating nervous system roles for novel or poorly studied genes; and (4) probing network interactions within implicated regulatory pathways. Drosophila genetic approaches are ideally suited to address each of these potential translational roadblocks, and will therefore contribute to mechanistic insights and potential breakthrough therapies for complex genetic disorders in the coming years. Strategic collaboration between neurologists, human geneticists, and the Drosophila research community holds great promise to accelerate progress in the post-genomic era.
Collapse
Affiliation(s)
- Joshua M Shulman
- Departments of Neurology, Molecular and Human Genetics, and Neuroscience, and Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
3067
|
Liao F, Jiang H, Srivatsan S, Xiao Q, Lefton KB, Yamada K, Mahan TE, Lee JM, Shaw AS, Holtzman DM. Effects of CD2-associated protein deficiency on amyloid-β in neuroblastoma cells and in an APP transgenic mouse model. Mol Neurodegener 2015; 10:12. [PMID: 25887956 PMCID: PMC4374406 DOI: 10.1186/s13024-015-0006-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/26/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND CD2-associated protein (CD2AP) is an SH3-containing scaffold adaptor protein which regulates the actin cytoskeleton. Recently, CD2AP was identified as a genetic risk factor for Alzheimer's disease (AD) by several genome-wide association studies. One of the hallmarks of AD is the accumulation of aggregated forms of Amyloid-β (Aβ) in the brain. In humans, CD2AP AD susceptibility locus (rs9349407) is associated with an increased plaque burden. Aβ production is highly regulated by endocytosis and is influenced by lysosomal function. Lysosomal trafficking is influenced by CD2AP. In this study, we decreased CD2AP levels in N2a neuroblastoma cultures and PS1APP mice and analyzed Aβ levels and plaque burden. RESULTS Our data show that suppressing CD2AP expression using shRNA in N2a-APP695 cells results in decreased cell membrane amyloid precursor protein, decreased Aβ release and a lower Aβ42/Aβ40 ratio. CD2AP protein is expressed in the brain as detected by western blot, and the expression level is dependent on gene dosage. In 1-month old PS1APP mice, complete loss of CD2AP in brain resulted in a decreased Aβ42/Aβ40 ratio in brain tissue lysates while there was no effect on Aβ deposition or accumulation in PS1APP mice expressing one copy of CD2AP. CONCLUSION CD2-Associated Protein affects Aβ levels and Aβ42/Aβ40 ratio in vitro. The effect of CD2-Associated Protein on Aβ metabolism is subtle in vivo.
Collapse
Affiliation(s)
- Fan Liao
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| | - Hong Jiang
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| | - Subhashini Srivatsan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Qingli Xiao
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| | - Katheryn B Lefton
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Kaoru Yamada
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Thomas E Mahan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Jin-Moo Lee
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| | - Andrey S Shaw
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
3068
|
Yu L, Chibnik LB, Srivastava GP, Pochet N, Yang J, Xu J, Kozubek J, Obholzer N, Leurgans SE, Schneider JA, Meissner A, De Jager PL, Bennett DA. Association of Brain DNA methylation in SORL1, ABCA7, HLA-DRB5, SLC24A4, and BIN1 with pathological diagnosis of Alzheimer disease. JAMA Neurol 2015; 72:15-24. [PMID: 25365775 DOI: 10.1001/jamaneurol.2014.3049] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE Recent large-scale genome-wide association studies have discovered several genetic variants associated with Alzheimer disease (AD); however, the extent to which DNA methylation in these AD loci contributes to the disease susceptibility remains unknown. OBJECTIVE To examine the association of brain DNA methylation in 28 reported AD loci with AD pathologies. DESIGN, SETTING, AND PARTICIPANTS Ongoing community-based clinical pathological cohort studies of aging and dementia (the Religious Orders Study and the Rush Memory and Aging Project) among 740 autopsied participants 66.0 to 108.3 years old. EXPOSURES DNA methylation levels at individual CpG sites generated from dorsolateral prefrontal cortex tissue using a bead assay. MAIN OUTCOMES AND MEASURES Pathological diagnosis of AD by National Institute on Aging-Reagan criteria following a standard postmortem examination. RESULTS Overall, 447 participants (60.4%) met the criteria for pathological diagnosis of AD. Brain DNA methylation in SORL1, ABCA7, HLA-DRB5, SLC24A4, and BIN1 was associated with pathological AD. The association was robustly retained after replacing the binary trait of pathological AD with 2 quantitative and molecular specific hallmarks of AD, namely, Aβ load and paired helical filament tau tangle density. Furthermore, RNA expression of transcripts of SORL1 and ABCA7 was associated with paired helical filament tau tangle density, and the expression of BIN1 was associated with Aβ load. CONCLUSIONS AND RELEVANCE Brain DNA methylation in multiple AD loci is associated with AD pathologies. The results provide further evidence that disruption of DNA methylation is involved in the pathological process of AD.
Collapse
Affiliation(s)
- Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois2Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Lori B Chibnik
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts4Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departme
| | - Gyan P Srivastava
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts4Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departme
| | - Nathalie Pochet
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts4Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departme
| | - Jingyun Yang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois2Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Jishu Xu
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts4Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departme
| | - James Kozubek
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts4Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departme
| | - Nikolaus Obholzer
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts4Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departme
| | - Sue E Leurgans
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois2Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois7Department of Preventive Medicine, Rush University Medical Center, Chicago, Illinois
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois2Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois8Department of Pathology, Rush University Medical Center, Chicago, Illinois
| | - Alexander Meissner
- Epigenomics Program, Broad Institute, Cambridge Center, Cambridge, Massachusetts10Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| | - Philip L De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts4Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departme
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois2Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
3069
|
Affiliation(s)
- Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland2Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Alan E Renton
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland
| |
Collapse
|
3070
|
Lacour A, Schüller V, Drichel D, Herold C, Jessen F, Leber M, Maier W, Noethen MM, Ramirez A, Vaitsiakhovich T, Becker T. Novel genetic matching methods for handling population stratification in genome-wide association studies. BMC Bioinformatics 2015; 16:84. [PMID: 25880419 PMCID: PMC4367953 DOI: 10.1186/s12859-015-0521-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 02/27/2015] [Indexed: 11/21/2022] Open
Abstract
Background A usually confronted problem in association studies is the occurrence of population stratification. In this work, we propose a novel framework to consider population matchings in the contexts of genome-wide and sequencing association studies. We employ pairwise and groupwise optimal case-control matchings and present an agglomerative hierarchical clustering, both based on a genetic similarity score matrix. In order to ensure that the resulting matches obtained from the matching algorithm capture correctly the population structure, we propose and discuss two stratum validation methods. We also invent a decisive extension to the Cochran-Armitage Trend test to explicitly take into account the particular population structure. Results We assess our framework by simulations of genotype data under the null hypothesis, to affirm that it correctly controls for the type-1 error rate. By a power study we evaluate that structured association testing using our framework displays reasonable power. We compare our result with those obtained from a logistic regression model with principal component covariates. Using the principal components approaches we also find a possible false-positive association to Alzheimer’s disease, which is neither supported by our new methods, nor by the results of a most recent large meta analysis or by a mixed model approach. Conclusions Matching methods provide an alternative handling of confounding due to population stratification for statistical tests for which covariates are hard to model. As a benchmark, we show that our matching framework performs equally well to state of the art models on common variants. Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0521-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- André Lacour
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 25, Bonn, 53127, Germany.
| | - Vitalia Schüller
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 25, Bonn, 53127, Germany.
| | - Dmitriy Drichel
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 25, Bonn, 53127, Germany.
| | - Christine Herold
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 25, Bonn, 53127, Germany.
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 25, Bonn, 53127, Germany. .,Abteilung für Psychiatrie und Psychotherapie, Universitätsklinikum Bonn, Sigmund-Freud-Str. 25, Bonn, 53127, Germany.
| | - Markus Leber
- Institut für Medizinische Biometrie, Informatik und Epidemiologie, Universität Bonn, Sigmund-Freud-Str. 25, Bonn, 53127, Germany.
| | - Wolfgang Maier
- Abteilung für Psychiatrie und Psychotherapie, Universitätsklinikum Bonn, Sigmund-Freud-Str. 25, Bonn, 53127, Germany.
| | - Markus M Noethen
- Institut für Humangenetik and Life & Brain Center, Universität Bonn, Sigmund-Freud-Str. 25, Bonn, 53127, Germany.
| | - Alfredo Ramirez
- Abteilung für Psychiatrie und Psychotherapie, Universitätsklinikum Bonn, Sigmund-Freud-Str. 25, Bonn, 53127, Germany.
| | - Tatsiana Vaitsiakhovich
- Institut für Medizinische Biometrie, Informatik und Epidemiologie, Universität Bonn, Sigmund-Freud-Str. 25, Bonn, 53127, Germany.
| | - Tim Becker
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 25, Bonn, 53127, Germany. .,Institut für Medizinische Biometrie, Informatik und Epidemiologie, Universität Bonn, Sigmund-Freud-Str. 25, Bonn, 53127, Germany.
| |
Collapse
|
3071
|
Young JE, Boulanger-Weill J, Williams DA, Woodruff G, Buen F, Revilla AC, Herrera C, Israel MA, Yuan SH, Edland SD, Goldstein LSB. Elucidating molecular phenotypes caused by the SORL1 Alzheimer's disease genetic risk factor using human induced pluripotent stem cells. Cell Stem Cell 2015; 16:373-85. [PMID: 25772071 DOI: 10.1016/j.stem.2015.02.004] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 01/11/2015] [Accepted: 02/10/2015] [Indexed: 12/20/2022]
Abstract
Predisposition to sporadic Alzheimer's disease (SAD) involves interactions between a person's unique combination of genetic variants and the environment. The molecular effect of these variants may be subtle and difficult to analyze with standard in vitro or in vivo models. Here we used hIPSCs to examine genetic variation in the SORL1 gene and possible contributions to SAD-related phenotypes in human neurons. We found that human neurons carrying SORL1 variants associated with an increased SAD risk show a reduced response to treatment with BDNF, at the level of both SORL1 expression and APP processing. shRNA knockdown of SORL1 demonstrates that the differences in BDNF-induced APP processing between genotypes are dependent on SORL1 expression. We propose that the variation in SORL1 expression induction by BDNF is modulated by common genetic variants and can explain how genetic variation in this one locus can contribute to an individual's risk of developing SAD.
Collapse
Affiliation(s)
- Jessica E Young
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jonathan Boulanger-Weill
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel A Williams
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Grace Woodruff
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Floyd Buen
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Arra C Revilla
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Cheryl Herrera
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mason A Israel
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shauna H Yuan
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Steven D Edland
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA; Division of Biostatistics, Department of Family and Preventive Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lawrence S B Goldstein
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
3072
|
Malik M, Chiles J, Xi HS, Medway C, Simpson J, Potluri S, Howard D, Liang Y, Paumi CM, Mukherjee S, Crane P, Younkin S, Fardo DW, Estus S. Genetics of CD33 in Alzheimer's disease and acute myeloid leukemia. Hum Mol Genet 2015; 24:3557-70. [PMID: 25762156 DOI: 10.1093/hmg/ddv092] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 03/09/2015] [Indexed: 01/01/2023] Open
Abstract
The CD33 single-nucleotide polymorphism (SNP) rs3865444 has been associated with the risk of Alzheimer's disease (AD). Rs3865444 is in linkage disequilibrium with rs12459419 which has been associated with efficacy of an acute myeloid leukemia (AML) chemotherapeutic agent based on a CD33 antibody. We seek to evaluate the extent to which CD33 genetics in AD and AML can inform one another and advance human disease therapy. We have previously shown that these SNPs are associated with skipping of CD33 exon 2 in brain mRNA. Here, we report that these CD33 SNPs are associated with exon 2 skipping in leukocytes from AML patients and with a novel CD33 splice variant that retains CD33 intron 1. Each copy of the minor rs12459419T allele decreases prototypic full-length CD33 expression by ∼ 25% and decreases the AD odds ratio by ∼ 0.10. These results suggest that CD33 antagonists may be useful in reducing AD risk. CD33 inhibitors may include humanized CD33 antibodies such as lintuzumab which was safe but ineffective in AML clinical trials. Here, we report that lintuzumab downregulates cell-surface CD33 by 80% in phorbol-ester differentiated U937 cells, at concentrations as low as 10 ng/ml. Overall, we propose a model wherein a modest effect on RNA splicing is sufficient to mediate the CD33 association with AD risk and suggest the potential for an anti-CD33 antibody as an AD-relevant pharmacologic agent.
Collapse
Affiliation(s)
- Manasi Malik
- Department of Physiology, Sanders-Brown Center on Aging
| | - Joe Chiles
- Department of Physiology, Sanders-Brown Center on Aging
| | - Hualin S Xi
- Computational Sciences Center of Emphasis, Pfizer Inc., Cambridge, MA 02140, USA
| | - Christopher Medway
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL 32224, USA and
| | - James Simpson
- Department of Physiology, Sanders-Brown Center on Aging
| | | | | | | | | | | | - Paul Crane
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Steven Younkin
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL 32224, USA and
| | - David W Fardo
- Department of Biostatistics, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Steven Estus
- Department of Physiology, Sanders-Brown Center on Aging ,
| |
Collapse
|
3073
|
Abstract
The copper metabolism disorder Wilson's disease was first defined in 1912. Wilson's disease can present with hepatic and neurological deficits, including dystonia and parkinsonism. Early-onset presentations in infancy and late-onset manifestations in adults older than 70 years of age are now well recognised. Direct genetic testing for ATP7B mutations are increasingly available to confirm the clinical diagnosis of Wilson's disease, and results from biochemical and genetic prevalence studies suggest that Wilson's disease might be much more common than previously estimated. Early diagnosis of Wilson's disease is crucial to ensure that patients can be started on adequate treatment, but uncertainty remains about the best possible choice of medication. Furthermore, Wilson's disease needs to be differentiated from other conditions that also present clinically with hepatolenticular degeneration or share biochemical abnormalities with Wilson's disease, such as reduced serum ceruloplasmin concentrations. Disordered copper metabolism is also associated with other neurological conditions, including a subtype of axonal neuropathy due to ATP7A mutations and the late-onset neurodegenerative disorders Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Oliver Bandmann
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK.
| | - Karl Heinz Weiss
- University Hospital Heidelberg, Department of Internal Medicine IV, Heidelberg, Germany
| | - Stephen G Kaler
- Section on Translational Neuroscience, Molecular Medicine Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
3074
|
Bohm C, Chen F, Sevalle J, Qamar S, Dodd R, Li Y, Schmitt-Ulms G, Fraser PE, St George-Hyslop PH. Current and future implications of basic and translational research on amyloid-β peptide production and removal pathways. Mol Cell Neurosci 2015; 66:3-11. [PMID: 25748120 PMCID: PMC4503820 DOI: 10.1016/j.mcn.2015.02.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 01/12/2023] Open
Abstract
Inherited variants in multiple different genes are associated with increased risk for Alzheimer's disease (AD). In many of these genes, the inherited variants alter some aspect of the production or clearance of the neurotoxic amyloid β-peptide (Aβ). Thus missense, splice site or duplication mutants in the presenilin 1 (PS1), presenilin 2 (PS2) or the amyloid precursor protein (APP) genes, which alter the levels or shift the balance of Aβ produced, are associated with rare, highly penetrant autosomal dominant forms of Familial Alzheimer's Disease (FAD). Similarly, the more prevalent late-onset forms of AD are associated with both coding and non-coding variants in genes such as SORL1, PICALM and ABCA7 that affect the production and clearance of Aβ. This review summarises some of the recent molecular and structural work on the role of these genes and the proteins coded by them in the biology of Aβ. We also briefly outline how the emerging knowledge about the pathways involved in Aβ generation and clearance can be potentially targeted therapeutically. This article is part of Special Issue entitled "Neuronal Protein".
Collapse
Affiliation(s)
- C Bohm
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - F Chen
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - J Sevalle
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - S Qamar
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - R Dodd
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - Y Li
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - G Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - P E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - P H St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada; Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK.
| |
Collapse
|
3075
|
Gjoneska E, Pfenning AR, Mathys H, Quon G, Kundaje A, Tsai LH, Kellis M. Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer's disease. Nature 2015; 518:365-9. [PMID: 25693568 PMCID: PMC4530583 DOI: 10.1038/nature14252] [Citation(s) in RCA: 390] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 01/22/2015] [Indexed: 12/12/2022]
Abstract
Alzheimer’s disease (AD) is a severe1 age-related neurodegenerative disorder characterized by accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles, synaptic and neuronal loss, and cognitive decline. Several genes have been implicated in AD, but chromatin state alterations during neurodegeneration remain uncharacterized. Here, we profile transcriptional and chromatin state dynamics across early and late pathology in the hippocampus of an inducible mouse model of AD-like neurodegeneration. We find a coordinated downregulation of synaptic plasticity genes and regulatory regions, and upregulation of immune response genes and regulatory regions, which are targeted by factors that belong to the ETS family of transcriptional regulators, including PU.1. Human regions orthologous to increasing-level enhancers show immune cell-specific enhancer signatures as well as immune cell expression quantitative trait loci (eQTL), while decreasing-level enhancer orthologs show fetal-brain-specific enhancer activity. Notably, AD-associated genetic variants are specifically enriched in increasing-level enhancer orthologs implicating immune processes in AD predisposition. Indeed, increasing enhancers overlap known AD loci lacking protein-altering variants and implicate additional loci that do not reach genome-wide significance. Our results reveal new insights into the mechanisms of neurodegeneration and establish the mouse as a useful model for functional studies of AD regulatory regions.
Collapse
Affiliation(s)
- Elizabeta Gjoneska
- 1] The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA [2] Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Andreas R Pfenning
- 1] Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA [2] Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Hansruedi Mathys
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Gerald Quon
- 1] Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA [2] Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Anshul Kundaje
- 1] Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA [2] Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA [3] Department of Genetics, Department of Computer Science, Stanford University, Stanford, California 94305, USA
| | - Li-Huei Tsai
- 1] The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA [2] Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Manolis Kellis
- 1] Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA [2] Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
3076
|
Reynolds CA, Finkel D. A meta-analysis of heritability of cognitive aging: minding the "missing heritability" gap. Neuropsychol Rev 2015; 25:97-112. [PMID: 25732892 DOI: 10.1007/s11065-015-9280-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 01/26/2015] [Indexed: 12/19/2022]
Abstract
The etiologies underlying variation in adult cognitive performance and cognitive aging have enjoyed much attention in the literature, but much of that attention has focused on broad factors, principally general cognitive ability. The current review provides meta-analyses of age trends in heritability of specific cognitive abilities and considers the profile of genetic and environmental factors contributing to cognitive aging to address the 'missing heritability' issue. Our findings, based upon evaluating 27 reports in the literature, indicate that verbal ability demonstrated declining heritability, after about age 60, as did spatial ability and perceptual speed more modestly. Trends for general memory, working memory, and spatial ability generally indicated stability, or small increases in heritability in mid-life. Equivocal results were found for executive function. A second meta-analysis then considered the gap between twin-based versus SNP-based heritability derived from population-based GWAS studies. Specifically, we considered twin correlation ratios to agnostically re-evaluate biometrical models across age and by cognitive domain. Results modestly suggest that nonadditive genetic variance may become increasingly important with age, especially for verbal ability. If so, this would support arguments that lower SNP-based heritability estimates result in part from uncaptured non-additive influences (e.g., dominance, gene-gene interactions), and possibly gene-environment (GE) correlations. Moreover, consistent with longitudinal twin studies of aging, as rearing environment becomes a distal factor, increasing genetic variance may result in part from nonadditive genetic influences or possible GE correlations. Sensitivity to life course dynamics is crucial to understanding etiological contributions to adult cognitive performance and cognitive aging.
Collapse
Affiliation(s)
- Chandra A Reynolds
- Department of Psychology, University of California Riverside, Riverside, CA, 92521, USA,
| | | |
Collapse
|
3077
|
Alzheimer’s disease shares gene expression aberrations with purinergic dysregulation of HPRT deficiency (Lesch–Nyhan disease). Neurosci Lett 2015; 590:35-9. [DOI: 10.1016/j.neulet.2015.01.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 12/19/2022]
|
3078
|
Abstract
Alzheimer's disease (AD), the most common form of dementia in western societies, is a pathologically and clinically heterogeneous disease with a strong genetic component. The recent advances in high-throughput genome technologies allowing for the rapid analysis of millions of polymorphisms in thousands of subjects has significantly advanced our understanding of the genomic underpinnings of AD susceptibility. During the last 5 years, genome-wide association and whole-exome- and whole-genome sequencing studies have mapped more than 20 disease-associated loci, providing insights into the molecular pathways involved in AD pathogenesis and hinting at potential novel therapeutic targets. This review article summarizes the challenges and opportunities of when using genomic information for the diagnosis and prognosis of AD.
Collapse
Affiliation(s)
- Christiane Reitz
- Sergievsly Center/Taub Institute/Dept. of Neurology, Columbia University, 630 W 168th Street, Rm 19-308, New York, NY 10032, phone: (212) 305-0865, fax: (212) 305-2391
| |
Collapse
|
3079
|
Camargo LM, Zhang XD, Loerch P, Caceres RM, Marine SD, Uva P, Ferrer M, de Rinaldis E, Stone DJ, Majercak J, Ray WJ, Yi-An C, Shearman MS, Mizuguchi K. Pathway-based analysis of genome-wide siRNA screens reveals the regulatory landscape of APP processing. PLoS One 2015; 10:e0115369. [PMID: 25723573 PMCID: PMC4344212 DOI: 10.1371/journal.pone.0115369] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 11/14/2014] [Indexed: 01/08/2023] Open
Abstract
The progressive aggregation of Amyloid-β (Aβ) in the brain is a major trait of Alzheimer's Disease (AD). Aβ is produced as a result of proteolytic processing of the β-amyloid precursor protein (APP). Processing of APP is mediated by multiple enzymes, resulting in the production of distinct peptide products: the non-amyloidogenic peptide sAPPα and the amyloidogenic peptides sAPPβ, Aβ40, and Aβ42. Using a pathway-based approach, we analyzed a large-scale siRNA screen that measured the production of different APP proteolytic products. Our analysis identified many of the biological processes/pathways that are known to regulate APP processing and have been implicated in AD pathogenesis, as well as revealing novel regulatory mechanisms. Furthermore, we also demonstrate that some of these processes differentially regulate APP processing, with some mechanisms favouring production of certain peptide species over others. For example, synaptic transmission having a bias towards regulating Aβ40 production over Aβ42 as well as processes involved in insulin and pancreatic biology having a bias for sAPPβ production over sAPPα. In addition, some of the pathways identified as regulators of APP processing contain genes (CLU, BIN1, CR1, PICALM, TREM2, SORL1, MEF2C, DSG2, EPH1A) recently implicated with AD through genome wide association studies (GWAS) and associated meta-analysis. In addition, we provide supporting evidence and a deeper mechanistic understanding of the role of diabetes in AD. The identification of these processes/pathways, their differential impact on APP processing, and their relationships to each other, provide a comprehensive systems biology view of the “regulatory landscape” of APP.
Collapse
Affiliation(s)
- Luiz Miguel Camargo
- Merck Research Laboratories, Merck & Co, Boston, United States of America
- * E-mail:
| | | | - Patrick Loerch
- Merck Research Laboratories, Merck & Co, Boston, United States of America
| | | | - Shane D. Marine
- Merck Research Laboratories, Merck & Co, North Wales, United States of America
| | - Paolo Uva
- Merck Research Laboratories, Instituto di Recerca di Biologia Molecolare, Pomezia, Italy
| | - Marc Ferrer
- Merck Research Laboratories, Merck & Co, North Wales, United States of America
| | - Emanuele de Rinaldis
- Merck Research Laboratories, Instituto di Recerca di Biologia Molecolare, Pomezia, Italy
| | - David J. Stone
- Merck Research Laboratories, Merck & Co, West Point, United States of America
| | - John Majercak
- Merck Research Laboratories, Merck & Co, West Point, United States of America
| | - William J. Ray
- Merck Research Laboratories, Merck & Co, West Point, United States of America
| | - Chen Yi-An
- National Institute of Biomedical Innovation, Osaka, Japan
| | - Mark S. Shearman
- Merck Research Laboratories, Merck & Co, Boston, United States of America
| | | |
Collapse
|
3080
|
Shuai P, Liu Y, Lu W, Liu Q, Li T, Gong B. Genetic associations of CLU rs9331888 polymorphism with Alzheimer's disease: A meta-analysis. Neurosci Lett 2015; 591:160-165. [PMID: 25703218 DOI: 10.1016/j.neulet.2015.02.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 02/06/2015] [Accepted: 02/14/2015] [Indexed: 11/29/2022]
Abstract
Recent studies showed the Clusterin gene (CLU) is associated with Alzheimer's disease (AD). However, studies investigating the association of CLU single-nucleotide polymorphism (SNP) rs9331888 with AD are controversial. We then performed a meta-analysis to assess the association between CLU SNP rs9331888 and AD. Computerized bibliographic searches of PUBMED and AlzGene database were conducted for the period up to July, 2014. The strength of the association between SNP rs9331888 and AD was estimated by odds ratios (ORs) and OR 95% confidence intervals (CIs). A total of 11 studies composed of 8766 AD cases and 11,366 controls were included in this study. Significant association of SNP rs9331888 with AD was found in Caucasian population among allelic model (C vs. G: OR=1.12, 95%CI=1.06-1.17, P<0.001), additive model (CC vs. GG: OR=1.25, 95%CI=1.12-1.40, P<0.001), recessive model (CC vs. CG+GG: OR=1.20, 95%CI=1.07-1.34, P=0.001), and dominant model (CC+CG vs. GG: OR=1.13, 95%CI=1.06-1.21, P<0.001). No significant association among these models was found in Asian and overall populations. Sensitivity analysis found that one study caused the distinct heterogeneity in Asian subgroup. Our analysis demonstrated that CLU SNP rs9331888 might be associated with an increased AD risk in Caucasian population, but not in Asian population.
Collapse
Affiliation(s)
- Ping Shuai
- Health Management Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; Sichuan Provincial Key Laboratory for Disease Gene Study, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yuping Liu
- Health Management Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Wenxue Lu
- West China School of Public Health, Sichuan University, Chengdu, China
| | - Qiaolan Liu
- West China School of Public Health, Sichuan University, Chengdu, China
| | - Tinxin Li
- Health Management Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Bo Gong
- Sichuan Provincial Key Laboratory for Disease Gene Study, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
3081
|
Hossini AM, Megges M, Prigione A, Lichtner B, Toliat MR, Wruck W, Schröter F, Nuernberg P, Kroll H, Makrantonaki E, Zouboulis CC, Zoubouliss CC, Adjaye J. Induced pluripotent stem cell-derived neuronal cells from a sporadic Alzheimer's disease donor as a model for investigating AD-associated gene regulatory networks. BMC Genomics 2015; 16:84. [PMID: 25765079 PMCID: PMC4344782 DOI: 10.1186/s12864-015-1262-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 01/22/2015] [Indexed: 02/07/2023] Open
Abstract
Background Alzheimer’s disease (AD) is a complex, irreversible neurodegenerative disorder. At present there are neither reliable markers to diagnose AD at an early stage nor therapy. To investigate underlying disease mechanisms, induced pluripotent stem cells (iPSCs) allow the generation of patient-derived neuronal cells in a dish. Results In this study, employing iPS technology, we derived and characterized iPSCs from dermal fibroblasts of an 82-year-old female patient affected by sporadic AD. The AD-iPSCs were differentiated into neuronal cells, in order to generate disease-specific protein association networks modeling the molecular pathology on the transcriptome level of AD, to analyse the reflection of the disease phenotype in gene expression in AD-iPS neuronal cells, in particular in the ubiquitin-proteasome system (UPS), and to address expression of typical AD proteins. We detected the expression of p-tau and GSK3B, a physiological kinase of tau, in neuronal cells derived from AD-iPSCs. Treatment of neuronal cells differentiated from AD-iPSCs with an inhibitor of γ-secretase resulted in the down-regulation of p-tau. Transcriptome analysis of AD-iPS derived neuronal cells revealed significant changes in the expression of genes associated with AD and with the constitutive as well as the inducible subunits of the proteasome complex. The neuronal cells expressed numerous genes associated with sub-regions within the brain thus suggesting the usefulness of our in-vitro model. Moreover, an AD-related protein interaction network composed of APP and GSK3B among others could be generated using neuronal cells differentiated from two AD-iPS cell lines. Conclusions Our study demonstrates how an iPSC-based model system could represent (i) a tool to study the underlying molecular basis of sporadic AD, (ii) a platform for drug screening and toxicology studies which might unveil novel therapeutic avenues for this debilitating neuronal disorder. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1262-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Amir M Hossini
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, 06847, Dessau, Germany.
| | - Matthias Megges
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany. .,Molecular Embryology and Aging Group, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195, Berlin, Germany. .,Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany.
| | - Alessandro Prigione
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany. .,Current address: Max Delbrueck Center for Molecular Medicine (MDC), Robert Roessle Str. 10, D-13125, Berlin, Germany.
| | - Bjoern Lichtner
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.
| | - Mohammad R Toliat
- Cologne Center for Genomics (CCG), Institute for Genetics, University of Cologne, 50931, Cologne, Germany.
| | - Wasco Wruck
- Molecular Embryology and Aging Group, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195, Berlin, Germany.
| | - Friederike Schröter
- Molecular Embryology and Aging Group, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195, Berlin, Germany.
| | - Peter Nuernberg
- Cologne Center for Genomics (CCG), Institute for Genetics, University of Cologne, 50931, Cologne, Germany.
| | - Hartmut Kroll
- Institute for Transfusion Medicine Dessau, Red Cross Blood Transfusion Service NSTOB, 06847, Dessau, Germany.
| | - Eugenia Makrantonaki
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, 06847, Dessau, Germany. .,Geriatrics Research Group, Department of Geriatric Medicine, Charité Universitätsmedizin Berlin, Reinickendorfer Str. 61, 13447, Berlin, Germany.
| | | | - Christos C Zoubouliss
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, 06847, Dessau, Germany.
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany. .,Molecular Embryology and Aging Group, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195, Berlin, Germany.
| |
Collapse
|
3082
|
Nho K, Kim S, Risacher SL, Shen L, Corneveaux JJ, Swaminathan S, Lin H, Ramanan VK, Liu Y, Foroud TM, Inlow MH, Siniard AL, Reiman RA, Aisen PS, Petersen RC, Green RC, Jack CR, Weiner MW, Baldwin CT, Lunetta KL, Farrer LA, Furney SJ, Lovestone S, Simmons A, Mecocci P, Vellas B, Tsolaki M, Kloszewska I, Soininen H, McDonald BC, Farlow MR, Ghetti B, Huentelman MJ, Saykin AJ. Protective variant for hippocampal atrophy identified by whole exome sequencing. Ann Neurol 2015; 77:547-52. [PMID: 25559091 DOI: 10.1002/ana.24349] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 12/16/2014] [Accepted: 12/24/2014] [Indexed: 01/25/2023]
Abstract
We used whole-exome sequencing to identify variants other than APOE associated with the rate of hippocampal atrophy in amnestic mild cognitive impairment. An in-silico predicted missense variant in REST (rs3796529) was found exclusively in subjects with slow hippocampal volume loss and validated using unbiased whole-brain analysis and meta-analysis across 5 independent cohorts. REST is a master regulator of neurogenesis and neuronal differentiation that has not been previously implicated in Alzheimer's disease. These findings nominate REST and its functional pathways as protective and illustrate the potential of combining next-generation sequencing with neuroimaging to discover novel disease mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3083
|
Morgen K, Frölich L. The metabolism hypothesis of Alzheimer’s disease: from the concept of central insulin resistance and associated consequences to insulin therapy. J Neural Transm (Vienna) 2015; 122:499-504. [DOI: 10.1007/s00702-015-1377-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/04/2015] [Indexed: 10/24/2022]
|
3084
|
Guerreiro R, Bras J, Toombs J, Heslegrave A, Hardy J, Zetterberg H. Genetic Variants and Related Biomarkers in Sporadic Alzheimer's Disease. CURRENT GENETIC MEDICINE REPORTS 2015; 3:19-25. [PMID: 25664224 PMCID: PMC4317514 DOI: 10.1007/s40142-014-0062-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
From a neuropathological perspective, elderly patients who die with a clinical diagnosis of sporadic Alzheimer's disease (AD) are a heterogeneous group with several different pathologies contributing to the AD phenotype. This poses a challenge when searching for low effect size susceptibility genes for AD. Further, control groups may be contaminated by significant numbers of preclinical AD patients, which also reduces the power of genetic association studies. Here, we discuss how cerebrospinal fluid and imaging biomarkers can be used to increase the chance of finding novel susceptibility genes and as a means to study the functional consequences of risk alleles.
Collapse
Affiliation(s)
- Rita Guerreiro
- Department of Molecular Neuroscience, UCL Institute of Neurology, 1 Wakefield Street (1st Floor), London, WC1N 1PJ UK
| | - Jose Bras
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
| | - Jamie Toombs
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
| | - Amanda Heslegrave
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, 1 Wakefield Street (1st Floor), London, WC1N 1PJ UK
| | - Henrik Zetterberg
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
| |
Collapse
|
3085
|
Retromer in Alzheimer disease, Parkinson disease and other neurological disorders. Nat Rev Neurosci 2015; 16:126-32. [PMID: 25669742 DOI: 10.1038/nrn3896] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Retromer is a protein assembly that has a central role in endosomal trafficking, and retromer dysfunction has been linked to a growing number of neurological disorders. First linked to Alzheimer disease, retromer dysfunction causes a range of pathophysiological consequences that have been shown to contribute to the core pathological features of the disease. Genetic studies have established that retromer dysfunction is also pathogenically linked to Parkinson disease, although the biological mechanisms that mediate this link are only now being elucidated. Most recently, studies have shown that retromer is a tractable target in drug discovery for these and other disorders of the nervous system.
Collapse
|
3086
|
Examination of candidate exonic variants for association to Alzheimer disease in the Amish. PLoS One 2015; 10:e0118043. [PMID: 25668194 PMCID: PMC4323242 DOI: 10.1371/journal.pone.0118043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 01/05/2015] [Indexed: 11/24/2022] Open
Abstract
Alzheimer disease (AD) is the most common cause of dementia. As with many complex diseases, the identified variants do not explain the total expected genetic risk that is based on heritability estimates for AD. Isolated founder populations, such as the Amish, are advantageous for genetic studies as they overcome heterogeneity limitations associated with complex population studies. We determined that Amish AD cases harbored a significantly higher burden of the known risk alleles compared to Amish cognitively normal controls, but a significantly lower burden when compared to cases from a dataset of unrelated individuals. Whole-exome sequencing of a selected subset of the overall study population was used as a screening tool to identify variants located in the regions of the genome that are most likely to contribute risk. By then genotyping the top candidate variants from the known AD genes and from linkage regions implicated previous studies in the full dataset, new associations could be confirmed. The most significant result (p = 0.0012) was for rs73938538, a synonymous variant in LAMA1 within the previously identified linkage peak on chromosome 18. However, this association is specific to the Amish and did not generalize when tested in a dataset of unrelated individuals. These results suggest that additional risk variation in the Amish remains to be identified and likely resides outside of the classical protein coding gene regions.
Collapse
|
3087
|
Apolipoprotein E4 produced in GABAergic interneurons causes learning and memory deficits in mice. J Neurosci 2015; 34:14069-78. [PMID: 25319703 DOI: 10.1523/jneurosci.2281-14.2014] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Apolipoprotein (apo) E4 is expressed in many types of brain cells, is associated with age-dependent decline of learning and memory in humans, and is the major genetic risk factor for AD. To determine whether the detrimental effects of apoE4 depend on its cellular sources, we generated human apoE knock-in mouse models in which the human APOE gene is conditionally deleted in astrocytes, neurons, or GABAergic interneurons. Here we report that deletion of apoE4 in astrocytes does not protect aged mice from apoE4-induced GABAergic interneuron loss and learning and memory deficits. In contrast, deletion of apoE4 in neurons does protect aged mice from both deficits. Furthermore, deletion of apoE4 in GABAergic interneurons is sufficient to gain similar protection. This study demonstrates a detrimental effect of endogenously produced apoE4 on GABAergic interneurons that leads to learning and memory deficits in mice and provides a novel target for drug development for AD related to apoE4.
Collapse
|
3088
|
Davies G, Armstrong N, Bis JC, Bressler J, Chouraki V, Giddaluru S, Hofer E, Ibrahim-Verbaas CA, Kirin M, Lahti J, van der Lee SJ, Le Hellard S, Liu T, Marioni RE, Oldmeadow C, Postmus I, Smith AV, Smith JA, Thalamuthu A, Thomson R, Vitart V, Wang J, Yu L, Zgaga L, Zhao W, Boxall R, Harris SE, Hill WD, Liewald DC, Luciano M, Adams H, Ames D, Amin N, Amouyel P, Assareh AA, Au R, Becker JT, Beiser A, Berr C, Bertram L, Boerwinkle E, Buckley BM, Campbell H, Corley J, De Jager PL, Dufouil C, Eriksson JG, Espeseth T, Faul JD, Ford I, Scotland G, Gottesman RF, Griswold ME, Gudnason V, Harris TB, Heiss G, Hofman A, Holliday EG, Huffman J, Kardia SLR, Kochan N, Knopman DS, Kwok JB, Lambert JC, Lee T, Li G, Li SC, Loitfelder M, Lopez OL, Lundervold AJ, Lundqvist A, Mather KA, Mirza SS, Nyberg L, Oostra BA, Palotie A, Papenberg G, Pattie A, Petrovic K, Polasek O, Psaty BM, Redmond P, Reppermund S, Rotter JI, Schmidt H, Schuur M, Schofield PW, Scott RJ, Steen VM, Stott DJ, van Swieten JC, Taylor KD, Trollor J, Trompet S, Uitterlinden AG, Weinstein G, Widen E, Windham BG, Jukema JW, Wright AF, et alDavies G, Armstrong N, Bis JC, Bressler J, Chouraki V, Giddaluru S, Hofer E, Ibrahim-Verbaas CA, Kirin M, Lahti J, van der Lee SJ, Le Hellard S, Liu T, Marioni RE, Oldmeadow C, Postmus I, Smith AV, Smith JA, Thalamuthu A, Thomson R, Vitart V, Wang J, Yu L, Zgaga L, Zhao W, Boxall R, Harris SE, Hill WD, Liewald DC, Luciano M, Adams H, Ames D, Amin N, Amouyel P, Assareh AA, Au R, Becker JT, Beiser A, Berr C, Bertram L, Boerwinkle E, Buckley BM, Campbell H, Corley J, De Jager PL, Dufouil C, Eriksson JG, Espeseth T, Faul JD, Ford I, Scotland G, Gottesman RF, Griswold ME, Gudnason V, Harris TB, Heiss G, Hofman A, Holliday EG, Huffman J, Kardia SLR, Kochan N, Knopman DS, Kwok JB, Lambert JC, Lee T, Li G, Li SC, Loitfelder M, Lopez OL, Lundervold AJ, Lundqvist A, Mather KA, Mirza SS, Nyberg L, Oostra BA, Palotie A, Papenberg G, Pattie A, Petrovic K, Polasek O, Psaty BM, Redmond P, Reppermund S, Rotter JI, Schmidt H, Schuur M, Schofield PW, Scott RJ, Steen VM, Stott DJ, van Swieten JC, Taylor KD, Trollor J, Trompet S, Uitterlinden AG, Weinstein G, Widen E, Windham BG, Jukema JW, Wright AF, Wright MJ, Yang Q, Amieva H, Attia JR, Bennett DA, Brodaty H, de Craen AJM, Hayward C, Ikram MA, Lindenberger U, Nilsson LG, Porteous DJ, Räikkönen K, Reinvang I, Rudan I, Sachdev PS, Schmidt R, Schofield PR, Srikanth V, Starr JM, Turner ST, Weir DR, Wilson JF, van Duijn C, Launer L, Fitzpatrick AL, Seshadri S, Mosley TH, Deary IJ. Genetic contributions to variation in general cognitive function: a meta-analysis of genome-wide association studies in the CHARGE consortium (N=53949). Mol Psychiatry 2015; 20:183-92. [PMID: 25644384 PMCID: PMC4356746 DOI: 10.1038/mp.2014.188] [Show More Authors] [Citation(s) in RCA: 276] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/11/2014] [Accepted: 11/24/2014] [Indexed: 01/14/2023]
Abstract
General cognitive function is substantially heritable across the human life course from adolescence to old age. We investigated the genetic contribution to variation in this important, health- and well-being-related trait in middle-aged and older adults. We conducted a meta-analysis of genome-wide association studies of 31 cohorts (N=53,949) in which the participants had undertaken multiple, diverse cognitive tests. A general cognitive function phenotype was tested for, and created in each cohort by principal component analysis. We report 13 genome-wide significant single-nucleotide polymorphism (SNP) associations in three genomic regions, 6q16.1, 14q12 and 19q13.32 (best SNP and closest gene, respectively: rs10457441, P=3.93 × 10(-9), MIR2113; rs17522122, P=2.55 × 10(-8), AKAP6; rs10119, P=5.67 × 10(-9), APOE/TOMM40). We report one gene-based significant association with the HMGN1 gene located on chromosome 21 (P=1 × 10(-6)). These genes have previously been associated with neuropsychiatric phenotypes. Meta-analysis results are consistent with a polygenic model of inheritance. To estimate SNP-based heritability, the genome-wide complex trait analysis procedure was applied to two large cohorts, the Atherosclerosis Risk in Communities Study (N=6617) and the Health and Retirement Study (N=5976). The proportion of phenotypic variation accounted for by all genotyped common SNPs was 29% (s.e.=5%) and 28% (s.e.=7%), respectively. Using polygenic prediction analysis, ~1.2% of the variance in general cognitive function was predicted in the Generation Scotland cohort (N=5487; P=1.5 × 10(-17)). In hypothesis-driven tests, there was significant association between general cognitive function and four genes previously associated with Alzheimer's disease: TOMM40, APOE, ABCG1 and MEF2C.
Collapse
Affiliation(s)
- G Davies
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - N Armstrong
- School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| | - J C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - J Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - V Chouraki
- Inserm-UMR744, Institut Pasteur de Lille, Unité d'Epidémiologie et de Santé Publique, Lille, France,Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - S Giddaluru
- K.G. Jebsen Centre for Psychosis Research and the Norwegian Centre for Mental Disorders Research (NORMENT), Department of Clinical Science, University of Bergen, Bergen, Norway,Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - E Hofer
- Department of Neurology, Medical University of Graz, Graz, Austria,Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - C A Ibrahim-Verbaas
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands,Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M Kirin
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, UK
| | - J Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland,Folkhälsan Research Centre, Helsinki, Finland
| | - S J van der Lee
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - S Le Hellard
- K.G. Jebsen Centre for Psychosis Research and the Norwegian Centre for Mental Disorders Research (NORMENT), Department of Clinical Science, University of Bergen, Bergen, Norway,Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - T Liu
- Max Planck Institute for Human Development, Berlin, Germany,Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - R E Marioni
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, UK,Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - C Oldmeadow
- Hunter Medical Research Institute and Faculty of Health, University of Newcastle, Newcastle, NSW, Australia
| | - I Postmus
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands,Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - A V Smith
- Icelandic Heart Association, Kopavogur, Iceland,University of Iceland, Reykjavik, Iceland
| | - J A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - A Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - R Thomson
- Menzies Research Institute, Hobart, Tasmania
| | - V Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - J Wang
- Framingham Heart Study, Framingham, MA, USA,Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - L Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - L Zgaga
- Department of Public Health and Primary Care, Trinity College Dublin, Dublin, Ireland,Andrija Stampar School of Public Health, Medical School, University of Zagreb, Zagreb, Croatia
| | - W Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - R Boxall
- Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - S E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - W D Hill
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - D C Liewald
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - M Luciano
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - H Adams
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands,Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - D Ames
- National Ageing Research Institute, Royal Melbourne Hospital, Melbourne, VIC, Australia,Academic Unit for Psychiatry of Old Age, St George's Hospital, University of Melbourne, Kew, Australia
| | - N Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands,Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - P Amouyel
- Inserm-UMR744, Institut Pasteur de Lille, Unité d'Epidémiologie et de Santé Publique, Lille, France
| | - A A Assareh
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - R Au
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA,Framingham Heart Study, Framingham, MA, USA
| | - J T Becker
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA,Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - A Beiser
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA,Framingham Heart Study, Framingham, MA, USA
| | - C Berr
- Inserm, U106, Montpellier, France,Université Montpellier I, Montpellier, France
| | - L Bertram
- Max Planck Institute for Molecular Genetics, Berlin, Germany,Faculty of Medicine, School of Public Health, Imperial College, London, UK
| | - E Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA,Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - B M Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - H Campbell
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, UK
| | - J Corley
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - P L De Jager
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA,Harvard Medical School, Boston, MA, USA,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - C Dufouil
- Inserm U708, Neuroepidemiology, Paris, France,Inserm U897, Université Bordeaux Segalen, Bordeaux, France
| | - J G Eriksson
- Folkhälsan Research Centre, Helsinki, Finland,National Institute for Health and Welfare, Helsinki, Finland,Department of General Practice and Primary health Care, University of Helsinki, Helsinki, Finland,Unit of General Practice, Helsinki University Central Hospital, Helsinki, Finland
| | - T Espeseth
- K.G. Jebsen Centre for Psychosis Research, Norwegian Centre For Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Department of Psychology, University of Oslo, Oslo, Norway
| | - J D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - I Ford
- Robertson Center for Biostatistics, Glasgow, UK
| | - Generation Scotland
- Generation Scotland, University of Edinburgh Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - R F Gottesman
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - M E Griswold
- Center of Biostatistics and Bioinformatics, University of Mississippi Medical Center, Jackson, MS, USA
| | - V Gudnason
- Icelandic Heart Association, Kopavogur, Iceland,University of Iceland, Reykjavik, Iceland
| | - T B Harris
- Intramural Research Program National Institutes on Aging, National Institutes of Health, Bethesda, MD, USA
| | - G Heiss
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - A Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands,Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - E G Holliday
- Hunter Medical Research Institute and Faculty of Health, University of Newcastle, Newcastle, NSW, Australia
| | - J Huffman
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - S L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - N Kochan
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia,Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - D S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - J B Kwok
- Neuroscience Research Australia, Randwick, NSW, Australia,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - J-C Lambert
- Inserm-UMR744, Institut Pasteur de Lille, Unité d'Epidémiologie et de Santé Publique, Lille, France
| | - T Lee
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia,Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - G Li
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - S-C Li
- Max Planck Institute for Human Development, Berlin, Germany,Technische Universität Dresden, Dresden, Germany
| | - M Loitfelder
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - O L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - A J Lundervold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway,Kavli Research Centre for Aging and Dementia, Haraldsplass Deaconess Hospital, Bergen, Norway,K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| | - A Lundqvist
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - K A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - S S Mirza
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands,Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - L Nyberg
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden,Department of Radiation Sciences, Umeå University, Umeå, Sweden,Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - B A Oostra
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A Palotie
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland,Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - G Papenberg
- Max Planck Institute for Human Development, Berlin, Germany,Karolinska Institutet, Aging Research Center, Stockholm University, Stockholm, Sweden
| | - A Pattie
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - K Petrovic
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - O Polasek
- Faculty of Medicine, Department of Public Health, University of Split, Split, Croatia
| | - B M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA,Deparment of Epidemiology, University of Washington, Seattle, WA, USA,Deparment of Health Services, University of Washington, Seattle, WA, USA,Group Health Research Unit, Group Health Cooperative, Seattle, WA, USA
| | - P Redmond
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - S Reppermund
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - J I Rotter
- Institute for Translational Genomics and Population Sciences Los Angeles BioMedical Research Institute, Harbor-UCLA Medical Center, Los Angeles, CA, USA,Division of Genetic Outcomes, Department of Pediatrics, Harbor-UCLA Medical Center, Los Angeles, CA, USA
| | - H Schmidt
- Department of Neurology, Medical University of Graz, Graz, Austria,Centre for Molecular Medicine, Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - M Schuur
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands,Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - P W Schofield
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| | - R J Scott
- Hunter Medical Research Institute and Faculty of Health, University of Newcastle, Newcastle, NSW, Australia
| | - V M Steen
- K.G. Jebsen Centre for Psychosis Research and the Norwegian Centre for Mental Disorders Research (NORMENT), Department of Clinical Science, University of Bergen, Bergen, Norway,Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - D J Stott
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - J C van Swieten
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - K D Taylor
- Institute for Translational Genomics and Population Sciences Los Angeles BioMedical Research Institute, Harbor-UCLA Medical Center, Los Angeles, CA, USA,Department of Pediatrics, Harbor-UCLA Medical Center, Los Angeles, CA, USA
| | - J Trollor
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia,Department of Developmental Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - S Trompet
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands,Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - A G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands,Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands,Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - G Weinstein
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA,Framingham Heart Study, Framingham, MA, USA
| | - E Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - B G Windham
- Division of Geriatrics, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - J W Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands,Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands,Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - A F Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - M J Wright
- Neuroimaging Genetics Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Q Yang
- Framingham Heart Study, Framingham, MA, USA,Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - H Amieva
- Inserm U897, Université Bordeaux Segalen, Bordeaux, France
| | - J R Attia
- Hunter Medical Research Institute and Faculty of Health, University of Newcastle, Newcastle, NSW, Australia
| | - D A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - H Brodaty
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia,Dementia Collaborative Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - A J M de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands,Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - C Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - M A Ikram
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands,Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands,Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands,Department of Radiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - U Lindenberger
- Max Planck Institute for Human Development, Berlin, Germany
| | - L-G Nilsson
- ARC, Karolinska Institutet, Stockholm and UFBI, Umeå University, Umeå, Sweden
| | - D J Porteous
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, UK,Generation Scotland, University of Edinburgh Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - K Räikkönen
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - I Reinvang
- Department of Psychology, University of Oslo, Oslo, Norway
| | - I Rudan
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, UK
| | - P S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia,Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - R Schmidt
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - P R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia,Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - V Srikanth
- Menzies Research Institute, Hobart, Tasmania,Stroke and Ageing Research, Medicine, Southern Clinical School, Monash University, Melbourne, VIC, Australia
| | - J M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - S T Turner
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - D R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - J F Wilson
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, UK
| | - C van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands,Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - L Launer
- Intramural Research Program National Institutes on Aging, National Institutes of Health, Bethesda, MD, USA
| | - A L Fitzpatrick
- Deparment of Epidemiology, University of Washington, Seattle, WA, USA,Department of Global Health, University of Washington, Seattle, WA, USA
| | - S Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA,Framingham Heart Study, Framingham, MA, USA
| | - T H Mosley
- Division of Geriatrics, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - I J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Department of Psychology, University of Edinburgh, Edinburgh, UK,Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh EH8 9JZ, Scotland, UK. E-mail:
| |
Collapse
|
3089
|
Horr T, Messinger-Rapport B, Pillai JA. Systematic review of strengths and limitations of randomized controlled trials for non-pharmacological interventions in mild cognitive impairment: focus on Alzheimer's disease. J Nutr Health Aging 2015; 19:141-53. [PMID: 25651439 DOI: 10.1007/s12603-014-0565-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Non-pharmacological interventions may improve cognition and quality of life, reduce disruptive behaviors, slow progression from Mild Cognitive Impairment (MCI) to dementia, and delay institutionalization. It is important to look at their trial designs as well as outcomes to understand the state of the evidence supporting non-pharmacological interventions in Alzheimer's disease (AD). An analysis of trial design strengths and limitations may help researchers clarify treatment effect and design future studies of non-pharmacological interventions for MCI related to AD. METHODS A systematic review of the methodology of Randomized Controlled Trials (RCTs) targeting physical activity, cognitive interventions, and socialization among subjects with MCI in AD reported until March 2014 was undertaken. The primary outcome was CONSORT 2010 reporting quality. Secondary outcomes were qualitative assessments of specific methodology problems. RESULTS 23 RCT studies met criteria for this review. Eight focused on physical activity, fourteen on cognitive interventions, and one on the effects of socialization. Most studies found a benefit with the intervention compared to control. CONSORT reporting quality of physical activity interventions was higher than that of cognitive interventions. Reporting quality of recent studies was higher than older studies, particularly with respect to sample size, control characteristics, and methodology of intervention training and delivery. However, the heterogeneity of subjects identified as having MCI and variability in interventions and outcomes continued to limit generalizability. CONCLUSIONS The role for non-pharmacological interventions targeting MCI is promising. Future studies of RCTs for non-pharmacological interventions targeting MCI related to AD may benefit by addressing design limitations.
Collapse
Affiliation(s)
- T Horr
- J.A. Pillai, MBBS, PhD, Staff Neurologist, Lou Ruvo Center for Brain Health, Assistant Professor of Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave / U10, Cleveland, OH 44195, Tel: 216 636 9467, Fax: 216 445 7013, E-mail:
| | | | | |
Collapse
|
3090
|
Vardarajan BN, Zhang Y, Lee JH, Cheng R, Bohm C, Ghani M, Reitz C, Reyes-Dumeyer D, Shen Y, Rogaeva E, St George-Hyslop P, Mayeux R. Coding mutations in SORL1 and Alzheimer disease. Ann Neurol 2015; 77:215-27. [PMID: 25382023 PMCID: PMC4367199 DOI: 10.1002/ana.24305] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 10/10/2014] [Accepted: 10/17/2014] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Common single nucleotide polymorphisms in the SORL1 gene have been associated with late onset Alzheimer disease (LOAD), but causal variants have not been fully characterized nor has the mechanism been established. The study was undertaken to identify functional SORL1 mutations in patients with LOAD. METHODS This was a family- and cohort-based genetic association study. Caribbean Hispanics with familial and sporadic LOAD and similarly aged controls were recruited from the United States and the Dominican Republic, and patients with sporadic disease of Northern European origin were recruited from Canada. Prioritized coding variants in SORL1 were detected by targeted resequencing and validated by genotyping in additional family members and unrelated healthy controls. Variants transfected into human embryonic kidney 293 cell lines were tested for Aβ40 and Aβ42 secretion, and the amount of the amyloid precursor protein (APP) secreted at the cell surface was determined. RESULTS Seventeen coding exonic variants were significantly associated with disease. Two rare variants (rs117260922-E270K and rs143571823-T947M) with minor allele frequency (MAF) < 1% and 1 common variant (rs2298813-A528T) with MAF = 14.9% segregated within families and were deemed deleterious to the coding protein. Transfected cell lines showed increased Aβ40 and Aβ42 secretion for the rare variants (E270K and T947M) and increased Aβ42 secretion for the common variant (A528T). All mutants increased the amount of APP at the cell surface, although in slightly different ways, thereby failing to direct full-length APP into the retromer-recycling endosome pathway. INTERPRETATION Common and rare variants in SORL1 elevate the risk of LOAD by directly affecting APP processing, which in turn can result in increased Aβ40 and Aβ42 secretion.
Collapse
Affiliation(s)
- Badri N. Vardarajan
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Yalun Zhang
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Medicine, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, Ontario, Canada, M5T 2S8
| | - Joseph H. Lee
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Rong Cheng
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Christopher Bohm
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Medicine, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, Ontario, Canada, M5T 2S8
| | - Mahdi Ghani
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Medicine, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, Ontario, Canada, M5T 2S8
| | - Christiane Reitz
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- The Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Dolly Reyes-Dumeyer
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Yufeng Shen
- The Department Systems Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Medicine, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, Ontario, Canada, M5T 2S8
| | - Peter St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Medicine, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, Ontario, Canada, M5T 2S8
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Richard Mayeux
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- The Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- The Department Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- The Department of Epidemiology, School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
3091
|
Chaudhry M, Hasnain S, Wang X, Snitz BE, Bamne MN, Rosenthal S, Yesim Demirci F, Ilyas Kamboh M. Association analysis of 23 susceptibility loci with risk of dementia in a Pakistani population. Psychiatry Res 2015; 225:223-224. [PMID: 25467701 DOI: 10.1016/j.psychres.2014.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 10/11/2014] [Accepted: 10/30/2014] [Indexed: 11/24/2022]
Affiliation(s)
- Mamoonah Chaudhry
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan.
| | - Shahida Hasnain
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan; The Women University Multan, Multan, Pakistan
| | - Xingbin Wang
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beth E Snitz
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mikhil N Bamne
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Samantha Rosenthal
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - F Yesim Demirci
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
3092
|
CLU rs9331888 Polymorphism Contributes to Alzheimer's Disease Susceptibility in Caucasian But Not East Asian Populations. Mol Neurobiol 2015; 53:1446-1451. [PMID: 25633098 DOI: 10.1007/s12035-015-9098-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 01/12/2015] [Indexed: 12/22/2022]
Abstract
Large-scale genome-wide association studies (GWAS) identified three single nucleotide polymorphisms rs11136000, rs2279590, and rs9331888 in CLU gene to be significantly associated with Alzheimer's disease (AD) in Caucasian ancestry. Both rs11136000 and rs2279590 variants were successfully replicated in Asian population. However, previous studies reported either a weak association or no association between rs9331888 polymorphism and AD in Asian population. Here, we searched the PubMed, AlzGene, and Google Scholar databases. We selected 12 independent studies that evaluated the association between the rs9331888 polymorphism and AD using a case-control design. Using an additive model, we did not identify significant heterogeneity among these 12 studies. We observed significant association between rs9331888 polymorphism and AD in pooled populations (P = 2.26E - 07, odds ratio (OR) = 1.10, 95% confidence interval (CI) 1.06-1.14). In subgroup analysis, we did not identify significant heterogeneity in both Asian and Caucasian populations. We identified significant association in Caucasian population (P = 1.67E - 08, OR = 1.13, 95% CI 1.08-1.18) but not in East Asian population (P = 0.49, OR = 1.02, 95% CI 0.96-1.10).
Collapse
|
3093
|
Baleriola J, Walker CA, Jean YY, Crary JF, Troy CM, Nagy PL, Hengst U. Axonally synthesized ATF4 transmits a neurodegenerative signal across brain regions. Cell 2015; 158:1159-1172. [PMID: 25171414 DOI: 10.1016/j.cell.2014.07.001] [Citation(s) in RCA: 264] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 05/23/2014] [Accepted: 07/01/2014] [Indexed: 11/16/2022]
Abstract
In Alzheimer's disease (AD) brain, exposure of axons to Aβ causes pathogenic changes that spread retrogradely by unknown mechanisms, affecting the entire neuron. We found that locally applied Aβ1-42 initiates axonal synthesis of a defined set of proteins including the transcription factor ATF4. Inhibition of local translation and retrograde transport or knockdown of axonal Atf4 mRNA abolished Aβ-induced ATF4 transcriptional activity and cell loss. Aβ1-42 injection into the dentate gyrus (DG) of mice caused loss of forebrain neurons whose axons project to the DG. Protein synthesis and Atf4 mRNA were upregulated in these axons, and coinjection of Atf4 siRNA into the DG reduced the effects of Aβ1-42 in the forebrain. ATF4 protein and transcripts were found with greater frequency in axons in the brain of AD patients. These results reveal an active role for intra-axonal translation in neurodegeneration and identify ATF4 as a mediator for the spread of AD pathology.
Collapse
Affiliation(s)
- Jimena Baleriola
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Chandler A Walker
- Integrated Program in Cellular, Molecular and Biomedical Studies, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Ying Y Jean
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - John F Crary
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Carol M Troy
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Peter L Nagy
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Ulrich Hengst
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
3094
|
Beach TG, Adler CH, Sue LI, Serrano G, Shill HA, Walker DG, Lue L, Roher AE, Dugger BN, Maarouf C, Birdsill AC, Intorcia A, Saxon-Labelle M, Pullen J, Scroggins A, Filon J, Scott S, Hoffman B, Garcia A, Caviness JN, Hentz JG, Driver-Dunckley E, Jacobson SA, Davis KJ, Belden CM, Long KE, Malek-Ahmadi M, Powell JJ, Gale LD, Nicholson LR, Caselli RJ, Woodruff BK, Rapscak SZ, Ahern GL, Shi J, Burke AD, Reiman EM, Sabbagh MN. Arizona Study of Aging and Neurodegenerative Disorders and Brain and Body Donation Program. Neuropathology 2015; 35:354-89. [PMID: 25619230 DOI: 10.1111/neup.12189] [Citation(s) in RCA: 356] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/11/2014] [Indexed: 12/13/2022]
Abstract
The Brain and Body Donation Program (BBDP) at Banner Sun Health Research Institute (http://www.brainandbodydonationprogram.org) started in 1987 with brain-only donations and currently has banked more than 1600 brains. More than 430 whole-body donations have been received since this service was commenced in 2005. The collective academic output of the BBDP is now described as the Arizona Study of Aging and Neurodegenerative Disorders (AZSAND). Most BBDP subjects are enrolled as cognitively normal volunteers residing in the retirement communities of metropolitan Phoenix, Arizona. Specific recruitment efforts are also directed at subjects with Alzheimer's disease, Parkinson's disease and cancer. The median age at death is 82. Subjects receive standardized general medical, neurological, neuropsychological and movement disorders assessments during life and more than 90% receive full pathological examinations by medically licensed pathologists after death. The Program has been funded through a combination of internal, federal and state of Arizona grants as well as user fees and pharmaceutical industry collaborations. Subsets of the Program are utilized by the US National Institute on Aging Arizona Alzheimer's Disease Core Center and the US National Institute of Neurological Disorders and Stroke National Brain and Tissue Resource for Parkinson's Disease and Related Disorders. Substantial funding has also been received from the Michael J. Fox Foundation for Parkinson's Research. The Program has made rapid autopsy a priority, with a 3.0-hour median post-mortem interval for the entire collection. The median RNA Integrity Number (RIN) for frozen brain and body tissue is 8.9 and 7.4, respectively. More than 2500 tissue requests have been served and currently about 200 are served annually. These requests have been made by more than 400 investigators located in 32 US states and 15 countries. Tissue from the BBDP has contributed to more than 350 publications and more than 200 grant-funded projects.
Collapse
Affiliation(s)
- Thomas G Beach
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Lucia I Sue
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Geidy Serrano
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Holly A Shill
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - LihFen Lue
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Alex E Roher
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Chera Maarouf
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Alex C Birdsill
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | | - Joel Pullen
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Jessica Filon
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Sarah Scott
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Angelica Garcia
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | | | | | | - Kathryn J Davis
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Kathy E Long
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | | - Lisa D Gale
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | | | | | | | | - Jiong Shi
- Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Anna D Burke
- Banner Alzheimer Institute, Phoenix, Arizona, USA
| | | | | |
Collapse
|
3095
|
Ramsden HL, Sürmeli G, McDonagh SG, Nolan MF. Laminar and dorsoventral molecular organization of the medial entorhinal cortex revealed by large-scale anatomical analysis of gene expression. PLoS Comput Biol 2015; 11:e1004032. [PMID: 25615592 PMCID: PMC4304787 DOI: 10.1371/journal.pcbi.1004032] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 11/10/2014] [Indexed: 12/14/2022] Open
Abstract
Neural circuits in the medial entorhinal cortex (MEC) encode an animal's position and orientation in space. Within the MEC spatial representations, including grid and directional firing fields, have a laminar and dorsoventral organization that corresponds to a similar topography of neuronal connectivity and cellular properties. Yet, in part due to the challenges of integrating anatomical data at the resolution of cortical layers and borders, we know little about the molecular components underlying this organization. To address this we develop a new computational pipeline for high-throughput analysis and comparison of in situ hybridization (ISH) images at laminar resolution. We apply this pipeline to ISH data for over 16,000 genes in the Allen Brain Atlas and validate our analysis with RNA sequencing of MEC tissue from adult mice. We find that differential gene expression delineates the borders of the MEC with neighboring brain structures and reveals its laminar and dorsoventral organization. We propose a new molecular basis for distinguishing the deep layers of the MEC and show that their similarity to corresponding layers of neocortex is greater than that of superficial layers. Our analysis identifies ion channel-, cell adhesion- and synapse-related genes as candidates for functional differentiation of MEC layers and for encoding of spatial information at different scales along the dorsoventral axis of the MEC. We also reveal laminar organization of genes related to disease pathology and suggest that a high metabolic demand predisposes layer II to neurodegenerative pathology. In principle, our computational pipeline can be applied to high-throughput analysis of many forms of neuroanatomical data. Our results support the hypothesis that differences in gene expression contribute to functional specialization of superficial layers of the MEC and dorsoventral organization of the scale of spatial representations.
Collapse
Affiliation(s)
- Helen L. Ramsden
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- Neuroinformatics Doctoral Training Centre, School of Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Gülşen Sürmeli
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Steven G. McDonagh
- Institute of Perception, Action and Behaviour, School of Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Matthew F. Nolan
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Brain Development and Repair, inStem, Bangalore, India
- * E-mail:
| |
Collapse
|
3096
|
Chakrabarty P, Li A, Ceballos-Diaz C, Eddy JA, Funk CC, Moore B, DiNunno N, Rosario AM, Cruz PE, Verbeeck C, Sacino A, Nix S, Janus C, Price ND, Das P, Golde TE. IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron 2015; 85:519-33. [PMID: 25619653 DOI: 10.1016/j.neuron.2014.11.020] [Citation(s) in RCA: 286] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 10/24/2014] [Accepted: 11/20/2014] [Indexed: 01/27/2023]
Abstract
Anti-inflammatory strategies are proposed to have beneficial effects in Alzheimer's disease. To explore how anti-inflammatory cytokine signaling affects Aβ pathology, we investigated the effects of adeno-associated virus (AAV2/1)-mediated expression of Interleukin (IL)-10 in the brains of APP transgenic mouse models. IL-10 expression resulted in increased Aβ accumulation and impaired memory in APP mice. A focused transcriptome analysis revealed changes consistent with enhanced IL-10 signaling and increased ApoE expression in IL-10-expressing APP mice. ApoE protein was selectively increased in the plaque-associated insoluble cellular fraction, likely because of direct interaction with aggregated Aβ in the IL-10-expressing APP mice. Ex vivo studies also show that IL-10 and ApoE can individually impair glial Aβ phagocytosis. Our observations that IL-10 has an unexpected negative effect on Aβ proteostasis and cognition in APP mouse models demonstrate the complex interplay between innate immunity and proteostasis in neurodegenerative diseases, an interaction we call immunoproteostasis.
Collapse
Affiliation(s)
- Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Andrew Li
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Carolina Ceballos-Diaz
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - James A Eddy
- Institute for Systems Biology, 401 Terry Avenue N, Seattle, WA 98109, USA
| | - Cory C Funk
- Institute for Systems Biology, 401 Terry Avenue N, Seattle, WA 98109, USA
| | - Brenda Moore
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Nadia DiNunno
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Awilda M Rosario
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Pedro E Cruz
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Christophe Verbeeck
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | - Amanda Sacino
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Sarah Nix
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | - Christopher Janus
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Nathan D Price
- Institute for Systems Biology, 401 Terry Avenue N, Seattle, WA 98109, USA
| | - Pritam Das
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | - Todd E Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
3097
|
Ge T, Nichols TE, Ghosh D, Mormino EC, Smoller JW, Sabuncu MR. A kernel machine method for detecting effects of interaction between multidimensional variable sets: an imaging genetics application. Neuroimage 2015; 109:505-514. [PMID: 25600633 DOI: 10.1016/j.neuroimage.2015.01.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 01/06/2015] [Accepted: 01/09/2015] [Indexed: 11/19/2022] Open
Abstract
Measurements derived from neuroimaging data can serve as markers of disease and/or healthy development, are largely heritable, and have been increasingly utilized as (intermediate) phenotypes in genetic association studies. To date, imaging genetic studies have mostly focused on discovering isolated genetic effects, typically ignoring potential interactions with non-genetic variables such as disease risk factors, environmental exposures, and epigenetic markers. However, identifying significant interaction effects is critical for revealing the true relationship between genetic and phenotypic variables, and shedding light on disease mechanisms. In this paper, we present a general kernel machine based method for detecting effects of the interaction between multidimensional variable sets. This method can model the joint and epistatic effect of a collection of single nucleotide polymorphisms (SNPs), accommodate multiple factors that potentially moderate genetic influences, and test for nonlinear interactions between sets of variables in a flexible framework. As a demonstration of application, we applied the method to the data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) to detect the effects of the interactions between candidate Alzheimer's disease (AD) risk genes and a collection of cardiovascular disease (CVD) risk factors, on hippocampal volume measurements derived from structural brain magnetic resonance imaging (MRI) scans. Our method identified that two genes, CR1 and EPHA1, demonstrate significant interactions with CVD risk factors on hippocampal volume, suggesting that CR1 and EPHA1 may play a role in influencing AD-related neurodegeneration in the presence of CVD risks.
Collapse
Affiliation(s)
- Tian Ge
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital / Harvard Medical School, Charlestown, MA 02129, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Thomas E Nichols
- Department of Statistics & Warwick Manufacturing Group, The University of Warwick, Coventry CV4 7AL, UK
| | - Debashis Ghosh
- Department of Statistics, The Pennsylvania State University, PA 16802, USA
| | - Elizabeth C Mormino
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02138, USA
| | - Mert R Sabuncu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital / Harvard Medical School, Charlestown, MA 02129, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
3098
|
Li X, Montine KS, Keene CD, Montine TJ. Different mechanisms of apolipoprotein E isoform-dependent modulation of prostaglandin E2 production and triggering receptor expressed on myeloid cells 2 (TREM2) expression after innate immune activation of microglia. FASEB J 2015; 29:1754-62. [PMID: 25593125 DOI: 10.1096/fj.14-262683] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/15/2014] [Indexed: 01/14/2023]
Abstract
Several lines of evidence support immune response in brain as a mechanism of injury in Alzheimer disease (AD). Moreover, immune activation is heightened in apolipoprotein E (APOE) ε4 carriers; inhibitors of prostaglandin (PG) synthesis show a partially protective effect on AD risk from APOE ε4; and genetic variants in triggering receptor expressed on myeloid cells 2 (TREM2) are a rare but potent risk for AD. We tested the hypothesis that APOE ε4 inheritance modulates both the PGE2 pathway and TREM2 expression using primary murine microglia from targeted replacement (TR) APOE3/3 and APOE4/4 mice. Microglial cyclooxygenase-2, microsomal PGE synthase, and PGE2 expression were increased 2- to 25-fold in both genotypes by TLR activators; however, this induction was significantly (P < 0.01) greater in TR APOE4/4 microglia with TLR3 and TLR4 activators. Microglial TREM2 expression was reduced approximately 85% by all TLR activators; this reduction was approximately one-third greater in microglia from TR APOE4/4 mice. Importantly, both receptor-associated protein and a nuclear factor κ-light-chain-enhancer inhibitor blocked TR APOE4/4-dependent effects on the PGE2 pathway but not on TREM2 expression. These data demonstrate complementary, but mechanistically distinct, regulation of pro- and anti-inflammatory mediators in TR APOE4/4 murine microglia that yields a more proinflammatory state than with TR APOE3/3.
Collapse
Affiliation(s)
- Xianwu Li
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Kathleen S Montine
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Thomas J Montine
- Department of Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
3099
|
Proitsi P, Kim M, Whiley L, Pritchard M, Leung R, Soininen H, Kloszewska I, Mecocci P, Tsolaki M, Vellas B, Sham P, Lovestone S, Powell JF, Dobson RJB, Legido-Quigley C. Plasma lipidomics analysis finds long chain cholesteryl esters to be associated with Alzheimer's disease. Transl Psychiatry 2015; 5:e494. [PMID: 25585166 PMCID: PMC4312824 DOI: 10.1038/tp.2014.127] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 09/28/2014] [Accepted: 10/26/2014] [Indexed: 12/17/2022] Open
Abstract
There is an urgent need for the identification of Alzheimer's disease (AD) biomarkers. Studies have now suggested the promising use of associations with blood metabolites as functional intermediate phenotypes in biomedical and pharmaceutical research. The aim of this study was to use lipidomics to identify a battery of plasma metabolite molecules that could predict AD patients from controls. We performed a comprehensive untargeted lipidomic analysis, using ultra-performance liquid chromatography/mass spectrometry on plasma samples from 35 AD patients, 40 elderly controls and 48 individuals with mild cognitive impairment (MCI) and used multivariate analysis methods to identify metabolites associated with AD status. A combination of 10 metabolites could discriminate AD patients from controls with 79.2% accuracy (81.8% sensitivity, 76.9% specificity and an area under curve of 0.792) in a novel test set. Six of the metabolites were identified as long chain cholesteryl esters (ChEs) and were reduced in AD (ChE 32:0, odds ratio (OR)=0.237, 95% confidence interval (CI)=0.10-0.48, P=4.19E-04; ChE 34:0, OR=0.152, 95% CI=0.05-0.37, P=2.90E-04; ChE 34:6, OR=0.126, 95% CI=0.03-0.35, P=5.40E-04; ChE 32:4, OR=0.056, 95% CI=0.01-0.24, P=6.56E-04 and ChE 33:6, OR=0.205, 95% CI=0.06-0.50, P=2.21E-03, per (log2) metabolite unit). The levels of these metabolites followed the trend control>MCI>AD. We, additionally, found no association between cholesterol, the precursor of ChE and AD. This study identified new ChE molecules, involved in cholesterol metabolism, implicated in AD, which may help identify new therapeutic targets; although, these findings need to be replicated in larger well-phenotyped cohorts.
Collapse
Affiliation(s)
- P Proitsi
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - M Kim
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - L Whiley
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - M Pritchard
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - R Leung
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - H Soininen
- Department of Neurology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland
| | - I Kloszewska
- Department of Old Age Psychiatry & Psychotic Disorders, Medical University of Łódź, Łódź, Poland
| | - P Mecocci
- Section of Gerontology and Geriatrics, Department of Medicine, University of Perugia, Perugia, Italy
| | - M Tsolaki
- Memory and Dementia Center, 3rd Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - B Vellas
- Department of Internal and Geriatrics Medicine, INSERM U 1027, Gerontopole, Hôpitaux de Toulouse, Toulouse, France
| | - P Sham
- Department of Psychiatry, State Key Laboratory of Brain and Cognitive Sciences, and Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - S Lovestone
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - J F Powell
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - R J B Dobson
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- National Institute for Health Research Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - C Legido-Quigley
- Institute of Pharmaceutical Science, King's College London, London, UK
| |
Collapse
|
3100
|
Mahmoudi R, Kisserli A, Novella JL, Donvito B, Dramé M, Réveil B, Duret V, Jolly D, Pham BN, Cohen JH. Alzheimer's disease is associated with low density of the long CR1 isoform. Neurobiol Aging 2015; 36:1766.e5-1766.e12. [PMID: 25666996 DOI: 10.1016/j.neurobiolaging.2015.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 10/31/2014] [Accepted: 01/05/2015] [Indexed: 01/21/2023]
Abstract
The long complement receptor type 1 (CR1) isoform, CR1*2 (S), has been identified as being associated with Alzheimer's disease (AD) risk. We aimed to analyze the phenotypic structural and expression aspects (length and density) of CR1 in erythrocytes of 135 Caucasian subjects (100 AD and 35 controls). CR1 length polymorphism was assessed at protein and gene levels using Western blot and high-resolution melting, respectively. CR1 sites on erythrocytes were enumerated by flow cytometry. CR1 gene analysis, spotting the rs6656401 and rs3818361 polymorphisms, was performed by pyrosequencing. The CR1 density was significantly lower in AD patients expressing the CR1*2 isoform compared with the controls (p = 0.001), demonstrating lower expression of CR1 in CR1*2 carriers. Our data suggested the existence of silent CR1 alleles. Finally, rs6656401 and rs3818361 were strongly associated with CR1 length polymorphism (p < 0.0001). These observations indicate that AD susceptibility is associated with the long CR1 isoform (CR1*2), albeit at a lower density, suggesting that AD results from insufficient clearance of plaque deposits rather than increased inflammation.
Collapse
Affiliation(s)
- Rachid Mahmoudi
- Champagne-Ardenne Resource and Research Memory Center (CMRR), Maison Blanche Hospital, Reims University Hospitals, Reims, France; Department of Internal Medicine and Geriatrics, Maison Blanche Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, EA 3797, Reims, France.
| | - Aymric Kisserli
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| | - Jean-Luc Novella
- Champagne-Ardenne Resource and Research Memory Center (CMRR), Maison Blanche Hospital, Reims University Hospitals, Reims, France; Department of Internal Medicine and Geriatrics, Maison Blanche Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, EA 3797, Reims, France
| | - Béatrice Donvito
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| | - Moustapha Dramé
- Faculty of Medicine, University of Reims Champagne-Ardenne, EA 3797, Reims, France; Department of Research and Innovation, Robert Debré Hospital, Reims University Hospitals, Reims, France
| | - Brigitte Réveil
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| | - Valérie Duret
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| | - Damien Jolly
- Faculty of Medicine, University of Reims Champagne-Ardenne, EA 3797, Reims, France; Department of Research and Innovation, Robert Debré Hospital, Reims University Hospitals, Reims, France
| | - Bach-Nga Pham
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| | - Jacques H Cohen
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| |
Collapse
|