3151
|
Medway C, Morgan K. Review: The genetics of Alzheimer's disease; putting flesh on the bones. Neuropathol Appl Neurobiol 2014; 40:97-105. [PMID: 24443964 PMCID: PMC4282344 DOI: 10.1111/nan.12101] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 11/21/2013] [Indexed: 01/14/2023]
Abstract
For two decades the search for genes involved in Alzheimer's disease brought little reward; it was not until the advent of genome-wide association studies (GWAS) that genetic associations started to be revealed. Since 2009 increasingly large GWAS have revealed 20 loci, which in itself is a substantial increase in our understanding, but perhaps the more important feature is that these studies have highlighted novel pathways that are potentially involved in the disease process. This commentary assembles our latest knowledge while acknowledging that the casual functional variants, and undoubtedly, other genes are still yet to be discovered. This is the challenge that remains and the promise of next-generation sequencing is anticipated as there are a number of large initiatives which themselves should start to yield information before long.
Collapse
Affiliation(s)
- C Medway
- Translation Cell Sciences - Human Genetics, School of Life Sciences, Queens Medical Centre, Nottingham, UK
| | | |
Collapse
|
3152
|
Common genetic variants associated with cognitive performance identified using the proxy-phenotype method. Proc Natl Acad Sci U S A 2014; 111:13790-4. [PMID: 25201988 DOI: 10.1073/pnas.1404623111] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
We identify common genetic variants associated with cognitive performance using a two-stage approach, which we call the proxy-phenotype method. First, we conduct a genome-wide association study of educational attainment in a large sample (n = 106,736), which produces a set of 69 education-associated SNPs. Second, using independent samples (n = 24,189), we measure the association of these education-associated SNPs with cognitive performance. Three SNPs (rs1487441, rs7923609, and rs2721173) are significantly associated with cognitive performance after correction for multiple hypothesis testing. In an independent sample of older Americans (n = 8,652), we also show that a polygenic score derived from the education-associated SNPs is associated with memory and absence of dementia. Convergent evidence from a set of bioinformatics analyses implicates four specific genes (KNCMA1, NRXN1, POU2F3, and SCRT). All of these genes are associated with a particular neurotransmitter pathway involved in synaptic plasticity, the main cellular mechanism for learning and memory.
Collapse
|
3153
|
Transcriptomics and mechanistic elucidation of Alzheimer's disease risk genes in the brain and in vitro models. Neurobiol Aging 2014; 36:1221.e15-28. [PMID: 25281018 DOI: 10.1016/j.neurobiolaging.2014.09.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 08/08/2014] [Accepted: 09/01/2014] [Indexed: 01/10/2023]
Abstract
In this study, we have assessed the expression and splicing status of genes involved in the pathogenesis or affecting the risk of Alzheimer's disease (AD) in the postmortem inferior temporal cortex samples obtained from 60 subjects with varying degree of AD-related neurofibrillary pathology. These subjects were grouped based on neurofibrillary pathology into 3 groups: Braak stages 0-II, Braak stages III-IV, and Braak stages V-VI. We also examined the right frontal cortical biopsies obtained during life from 22 patients with idiopathic shunt-responding normal pressure hydrocephalus, a disease that displays similar pathologic alterations as seen in AD. These 22 patients were categorized according to dichotomized amyloid-β positive or negative pathology in the biopsies. We observed that the expression of FRMD4A significantly decreased, and the expression of MS4A6A significantly increased in relation to increasing AD-related neurofibrillary pathology. Moreover, the expression of 2 exons in both CLU and TREM2 significantly increased with increase in AD-related neurofibrillary pathology. However, a similar trend toward increased expression in CLU and TREM2 was observed with most of the studied exons, suggesting a global change in the expression rather than altered splicing. Correlation of gene expression with well-established AD-related factors, such as α-, β-, and γ-secretase activities, brain amyloid-β42 levels, and cerebrospinal fluid biomarkers, revealed a positive correlation between β-secretase activity and the expression of TREM2 and BIN1. In expression quantitative trait loci analysis, we did not detect significant effects of the risk alleles on gene expression or splicing. Analysis of the normal pressure hydrocephalus biopsies revealed no differences in the expression or splicing profiles of the studied genes between amyloid-β positive and negative patients. Using the protein-protein interaction-based in vitro pathway analysis tools, we found that downregulation of FRMD4A associated with increased APP-β-secretase interaction, increased amyloid-β40 secretion, and altered phosphorylation of tau. Taken together, our results suggest that the expression of FRMD4A, MS4A6A, CLU, and TREM2 is altered in relation to increasing AD-related neurofibrillary pathology, and that FRMD4A may play a role in amyloidogenic and tau-related pathways in AD. Therefore, investigation of gene expression changes in the brain and effects of the identified genes on disease-associated pathways in vitro may provide mechanistic insights on how alterations in these genes may contribute to AD pathogenesis.
Collapse
|
3154
|
Beecham GW, Hamilton K, Naj AC, Martin ER, Huentelman M, Myers AJ, Corneveaux JJ, Hardy J, Vonsattel JP, Younkin SG, Bennett DA, De Jager PL, Larson EB, Crane PK, Kamboh MI, Kofler JK, Mash DC, Duque L, Gilbert JR, Gwirtsman H, Buxbaum JD, Kramer P, Dickson DW, Farrer LA, Frosch MP, Ghetti B, Haines JL, Hyman BT, Kukull WA, Mayeux RP, Pericak-Vance MA, Schneider JA, Trojanowski JQ, Reiman EM, the Alzheimer's Disease Genetics Consortium (ADGC), Schellenberg GD, Montine TJ. Genome-wide association meta-analysis of neuropathologic features of Alzheimer's disease and related dementias. PLoS Genet 2014; 10:e1004606. [PMID: 25188341 PMCID: PMC4154667 DOI: 10.1371/journal.pgen.1004606] [Citation(s) in RCA: 263] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 07/14/2014] [Indexed: 01/11/2023] Open
Abstract
Alzheimer's disease (AD) and related dementias are a major public health challenge and present a therapeutic imperative for which we need additional insight into molecular pathogenesis. We performed a genome-wide association study and analysis of known genetic risk loci for AD dementia using neuropathologic data from 4,914 brain autopsies. Neuropathologic data were used to define clinico-pathologic AD dementia or controls, assess core neuropathologic features of AD (neuritic plaques, NPs; neurofibrillary tangles, NFTs), and evaluate commonly co-morbid neuropathologic changes: cerebral amyloid angiopathy (CAA), Lewy body disease (LBD), hippocampal sclerosis of the elderly (HS), and vascular brain injury (VBI). Genome-wide significance was observed for clinico-pathologic AD dementia, NPs, NFTs, CAA, and LBD with a number of variants in and around the apolipoprotein E gene (APOE). GalNAc transferase 7 (GALNT7), ATP-Binding Cassette, Sub-Family G (WHITE), Member 1 (ABCG1), and an intergenic region on chromosome 9 were associated with NP score; and Potassium Large Conductance Calcium-Activated Channel, Subfamily M, Beta Member 2 (KCNMB2) was strongly associated with HS. Twelve of the 21 non-APOE genetic risk loci for clinically-defined AD dementia were confirmed in our clinico-pathologic sample: CR1, BIN1, CLU, MS4A6A, PICALM, ABCA7, CD33, PTK2B, SORL1, MEF2C, ZCWPW1, and CASS4 with 9 of these 12 loci showing larger odds ratio in the clinico-pathologic sample. Correlation of effect sizes for risk of AD dementia with effect size for NFTs or NPs showed positive correlation, while those for risk of VBI showed a moderate negative correlation. The other co-morbid neuropathologic features showed only nominal association with the known AD loci. Our results discovered new genetic associations with specific neuropathologic features and aligned known genetic risk for AD dementia with specific neuropathologic changes in the largest brain autopsy study of AD and related dementias. Alzheimer's disease (AD) and related dementias are a major public health challenge and present a therapeutic imperative for which we need additional insight into molecular pathogenesis. We performed a genome-wide association study (GWAS), as well as an analysis of known genetic risk loci for AD dementia, using data from 4,914 brain autopsies. Genome-wide significance was observed for 7 genes and pathologic features of AD and related diseases. Twelve of the 22 genetic risk loci for clinically-defined AD dementia were confirmed in our pathologic sample. Correlation of effect sizes for risk of AD dementia with effect size for hallmark pathologic features of AD were strongly positive and linear. Our study discovered new genetic associations with specific pathologic features and aligned known genetic risk for AD dementia with specific pathologic changes in a large brain autopsy study of AD and related dementias.
Collapse
Affiliation(s)
- Gary W. Beecham
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Kara Hamilton
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Adam C. Naj
- Division of Epidemiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Eden R. Martin
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Matt Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Amanda J. Myers
- Department of Psychiatry & Behavioral Sciences, University of Miami, Miami, Florida, United States of America
| | - Jason J. Corneveaux
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - John Hardy
- Department of Molecular Neuroscience, University College London, London, United Kingdom
| | - Jean-Paul Vonsattel
- New York Brain Bank, Columbia University, New York, New York, United States of America
| | - Steven G. Younkin
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida, United States of America
| | - David A. Bennett
- Department of Neurological Sciences, Rush University, Chicago, Illinois, United States of America
| | - Philip L. De Jager
- Department of Neurology and Psychiatry, Brigham and Women's Hospital, Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Eric B. Larson
- Group Health Research Institute, Seattle, Washington, United States of America
| | - Paul K. Crane
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - M. Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Julia K. Kofler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Deborah C. Mash
- Department of Neurology, University of Miami, Miami, Florida, United States of America
| | - Linda Duque
- Department of Neurology, University of Miami, Miami, Florida, United States of America
| | - John R. Gilbert
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Harry Gwirtsman
- Department of Psychiatry, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Joseph D. Buxbaum
- Department of Psychiatry, Mount Sinai Hospital, New York, New York, United States of America
| | - Patricia Kramer
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida, United States of America
| | - Lindsay A. Farrer
- Biomedical Genetics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Matthew P. Frosch
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, Indiana, United States of America
| | - Jonathan L. Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Bradley T. Hyman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Walter A. Kukull
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, Washington, United States of America
| | - Richard P. Mayeux
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Margaret A. Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Julie A. Schneider
- Department of Neurological Sciences, Rush University, Chicago, Illinois, United States of America
- Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, Illinois, United States of America
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Eric M. Reiman
- Arizona Alzheimer's Consortium, Banner Alzheimer's Institute, Phoenix, Arizona, United States of America
- Department of Psychiatry, University of Arizona, Phoenix, Arizona, United States of America
| | | | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Thomas J. Montine
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
3155
|
Li X, Shen N, Zhang S, Liu J, Jiang Q, Liao M, Feng R, Zhang L, Wang G, Ma G, Zhou H, Chen Z, Jiang Y, Zhao B, Li K, Liu G. CD33 rs3865444 Polymorphism Contributes to Alzheimer's Disease Susceptibility in Chinese, European, and North American Populations. Mol Neurobiol 2014; 52:414-21. [PMID: 25186233 DOI: 10.1007/s12035-014-8880-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/25/2014] [Indexed: 11/30/2022]
Abstract
The CD33 rs3865444 polymorphism was first identified to be associated with Alzheimer's disease (AD) in European population. However, the following studies reported weak or no significant association in Chinese, Japanese, Korean, American, and Canadian populations. We think that these negative results may have been caused by either relatively small sample sizes compared with those used for the previous GWAS in European ancestry or the genetic heterogeneity of the rs3865444 polymorphism in different populations. Here, we reevaluated this association using the relatively large-scale samples from previous 27 studies (N = 86,759; 31,106 cases and 55,653 controls) by searching the PubMed, AlzGene, and Google Scholar databases. We identified significant heterogeneity and observed no significant association between the rs3865444 polymorphism and AD in pooled populations (P = 0.264, odds ratio (OR) = 0.97, 95% confidence interval (CI) 0.93-1.02). In subgroup analysis, we identified significant heterogeneity only in East Asian population and observed no significant association between the rs3865444 polymorphism and AD. We further identified significant heterogeneity and observed significant association between the rs3865444 polymorphism and AD in Chinese population. We identified no significant heterogeneity and significant association in North American and European populations. Collectively, our analysis shows that the CD33 rs3865444 polymorphism is associated with AD susceptibility in Chinese, European, and North American populations. We believe that our findings will be very useful for future genetic studies on AD.
Collapse
Affiliation(s)
- Xingwang Li
- Department of Anesthesiology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3156
|
Xu W, Tan L, Yu JT. The Role of PICALM in Alzheimer's Disease. Mol Neurobiol 2014; 52:399-413. [PMID: 25186232 DOI: 10.1007/s12035-014-8878-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/25/2014] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is a highly heritable disease (with heritability up to 76%) with a complex genetic profile of susceptibility, among which large genome-wide association studies (GWASs) pointed to the phosphatidylinositol-binding clathrin assembly protein (PICALM) gene as a susceptibility locus for late-onset Alzheimer's disease (LOAD) incidence. Here, we summarize the known functions of PICALM and discuss its genetic polymorphisms and their potential physiological effects associated with LOAD. Compelling data indicated that PICALM affects AD risk primarily by modulating production, transportation, and clearance of β-amyloid (Aβ) peptide, but other Aβ-independent pathways are discussed, including tauopathy, synaptic dysfunction, disorganized lipid metabolism, immune disorder, and disrupted iron homeostasis. Finally, given the potential involvement of PICALM in facilitating AD occurrence in multiple ways, it might be possible that targeting PICALM might provide promising and novel avenues for AD therapy.
Collapse
Affiliation(s)
- Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | | | | |
Collapse
|
3157
|
Dauncey MJ. Nutrition, the brain and cognitive decline: insights from epigenetics. Eur J Clin Nutr 2014; 68:1179-85. [PMID: 25182020 DOI: 10.1038/ejcn.2014.173] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 07/11/2014] [Indexed: 02/07/2023]
Abstract
Nutrition affects the brain throughout life, with profound implications for cognitive decline and dementia. These effects are mediated by changes in expression of multiple genes, and responses to nutrition are in turn affected by individual genetic variability. An important layer of regulation is provided by the epigenome: nutrition is one of the many epigenetic regulators that modify gene expression without changes in DNA sequence. Epigenetic mechanisms are central to brain development, structure and function, and include DNA methylation, histone modifications and non-protein-coding RNAs. They enable cell-specific and age-related gene expression. Although epigenetic events can be highly stable, they can also be reversible, highlighting a critical role for nutrition in prevention and treatment of disease. Moreover, they suggest key mechanisms by which nutrition is involved in the pathogenesis of age-related cognitive decline: many nutrients, foods and diets have both immediate and long-term effects on the epigenome, including energy status, that is, energy intake, physical activity, energy metabolism and related changes in body composition, and micronutrients involved in DNA methylation, for example, folate, vitamins B6 and B12, choline, methionine. Optimal brain function results from highly complex interactions between numerous genetic and environmental factors, including food intake, physical activity, age and stress. Future studies linking nutrition with advances in neuroscience, genomics and epigenomics should provide novel approaches to the prevention of cognitive decline, and treatment of dementia and Alzheimer's disease.
Collapse
Affiliation(s)
- M J Dauncey
- Wolfson College, University of Cambridge, Cambridge, UK
| |
Collapse
|
3158
|
Abstract
Alzheimer's disease (AD) is a clinically heterogeneous neurodegenerative disease with a strong genetic component. Several genes have been associated with AD risk for nearly 20 years. However, it was not until the recent technological advances that allow for the analysis of millions of polymorphisms in thousands of subjects that we have been able to advance our understanding of the genetic complexity of AD susceptibility. Genome-wide association studies and whole-exome and whole-genome sequencing have revealed more than 20 loci associated with AD risk. These studies have provided insights into the molecular pathways that are altered in AD pathogenesis, which have, in turn, provided insight into novel therapeutic targets.
Collapse
Affiliation(s)
- Celeste M Karch
- Department of Psychiatry and Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry and Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alison M Goate
- Department of Psychiatry and Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
3159
|
Bean DM, Heimbach J, Ficorella L, Micklem G, Oliver SG, Favrin G. esyN: network building, sharing and publishing. PLoS One 2014; 9:e106035. [PMID: 25181461 PMCID: PMC4152123 DOI: 10.1371/journal.pone.0106035] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/27/2014] [Indexed: 01/18/2023] Open
Abstract
The construction and analysis of networks is increasingly widespread in biological research. We have developed esyN ("easy networks") as a free and open source tool to facilitate the exchange of biological network models between researchers. esyN acts as a searchable database of user-created networks from any field. We have developed a simple companion web tool that enables users to view and edit networks using data from publicly available databases. Both normal interaction networks (graphs) and Petri nets can be created. In addition to its basic tools, esyN contains a number of logical templates that can be used to create models more easily. The ability to use previously published models as building blocks makes esyN a powerful tool for the construction of models and network graphs. Users are able to save their own projects online and share them either publicly or with a list of collaborators. The latter can be given the ability to edit the network themselves, allowing online collaboration on network construction. esyN is designed to facilitate unrestricted exchange of this increasingly important type of biological information. Ultimately, the aim of esyN is to bring the advantages of Open Source software development to the construction of biological networks.
Collapse
Affiliation(s)
- Daniel M. Bean
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Joshua Heimbach
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Lorenzo Ficorella
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Dipartimento di Biochimica, Universita’ degli studi di Pisa, Pisa, Italy
| | - Gos Micklem
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Stephen G. Oliver
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Giorgio Favrin
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
3160
|
Berchtold NC, Sabbagh MN, Beach TG, Kim RC, Cribbs DH, Cotman CW. Brain gene expression patterns differentiate mild cognitive impairment from normal aged and Alzheimer's disease. Neurobiol Aging 2014; 35:1961-72. [PMID: 24786631 PMCID: PMC4067010 DOI: 10.1016/j.neurobiolaging.2014.03.031] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/25/2014] [Accepted: 03/28/2014] [Indexed: 02/08/2023]
Abstract
Mild cognitive impairment (MCI) represents a cognitive state intermediate between normal aging and early Alzheimer's disease (AD). To investigate if the molecular signature of MCI parallels the clinical picture, we use microarrays to extensively profile gene expression in 4 cortical brain regions (entorhinal cortex, hippocampus, superior frontal gyrus, post-central gyrus) using the postmortem tissue from cognitively normal aged controls, MCI, and AD cases. Our data reveal that gene expression patterns in MCI are not an extension of aging, and for the most part, are not intermediate between aged controls and AD. Functional enrichment analysis of significant genes revealed prominent upregulation in MCI brains of genes associated with anabolic and biosynthetic pathways (notably transcription, protein biosynthesis, protein trafficking, and turnover) as well as mitochondrial energy generation. In addition, many synaptic genes showed altered expression in MCI, predominantly upregulation, including genes for central components of the vesicle fusion machinery at the synapse, synaptic vesicle trafficking, neurotransmitter receptors, and synaptic structure and stabilization. These data suggest that there is a rebalancing of synaptic transmission in the MCI brain. To investigate if synaptic gene expression levels in MCI were related to cognitive function, Pearson correlation coefficient between the Mini Mental State Examination (MMSE) and region-specific messenger RNA expression were computed for MCI cases. A number of synaptic genes showed strong significant correlations (r > 0.8, p < 0.01) most notably in the entorhinal cortex, with fewer in the hippocampus, and very few in neocortical regions. The synaptic genes with highly significant correlations were predominantly related to synaptic transmission and plasticity, and myelin composition. Unexpectedly, we found that gene expression changes that facilitate synaptic excitability and plasticity were overwhelmingly associated with poorer MMSE, and conversely that gene expression changes that inhibit plasticity were positively associated with MMSE. These data suggest that there are excessive excitability and apparent plasticity in limbic brain regions in MCI, that is associated with impaired synaptic and cognitive function. Such changes would be predicted to contribute to increased excitability, in turn leading to greater metabolic demand and ultimately progressive degeneration and AD, if not controlled.
Collapse
Affiliation(s)
- Nicole C Berchtold
- Institute for Mental Impairments and Neurological Disorders (MIND), University of California Irvine, Irvine, CA, USA.
| | | | | | - Ronald C Kim
- Institute for Mental Impairments and Neurological Disorders (MIND), University of California Irvine, Irvine, CA, USA
| | - David H Cribbs
- Institute for Mental Impairments and Neurological Disorders (MIND), University of California Irvine, Irvine, CA, USA; Departments of Neurology and Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Carl W Cotman
- Institute for Mental Impairments and Neurological Disorders (MIND), University of California Irvine, Irvine, CA, USA; Departments of Neurology and Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
3161
|
Jun G, Asai H, Zeldich E, Drapeau E, Chen C, Chung J, Park JH, Kim S, Haroutunian V, Foroud T, Kuwano R, Haines JL, Pericak-Vance MA, Schellenberg GD, Lunetta KL, Kim JW, Buxbaum JD, Mayeux R, Ikezu T, Abraham CR, Farrer LA. PLXNA4 is associated with Alzheimer disease and modulates tau phosphorylation. Ann Neurol 2014; 76:379-92. [PMID: 25043464 PMCID: PMC4830273 DOI: 10.1002/ana.24219] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 07/02/2014] [Accepted: 07/02/2014] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Much of the genetic basis for Alzheimer disease (AD) is unexplained. We sought to identify novel AD loci using a unique family-based approach that can detect robust associations with infrequent variants (minor allele frequency < 0.10). METHODS We conducted a genome-wide association study in the Framingham Heart Study (discovery) and NIA-LOAD (National Institute on Aging-Late-Onset Alzheimer Disease) Study (replication) family-based cohorts using an approach that accounts for family structure and calculates a risk score for AD as the outcome. Links between the most promising gene candidate and AD pathogenesis were explored in silico as well as experimentally in cell-based models and in human brain. RESULTS Genome-wide significant association was identified with a PLXNA4 single nucleotide polymorphism (rs277470) located in a region encoding the semaphorin-3A (SEMA3A) binding domain (meta-analysis p value [meta-P] = 4.1 × 10(-8) ). A test for association with the entire region was also significant (meta-P = 3.2 × 10(-4) ). Transfection of SH-SY5Y cells or primary rat neurons with full-length PLXNA4 (TS1) increased tau phosphorylation with stimulated by SEMA3A. The opposite effect was observed when cells were transfected with shorter isoforms (TS2 and TS3). However, transfection of any isoform into HEK293 cells stably expressing amyloid β (Aβ) precursor protein (APP) did not result in differential effects on APP processing or Aβ production. Late stage AD cases (n = 9) compared to controls (n = 5) had 1.9-fold increased expression of TS1 in cortical brain tissue (p = 1.6 × 10(-4) ). Expression of TS1 was significantly correlated with the Clinical Dementia Rating score (ρ = 0.75, p = 2.2 × 10(-4) ), plaque density (ρ = 0.56, p = 0.01), and Braak stage (ρ = 0.54, p = 0.02). INTERPRETATION Our results indicate that PLXNA4 has a role in AD pathogenesis through isoform-specific effects on tau phosphorylation.
Collapse
Affiliation(s)
- Gyungah Jun
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA,Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, USA,Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA,Corresponding Authors: Drs. Gyungah Jun and Lindsay A. Farrer, Biomedical Genetics E200, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118; tel – (617) 638-5393; fax – (617) 638-4275; or
| | - Hirohide Asai
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ella Zeldich
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Elodie Drapeau
- Department of Psychiatry and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - CiDi Chen
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jaeyoon Chung
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jong-Ho Park
- Department of Health Sciences and Technology, Graduate School, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sehwa Kim
- Department of Health Sciences and Technology, Graduate School, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Vahram Haroutunian
- Department of Psychiatry and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ryozo Kuwano
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Jonathan L. Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Jong-Won Kim
- Department of Health Sciences and Technology, Graduate School, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea,Department of Laboratory Medicine & Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joseph D. Buxbaum
- Department of Psychiatry and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Richard Mayeux
- Department of Neurology and the Taub Institute, Columbia University, New York, New York, USA
| | - Tsuneya Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, USA,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Carmela R. Abraham
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, USA,Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Lindsay A. Farrer
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA,Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, USA,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA,Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA,Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA,Corresponding Authors: Drs. Gyungah Jun and Lindsay A. Farrer, Biomedical Genetics E200, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118; tel – (617) 638-5393; fax – (617) 638-4275; or
| |
Collapse
|
3162
|
Lunnon K, Smith R, Hannon E, De Jager P, Srivastava G, Volta M, Troakes C, Al-Sarraj S, Burrage J, Macdonald R, Condliffe D, Harries LW, Katsel P, Haroutunian V, Kaminsky Z, Joachim C, Powell J, Lovestone S, Bennett DA, Schalkwyk L, Mill J. Methylomic profiling implicates cortical deregulation of ANK1 in Alzheimer's disease. Nat Neurosci 2014; 17:1164-70. [PMID: 25129077 PMCID: PMC4410018 DOI: 10.1038/nn.3782] [Citation(s) in RCA: 424] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 07/07/2014] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder that is characterized by progressive neuropathology and cognitive decline. We performed a cross-tissue analysis of methylomic variation in AD using samples from four independent human post-mortem brain cohorts. We identified a differentially methylated region in the ankyrin 1 (ANK1) gene that was associated with neuropathology in the entorhinal cortex, a primary site of AD manifestation. This region was confirmed as being substantially hypermethylated in two other cortical regions (superior temporal gyrus and prefrontal cortex), but not in the cerebellum, a region largely protected from neurodegeneration in AD, or whole blood obtained pre-mortem from the same individuals. Neuropathology-associated ANK1 hypermethylation was subsequently confirmed in cortical samples from three independent brain cohorts. This study represents, to the best of our knowledge, the first epigenome-wide association study of AD employing a sequential replication design across multiple tissues and highlights the power of this approach for identifying methylomic variation associated with complex disease.
Collapse
Affiliation(s)
- Katie Lunnon
- University of Exeter Medical School, Exeter University, Exeter, UK
| | - Rebecca Smith
- Institute of Psychiatry, King’s College London, London, UK
| | - Eilis Hannon
- University of Exeter Medical School, Exeter University, Exeter, UK
| | - Philip De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, USA
| | - Gyan Srivastava
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women’s Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, USA
| | - Manuela Volta
- Institute of Psychiatry, King’s College London, London, UK
| | - Claire Troakes
- Institute of Psychiatry, King’s College London, London, UK
| | - Safa Al-Sarraj
- Institute of Psychiatry, King’s College London, London, UK
| | - Joe Burrage
- University of Exeter Medical School, Exeter University, Exeter, UK
| | - Ruby Macdonald
- University of Exeter Medical School, Exeter University, Exeter, UK
| | | | - Lorna W. Harries
- University of Exeter Medical School, Exeter University, Exeter, UK
| | - Pavel Katsel
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, USA
| | - Vahram Haroutunian
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neuroscience, The Icahn School of Medicine at Mount Sinai, New York, USA
- JJ Peters VA Medical Center, Bronx, New York, USA
| | - Zachary Kaminsky
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Catharine Joachim
- Department of Neuropathology, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - John Powell
- Institute of Psychiatry, King’s College London, London, UK
| | - Simon Lovestone
- Institute of Psychiatry, King’s College London, London, UK
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | | | - Jonathan Mill
- University of Exeter Medical School, Exeter University, Exeter, UK
- Institute of Psychiatry, King’s College London, London, UK
| |
Collapse
|
3163
|
Cissé M, Checler F. Eph receptors: new players in Alzheimer's disease pathogenesis. Neurobiol Dis 2014; 73:137-49. [PMID: 25193466 DOI: 10.1016/j.nbd.2014.08.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/01/2014] [Accepted: 08/22/2014] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is devastating and leads to permanent losses of memory and other cognitive functions. Although recent genetic evidences strongly argue for a causative role of Aβ in AD onset and progression (Jonsson et al., 2012), its role in AD etiology remains a matter of debate. However, even if not the sole culprit or pathological trigger, genetic and anatomical evidences in conjunction with numerous pharmacological studies, suggest that Aβ peptides, at least contribute to the disease. How Aβ contributes to memory loss remains largely unknown. Soluble Aβ species referred to as Aβ oligomers have been shown to be neurotoxic and induce network failure and cognitive deficits in animal models of the disease. In recent years, several proteins were described as potential Aβ oligomers receptors, amongst which are the receptor tyrosine kinases of Eph family. These receptors together with their natural ligands referred to as ephrins have been involved in a plethora of physiological and pathological processes, including embryonic neurogenesis, learning and memory, diabetes, cancers and anxiety. Here we review recent discoveries on Eph receptors-mediated protection against Aβ oligomers neurotoxicity as well as their potential as therapeutic targets in AD pathogenesis.
Collapse
Affiliation(s)
- Moustapha Cissé
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| |
Collapse
|
3164
|
Lau P, Frigerio CS, De Strooper B. Variance in the identification of microRNAs deregulated in Alzheimer's disease and possible role of lincRNAs in the pathology: the need of larger datasets. Ageing Res Rev 2014; 17:43-53. [PMID: 24607832 DOI: 10.1016/j.arr.2014.02.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 12/19/2022]
Abstract
Non-coding RNAs, such as microRNAs and long non-coding RNAs, represent the next major step in understanding the complexity of gene regulation and expression. In the past decade, tremendous efforts have been put mainly into identifying microRNAs that are changed in Alzheimer's disease, with the goal to provide biomarkers of the disease and to better characterize molecular pathways that are deregulated concomitantly to the formation of Tau and amyloid aggregates. This review underlines the importance of correctly defining, in a deluge of high-throughput data, which microRNAs are abnormally expressed in Alzheimer's disease patients. Despite a clear lack of consensus on the topic, miR-132 is emerging as a neuronal microRNA being gradually down-regulated during disease and showing important roles in the maintenance of brain integrity. Insight into the biological importance of other classes of non-coding RNAs also rapidly increased over the last years and therefore we discuss the possible implication of long non-coding RNAs in Alzheimer's disease.
Collapse
Affiliation(s)
- Pierre Lau
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium; Center for Human Genetics, Leuven Institute for Neurodegenerative Disorders (LIND) and University of Leuven, O&N4, Herestraat 49 Box 602, 3000 Leuven, Belgium
| | - Carlo Sala Frigerio
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium; Center for Human Genetics, Leuven Institute for Neurodegenerative Disorders (LIND) and University of Leuven, O&N4, Herestraat 49 Box 602, 3000 Leuven, Belgium
| | - Bart De Strooper
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium; Center for Human Genetics, Leuven Institute for Neurodegenerative Disorders (LIND) and University of Leuven, O&N4, Herestraat 49 Box 602, 3000 Leuven, Belgium; Department of Molecular Neuroscience, University College London (UCL) Institute of Neurology, London, United Kingdom.
| |
Collapse
|
3165
|
Abstract
Two independent epigenome-wide association studies of Alzheimer’s disease cohorts have identified overlapping methylation signals in four loci, ANK1, RPL13, RHBDF2 and CDH23, not previously associated with Alzheimer’s disease. These studies also suggest that epigenetic changes contribute more to Alzheimer’s disease than expected.
Collapse
Affiliation(s)
- Jenny Lord
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carlos Cruchaga
- 1] Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA. [2] Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
3166
|
De Jager PL, Srivastava G, Lunnon K, Burgess J, Schalkwyk LC, Yu L, Eaton ML, Keenan BT, Ernst J, McCabe C, Tang A, Raj T, Replogle J, Brodeur W, Gabriel S, Chai HS, Younkin C, Younkin SG, Zou F, Szyf M, Epstein CB, Schneider JA, Bernstein BE, Meissner A, Ertekin-Taner N, Chibnik LB, Kellis M, Mill J, Bennett DA. Alzheimer's disease: early alterations in brain DNA methylation at ANK1, BIN1, RHBDF2 and other loci. Nat Neurosci 2014; 17:1156-63. [PMID: 25129075 PMCID: PMC4292795 DOI: 10.1038/nn.3786] [Citation(s) in RCA: 696] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/16/2014] [Indexed: 02/07/2023]
Abstract
We used a collection of 708 prospectively collected autopsied brains to assess the methylation state of the brain's DNA in relation to Alzheimer's disease (AD). We found that the level of methylation at 71 of the 415,848 interrogated CpGs was significantly associated with the burden of AD pathology, including CpGs in the ABCA7 and BIN1 regions, which harbor known AD susceptibility variants. We validated 11 of the differentially methylated regions in an independent set of 117 subjects. Furthermore, we functionally validated these CpG associations and identified the nearby genes whose RNA expression was altered in AD: ANK1, CDH23, DIP2A, RHBDF2, RPL13, SERPINF1 and SERPINF2. Our analyses suggest that these DNA methylation changes may have a role in the onset of AD given that we observed them in presymptomatic subjects and that six of the validated genes connect to a known AD susceptibility gene network.
Collapse
Affiliation(s)
- PL De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, NRB168, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
- Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - G Srivastava
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, NRB168, Boston, MA 02115
- Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - K Lunnon
- University of Exeter Medical School, University of Exeter, RILD (Level 4), Barrack Rd, Exeter, UK
- Institute of Psychiatry, King's College London, De Crespigny Park, Denmark Hill, London. SE5 8AF. UK
| | - J Burgess
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224
| | - LC Schalkwyk
- University of Exeter Medical School, University of Exeter, RILD (Level 4), Barrack Rd, Exeter, UK
- Institute of Psychiatry, King's College London, De Crespigny Park, Denmark Hill, London. SE5 8AF. UK
| | - L Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600 S Paulina St., Chicago, IL 60612; Computer Science and Artificial Intelligence Laboratory (CSAIL), Massachusetts Institute of Technology, 32 Vassar St., Cambridge, MA 02139
| | - ML Eaton
- Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
- Genetic Analysis Platform, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - BT Keenan
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, NRB168, Boston, MA 02115
- Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - J Ernst
- Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
- Genetic Analysis Platform, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - C McCabe
- Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - A Tang
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, NRB168, Boston, MA 02115
| | - T Raj
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, NRB168, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
- Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - J Replogle
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, NRB168, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
- Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - W Brodeur
- Genetic Analysis Platform, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - S Gabriel
- Genetic Analysis Platform, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - HS Chai
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224
| | - C Younkin
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | - SG Younkin
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | - F Zou
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | - M Szyf
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Québec, H3G 1Y6
| | - CB Epstein
- Epigenomics Program, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - JA Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600 S Paulina St., Chicago, IL 60612; Computer Science and Artificial Intelligence Laboratory (CSAIL), Massachusetts Institute of Technology, 32 Vassar St., Cambridge, MA 02139
| | - BE Bernstein
- Harvard Medical School, Boston, MA 02115
- Epigenomics Program, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
- Department of Pathology, Massachusetts General Hospital, 185 Cambridge St., Boston, MA 02114
| | - A Meissner
- Genetic Analysis Platform, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
- Harvard Stem Cell Institute, Harvard University, 1350 Massachusetts Ave., Cambridge MA 02138
| | - N Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224
| | - LB Chibnik
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, NRB168, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
- Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - M Kellis
- Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
- Genetic Analysis Platform, Broad Institute, 7 Cambridge Center, Cambridge, MA 02142
| | - J Mill
- University of Exeter Medical School, University of Exeter, RILD (Level 4), Barrack Rd, Exeter, UK
- Institute of Psychiatry, King's College London, De Crespigny Park, Denmark Hill, London. SE5 8AF. UK
| | - DA Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600 S Paulina St., Chicago, IL 60612; Computer Science and Artificial Intelligence Laboratory (CSAIL), Massachusetts Institute of Technology, 32 Vassar St., Cambridge, MA 02139
| |
Collapse
|
3167
|
Wes PD, Easton A, Corradi J, Barten DM, Devidze N, DeCarr LB, Truong A, He A, Barrezueta NX, Polson C, Bourin C, Flynn ME, Keenan S, Lidge R, Meredith J, Natale J, Sankaranarayanan S, Cadelina GW, Albright CF, Cacace AM. Tau overexpression impacts a neuroinflammation gene expression network perturbed in Alzheimer's disease. PLoS One 2014; 9:e106050. [PMID: 25153994 PMCID: PMC4143352 DOI: 10.1371/journal.pone.0106050] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 07/30/2014] [Indexed: 01/16/2023] Open
Abstract
Filamentous inclusions of the microtubule-associated protein, tau, define a variety of neurodegenerative diseases known as tauopathies, including Alzheimer's disease (AD). To better understand the role of tau-mediated effects on pathophysiology and global central nervous system function, we extensively characterized gene expression, pathology and behavior of the rTg4510 mouse model, which overexpresses a mutant form of human tau that causes Frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). We found that the most predominantly altered gene expression pathways in rTg4510 mice were in inflammatory processes. These results closely matched the causal immune function and microglial gene-regulatory network recently identified in AD. We identified additional gene expression changes by laser microdissecting specific regions of the hippocampus, which highlighted alterations in neuronal network activity. Expression of inflammatory genes and markers of neuronal activity changed as a function of age in rTg4510 mice and coincided with behavioral deficits. Inflammatory changes were tau-dependent, as they were reversed by suppression of the tau transgene. Our results suggest that the alterations in microglial phenotypes that appear to contribute to the pathogenesis of Alzheimer's disease may be driven by tau dysfunction, in addition to the direct effects of beta-amyloid.
Collapse
Affiliation(s)
- Paul D. Wes
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Amy Easton
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - John Corradi
- Department of Applied Genomics, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Donna M. Barten
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Nino Devidze
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Lynn B. DeCarr
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Amy Truong
- Department of Applied Genomics, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Aiqing He
- Department of Applied Genomics, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Nestor X. Barrezueta
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Craig Polson
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Clotilde Bourin
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Marianne E. Flynn
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Stefanie Keenan
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Regina Lidge
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Jere Meredith
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Joanne Natale
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Sethu Sankaranarayanan
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Greg W. Cadelina
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Charlie F. Albright
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
| | - Angela M. Cacace
- Department of Neuroscience, Bristol-Myers Squibb, Wallingford, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
3168
|
Fuller JP, Stavenhagen JB, Teeling JL. New roles for Fc receptors in neurodegeneration-the impact on Immunotherapy for Alzheimer's Disease. Front Neurosci 2014; 8:235. [PMID: 25191216 PMCID: PMC4139653 DOI: 10.3389/fnins.2014.00235] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/17/2014] [Indexed: 12/11/2022] Open
Abstract
There are an estimated 18 million Alzheimer's disease (AD) sufferers worldwide and with no disease modifying treatment currently available, development of new therapies represents an enormous unmet clinical need. AD is characterized by episodic memory loss followed by severe cognitive decline and is associated with many neuropathological changes. AD is characterized by deposits of amyloid beta (Aβ), neurofibrillary tangles, and neuroinflammation. Active immunization or passive immunization against Aβ leads to the clearance of deposits in transgenic mice expressing human Aβ. This clearance is associated with reversal of associated cognitive deficits, but these results have not translated to humans, with both active and passive immunotherapy failing to improve memory loss. One explanation for these observations is that certain anti-Aβ antibodies mediate damage to the cerebral vasculature limiting the top dose and potentially reducing efficacy. Fc gamma receptors (FcγR) are a family of immunoglobulin-like receptors which bind to the Fc portion of IgG, and mediate the response of effector cells to immune complexes. Data from both mouse and human studies suggest that cross-linking FcγR by therapeutic antibodies and the subsequent pro-inflammatory response mediates the vascular side effects seen following immunotherapy. Increasing evidence is emerging that FcγR expression on CNS resident cells, including microglia and neurons, is increased during aging and functionally involved in the pathogenesis of age-related neurodegenerative diseases. Therefore, we propose that increased expression and ligation of FcγR in the CNS, either by endogenous IgG or therapeutic antibodies, has the potential to induce vascular damage and exacerbate neurodegeneration. To produce safe and effective immunotherapies for AD and other neurodegenerative diseases it will be vital to understand the role of FcγR in the healthy and diseased brain. Here we review the literature on FcγR expression, function and proposed roles in multiple age-related neurological diseases. Lessons can be learnt from therapeutic antibodies used for the treatment of cancer where antibodies have been engineered for optimal efficacy.
Collapse
Affiliation(s)
- James P. Fuller
- CNS Inflammation Group, Centre for Biological Sciences, University of SouthamptonSouthampton, UK
| | | | - Jessica L. Teeling
- CNS Inflammation Group, Centre for Biological Sciences, University of SouthamptonSouthampton, UK
| |
Collapse
|
3169
|
Abstract
The formation of amyloid aggregates is a feature of most, if not all, polypeptide chains. In vivo modelling of this process has been undertaken in the fruitfly Drosophila melanogaster with remarkable success. Models of both neurological and systemic amyloid diseases have been generated and have informed our understanding of disease pathogenesis in two main ways. First, the toxic amyloid species have been at least partially characterized, for example in the case of the Aβ (amyloid β-peptide) associated with Alzheimer's disease. Secondly, the genetic underpinning of model disease-linked phenotypes has been characterized for a number of neurodegenerative disorders. The current challenge is to integrate our understanding of disease-linked processes in the fly with our growing knowledge of human disease, for the benefit of patients.
Collapse
|
3170
|
Zhao W, Zhang J, Davis EG, Rebeck GW. Aging reduces glial uptake and promotes extracellular accumulation of Aβ from a lentiviral vector. Front Aging Neurosci 2014; 6:210. [PMID: 25177293 PMCID: PMC4133689 DOI: 10.3389/fnagi.2014.00210] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/28/2014] [Indexed: 01/27/2023] Open
Abstract
We used a lentiviral system for expressing secreted human Aβ in the brains of young and old APOE knock-in mice. This system allowed us to examine Aβ metabolism in vivo, and test the effects of both aging and APOE genotype, two of the strongest risk factors for Alzheimer’s disease. We injected the Aβ1-42 lentivirus into the motor cortex of young (2 month old) and old (20–22 month old) APOE3 and APOE4 mice. After 2 weeks of lentiviral expression, we analyzed the pattern of Aβ accumulation, glial activation, and phosphor-tau. In young mice, Aβ accumulated mainly within neurons with no evidence of extracellular Aβ. Significantly higher levels of intraneuronal Aβ were observed in APOE4 mice compared to APOE3 mice. In old mice, APOE4 predisposed again to higher levels of Aβ accumulation, but the Aβ was mainly in extracellular spaces. In younger mice, we also observed Aβ in microglia but not astrocytes. The numbers of microglia containing Aβ were significantly higher in APOE3 mice compared to APOE4 mice, and were significantly lower in both genetic backgrounds with aging. The astrocytes in old mice were activated to a greater extent in the brain regions where Aβ was introduced, an effect that was again increased by the presence of APOE4. Finally, phospho-tau accumulated in the region of Aβ expression, with evidence of extracellular phospho-tau increasing with aging. These data suggest that APOE4 predisposes to less microglial clearance of Aβ, leading to more intraneuronal accumulation. In older brains, decreased clearance leads to more extracellular Aβ, and more downstream consequences relating to astrocyte activation and phospho-tau accumulation. We conclude that both aging and APOE genotype affect pathways related to Aβ metabolism by microglia.
Collapse
Affiliation(s)
- Wenjuan Zhao
- School of Pharmacy, Shanghai Jiao Tong University Shanghai, China
| | - Jiguo Zhang
- Department of Pharmacology, School of Pharmacy, Taishan Medical University Taian, China
| | - Elizabeth G Davis
- Department of Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - G William Rebeck
- Department of Neuroscience, Georgetown University Medical Center Washington, DC, USA
| |
Collapse
|
3171
|
Han G, Sun J, Wang J, Bai Z, Song F, Lei H. Genomics in neurological disorders. GENOMICS PROTEOMICS & BIOINFORMATICS 2014; 12:156-63. [PMID: 25108264 PMCID: PMC4411357 DOI: 10.1016/j.gpb.2014.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 07/07/2014] [Accepted: 07/07/2014] [Indexed: 11/26/2022]
Abstract
Neurological disorders comprise a variety of complex diseases in the central nervous system, which can be roughly classified as neurodegenerative diseases and psychiatric disorders. The basic and translational research of neurological disorders has been hindered by the difficulty in accessing the pathological center (i.e., the brain) in live patients. The rapid advancement of sequencing and array technologies has made it possible to investigate the disease mechanism and biomarkers from a systems perspective. In this review, recent progresses in the discovery of novel risk genes, treatment targets and peripheral biomarkers employing genomic technologies will be discussed. Our major focus will be on two of the most heavily investigated neurological disorders, namely Alzheimer’s disease and autism spectrum disorder.
Collapse
Affiliation(s)
- Guangchun Han
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiya Sun
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiajia Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhouxian Bai
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fuhai Song
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongxing Lei
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing 100053, China.
| |
Collapse
|
3172
|
Carrasquillo MM, Crook JE, Pedraza O, Thomas CS, Pankratz VS, Allen M, Nguyen T, Malphrus KG, Ma L, Bisceglio GD, Roberts RO, Lucas JA, Smith GE, Ivnik RJ, Machulda MM, Graff-Radford NR, Petersen RC, Younkin SG, Ertekin-Taner N. Late-onset Alzheimer's risk variants in memory decline, incident mild cognitive impairment, and Alzheimer's disease. Neurobiol Aging 2014; 36:60-7. [PMID: 25189118 DOI: 10.1016/j.neurobiolaging.2014.07.042] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/04/2014] [Accepted: 07/28/2014] [Indexed: 11/19/2022]
Abstract
We tested association of nine late-onset Alzheimer's disease (LOAD) risk variants from genome-wide association studies (GWAS) with memory and progression to mild cognitive impairment (MCI) or LOAD (MCI/LOAD) in older Caucasians, cognitively normal at baseline and longitudinally evaluated at Mayo Clinic Rochester and Jacksonville (n>2000). Each variant was tested both individually and collectively using a weighted risk score. APOE-e4 associated with worse baseline memory and increased decline with highly significant overall effect on memory. CLU-rs11136000-G associated with worse baseline memory and incident MCI/LOAD. MS4A6A-rs610932-C associated with increased incident MCI/LOAD and suggestively with lower baseline memory. ABCA7-rs3764650-C and EPHA1-rs11767557-A associated with increased rates of memory decline in subjects with a final diagnosis of MCI/LOAD. PICALM-rs3851179-G had an unexpected protective effect on incident MCI/LOAD. Only APOE-inclusive risk scores associated with worse memory and incident MCI/LOAD. The collective influence of the nine top LOAD GWAS variants on memory decline and progression to MCI/LOAD appears limited. Discovery of biologically functional variants at these loci may uncover stronger effects on memory and incident disease.
Collapse
Affiliation(s)
| | - Julia E Crook
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Otto Pedraza
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, USA
| | - Colleen S Thomas
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - V Shane Pankratz
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Thuy Nguyen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Li Ma
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Rosebud O Roberts
- Department of Health Sciences Research, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - John A Lucas
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, USA
| | - Glenn E Smith
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Robert J Ivnik
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of Neurology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
3173
|
Ghani M, Lang AE, Zinman L, Nacmias B, Sorbi S, Bessi V, Tedde A, Tartaglia MC, Surace EI, Sato C, Moreno D, Xi Z, Hung R, Nalls MA, Singleton A, St George-Hyslop P, Rogaeva E. Mutation analysis of patients with neurodegenerative disorders using NeuroX array. Neurobiol Aging 2014; 36:545.e9-14. [PMID: 25174650 DOI: 10.1016/j.neurobiolaging.2014.07.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 07/27/2014] [Indexed: 12/13/2022]
Abstract
Genetic analyses of patients with neurodegenerative disorders have identified multiple genes that need to be investigated for the presence of damaging variants. However, mutation analysis by Sanger sequencing is costly and time consuming. We tested the utility of a recently designed semi-custom genome-wide array (NeuroX; Illumina, Inc) tailored to study neurodegenerative diseases (e.g., mutation screening). We investigated 192 patients with 4 different neurodegenerative disorders for the presence of rare damaging variations in 77 genes implicated in these diseases. Several causative mutations were identified and confirmed by Sanger sequencing, including PSEN1 p.M233T responsible for Alzheimer's disease in a large Italian family, as well as SOD1 p.A4V and p.I113T in patients with amyotrophic lateral sclerosis. In total, we identified 78 potentially damaging rare variants (frequency <1%), including ABCA7 p.L400V in a family with Alzheimer's disease and LRRK2 p.R1514Q in 6 of 98 patients with Parkinson's disease (6.1%). In conclusion, NeuroX appears to be helpful for rapid and accurate mutation screening, although further development may be still required to improve some current caveats.
Collapse
Affiliation(s)
- Mahdi Ghani
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Anthony E Lang
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada; Morton and Gloria Shulman Movement Disorders Center and the Edmond J. Safra Program in Parkinson's Disease, Toronto Western Hospital, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Lorne Zinman
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Andrea Tedde
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ezequiel I Surace
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Laboratorio de Biología Molecular, Instituto de Investigaciones Neurológicas Dr. Raúl Carrea (FLENI), Buenos Aires, Argentina
| | - Christine Sato
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Danielle Moreno
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Zhengrui Xi
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Hung
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Peter St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Clinical Neurosciences, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
3174
|
Khabirova E, Moloney A, Marciniak SJ, Williams J, Lomas DA, Oliver SG, Favrin G, Sattelle DB, Crowther DC. The TRiC/CCT chaperone is implicated in Alzheimer's disease based on patient GWAS and an RNAi screen in Aβ-expressing Caenorhabditis elegans. PLoS One 2014; 9:e102985. [PMID: 25080104 PMCID: PMC4117641 DOI: 10.1371/journal.pone.0102985] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 06/25/2014] [Indexed: 01/02/2023] Open
Abstract
The human Aβ peptide causes progressive paralysis when expressed in the muscles of the nematode worm, C. elegans. We have exploited this model of Aβ toxicity by carrying out an RNAi screen to identify genes whose reduced expression modifies the severity of this locomotor phenotype. Our initial finding was that none of the human orthologues of these worm genes is identical with the genome-wide significant GWAS genes reported to date (the “white zone”); moreover there was no identity between worm screen hits and the longer list of GWAS genes which included those with borderline levels of significance (the “grey zone”). This indicates that Aβ toxicity should not be considered as equivalent to sporadic AD. To increase the sensitivity of our analysis, we then considered the physical interactors (+1 interactome) of the products of the genes in both the worm and the white+grey zone lists. When we consider these worm and GWAS gene lists we find that 4 of the 60 worm genes have a +1 interactome overlap that is larger than expected by chance. Two of these genes form a chaperonin complex, the third is closely associated with this complex and the fourth gene codes for actin, the major substrate of the same chaperonin.
Collapse
Affiliation(s)
- Eleonora Khabirova
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Aileen Moloney
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Stefan J. Marciniak
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Julie Williams
- Department of Psychiatry and Neurology, MRC Centre for Neuropsychiatric Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - David A. Lomas
- Department of Medicine, University College London, London, United Kingdom
| | - Stephen G. Oliver
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- * E-mail: (SGO); (DCC)
| | - Giorgio Favrin
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - David B. Sattelle
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- The Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Damian C. Crowther
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- * E-mail: (SGO); (DCC)
| |
Collapse
|
3175
|
Moshkanbaryans L, Chan LS, Graham ME. The Biochemical Properties and Functions of CALM and AP180 in Clathrin Mediated Endocytosis. MEMBRANES 2014; 4:388-413. [PMID: 25090048 PMCID: PMC4194041 DOI: 10.3390/membranes4030388] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/03/2014] [Accepted: 07/22/2014] [Indexed: 01/26/2023]
Abstract
Clathrin-mediated endocytosis (CME) is a fundamental process for the regulated internalization of transmembrane cargo and ligands via the formation of vesicles using a clathrin coat. A vesicle coat is initially created at the plasma membrane by clathrin assembly into a lattice, while a specific cargo sorting process selects and concentrates proteins for inclusion in the new vesicle. Vesicles formed via CME traffic to different parts of the cell and fuse with target membranes to deliver cargo. Both clathrin assembly and cargo sorting functions are features of the two gene family consisting of assembly protein 180 kDa (AP180) and clathrin assembly lymphoid myeloid leukemia protein (CALM). In this review, we compare the primary structure and domain organization of CALM and AP180 and relate these properties to known functions and roles in CME and disease.
Collapse
Affiliation(s)
- Lia Moshkanbaryans
- Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW 2145, Australia.
| | - Ling-Shan Chan
- Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW 2145, Australia.
| | - Mark E Graham
- Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW 2145, Australia.
| |
Collapse
|
3176
|
Mitsui J, Tsuji S. Genomic aspects of sporadic neurodegenerative diseases. Biochem Biophys Res Commun 2014; 452:221-5. [PMID: 25078619 DOI: 10.1016/j.bbrc.2014.07.098] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/22/2014] [Indexed: 12/31/2022]
Abstract
Sporadic neurodegenerative diseases are complex in nature, that is, they involve multiple genetic and environmental factors that may play roles at the molecular level. In contrast to diseases with Mendelian inheritance, the genomic signatures of common sporadic forms of neurodegenerative diseases largely remain unknown. Over the past decade, genome-wide association studies employing common single-nucleotide polymorphisms have been intensively conducted, in which the theoretical framework is based on the "common disease-common variants" hypothesis. Another paradigm is a sequence-based association study under the "common disease-multiple rare variants" hypothesis. Because current next-generation sequencing technologies enable us to obtain virtually all the variants in human genome irrespective of allele frequencies, it is anticipated that sequence-based association studies will become the mainstream approach. In this review, we present brief overviews of molecular genetic approaches to elucidate the molecular bases of sporadic forms of neurodegenerative diseases, including Alzheimer disease, Parkinson disease, and multiple system atrophy as examples.
Collapse
Affiliation(s)
- Jun Mitsui
- Department of Neurology, The University of Tokyo, Graduate School of Medicine, Japan
| | - Shoji Tsuji
- Department of Neurology, The University of Tokyo, Graduate School of Medicine, Japan.
| |
Collapse
|
3177
|
Adaptors for disorders of the brain? The cancer signaling proteins NEDD9, CASS4, and PTK2B in Alzheimer's disease. Oncoscience 2014; 1:486-503. [PMID: 25594051 PMCID: PMC4278314 DOI: 10.18632/oncoscience.64] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/23/2014] [Indexed: 12/19/2022] Open
Abstract
No treatment strategies effectively limit the progression of Alzheimer's disease (AD), a common and debilitating neurodegenerative disorder. The absence of viable treatment options reflects the fact that the pathophysiology and genotypic causes of the disease are not well understood. The advent of genome-wide association studies (GWAS) has made it possible to broadly investigate genotypic alterations driving phenotypic occurrences. Recent studies have associated single nucleotide polymorphisms (SNPs) in two paralogous scaffolding proteins, NEDD9 and CASS4, and the kinase PTK2B, with susceptibility to late-onset AD (LOAD). Intriguingly, NEDD9, CASS4, and PTK2B have been much studied as interacting partners regulating oncogenesis and metastasis, and all three are known to be active in the brain during development and in cancer. However, to date, the majority of studies of these proteins have emphasized their roles in the directly cancer relevant processes of migration and survival signaling. We here discuss evidence for roles of NEDD9, CASS4 and PTK2B in additional processes, including hypoxia, vascular changes, inflammation, microtubule stabilization and calcium signaling, as potentially relevant to the pathogenesis of LOAD. Reciprocally, these functions can better inform our understanding of the action of NEDD9, CASS4 and PTK2B in cancer.
Collapse
|
3178
|
Affiliation(s)
- J M Schott
- Dementia Research Centre, UCL Institute of Neurology, London, UK.
| |
Collapse
|
3179
|
The impact of the human genome project on complex disease. Genes (Basel) 2014; 5:518-35. [PMID: 25032678 PMCID: PMC4198915 DOI: 10.3390/genes5030518] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/03/2014] [Accepted: 06/24/2014] [Indexed: 02/06/2023] Open
Abstract
In the decade that has passed since the initial release of the Human Genome, numerous advancements in science and technology within and beyond genetics and genomics have been encouraged and enhanced by the availability of this vast and remarkable data resource. Progress in understanding three common, complex diseases: age-related macular degeneration (AMD), Alzheimer's disease (AD), and multiple sclerosis (MS), are three exemplars of the incredible impact on the elucidation of the genetic architecture of disease. The approaches used in these diseases have been successfully applied to numerous other complex diseases. For example, the heritability of AMD was confirmed upon the release of the first genome-wide association study (GWAS) along with confirmatory reports that supported the findings of that state-of-the art method, thus setting the foundation for future GWAS in other heritable diseases. Following this seminal discovery and applying it to other diseases including AD and MS, the genetic knowledge of AD expanded far beyond the well-known APOE locus and now includes more than 20 loci. MS genetics saw a similar increase beyond the HLA loci and now has more than 100 known risk loci. Ongoing and future efforts will seek to define the remaining heritability of these diseases; the next decade could very well hold the key to attaining this goal.
Collapse
|
3180
|
Okamoto SI, Nakamura T, Cieplak P, Chan SF, Kalashnikova E, Liao L, Saleem S, Han X, Clemente A, Nutter A, Sances S, Brechtel C, Haus D, Haun F, Sanz-Blasco S, Huang X, Li H, Zaremba JD, Cui J, Gu Z, Nikzad R, Harrop A, McKercher SR, Godzik A, Yates JR, Lipton SA. S-nitrosylation-mediated redox transcriptional switch modulates neurogenesis and neuronal cell death. Cell Rep 2014; 8:217-28. [PMID: 25001280 DOI: 10.1016/j.celrep.2014.06.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 04/17/2014] [Accepted: 06/03/2014] [Indexed: 11/24/2022] Open
Abstract
Redox-mediated posttranslational modifications represent a molecular switch that controls major mechanisms of cell function. Nitric oxide (NO) can mediate redox reactions via S-nitrosylation, representing transfer of an NO group to a critical protein thiol. NO is known to modulate neurogenesis and neuronal survival in various brain regions in disparate neurodegenerative conditions. However, a unifying molecular mechanism linking these phenomena remains unknown. Here, we report that S-nitrosylation of myocyte enhancer factor 2 (MEF2) transcription factors acts as a redox switch to inhibit both neurogenesis and neuronal survival. Structure-based analysis reveals that MEF2 dimerization creates a pocket, facilitating S-nitrosylation at an evolutionally conserved cysteine residue in the DNA binding domain. S-Nitrosylation disrupts MEF2-DNA binding and transcriptional activity, leading to impaired neurogenesis and survival in vitro and in vivo. Our data define a molecular switch whereby redox-mediated posttranslational modification controls both neurogenesis and neurodegeneration via a single transcriptional signaling cascade.
Collapse
Affiliation(s)
- Shu-Ichi Okamoto
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Tomohiro Nakamura
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Piotr Cieplak
- Bioinformatics and Systems Biology Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Shing Fai Chan
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Evgenia Kalashnikova
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Lujian Liao
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sofiyan Saleem
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Xuemei Han
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Arjay Clemente
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Anthony Nutter
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sam Sances
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Christopher Brechtel
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Daniel Haus
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Florian Haun
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sara Sanz-Blasco
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Xiayu Huang
- Bioinformatics and Systems Biology Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hao Li
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jeffrey D Zaremba
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jiankun Cui
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Zezong Gu
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Rana Nikzad
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Anne Harrop
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Scott R McKercher
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Adam Godzik
- Bioinformatics and Systems Biology Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Stuart A Lipton
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
3181
|
Guerchet M, Mbelesso P, Ndamba-Bandzouzi B, Pilleron S, Desormais I, Lacroix P, Aboyans V, Jésus P, Desport JC, Tchalla AE, Marin B, Lambert JC, Clément JP, Dartigues JF, Preux PM. Epidemiology of dementia in Central Africa (EPIDEMCA): protocol for a multicentre population-based study in rural and urban areas of the Central African Republic and the Republic of Congo. SPRINGERPLUS 2014; 3:338. [PMID: 25045613 PMCID: PMC4096859 DOI: 10.1186/2193-1801-3-338] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 06/10/2014] [Indexed: 12/18/2022]
Abstract
Background The worldwide population is ageing and the proportion of elderly aged 60 and over is expected to dramatically rise in Low and Middle Income Countries (LMIC). The epidemic of dementia will not spare those countries, where the largest increases in numbers of people affected are estimated. Besides, dementia is still understudied in sub-Saharan Africa (SSA) compared to other regions. This paper describes the protocol for the ‘Epidemiology of Dementia in Central Africa’ population-based study, which aims at estimating the prevalence of dementia in two countries of Central Africa and investigating possible risk factors. Methods/Design A multicenter population-based study was carried out in Central African Republic and Republic of Congo between 2011 and 2012 including both urban and rural sites in each country. Around 2000 participants aged ≥65 years old were interviewed in total using the Community Screening Interview for Dementia (CSI-D), the GMS-AGECAT and the CERAD’s 10-word list. Elderly with low performance to the cognitive part of the CSI-D (COGSCORE ≤ 24.5) were then clinically assessed by neurologists and underwent further psychometrical tests. DSM-IV and NINCDS-ADRDA criteria were required for dementia and Alzheimer’s disease (AD) diagnoses respectively. The algorithmic 10/66 dementia diagnosis was also determined. Petersen’s criteria were required for the diagnosis of Mild Cognitive Impairment. Sociodemographic, and environmental factors including vascular, nutritional, biological, psychosocial and lifestyle factors were collected in each setting in order to investigate factors associated with dementia. Blood sampling was realized to investigate genetic variations that could modify the risk of dementia. Discussion For now, no large epidemiological study has been undertaken to compare the prevalence of dementia in both rural and urban areas within SSA countries. This programme will provide further evidence regarding the prevalence of dementia in SSA, and also the possible rural/urban disparities existing with associated factors. Furthermore, the genetics of AD in those populations will be addressed.
Collapse
Affiliation(s)
- Maëlenn Guerchet
- Tropical Neuroepidemiology, Faculty of Medicine, INSERM UMR 1094, 2 rue du Docteur Marcland, 87025 Limoges, France ; Institute of Neuroepidemiology and Tropical Neurology, University Limoges, School of Medicine, CNRS FR 3503 GEIST, Limoges, France ; King's College London, Centre for Global Mental Health, Institute of Psychiatry, Health Service and Population Research Department, London, UK
| | - Pascal Mbelesso
- Tropical Neuroepidemiology, Faculty of Medicine, INSERM UMR 1094, 2 rue du Docteur Marcland, 87025 Limoges, France ; Institute of Neuroepidemiology and Tropical Neurology, University Limoges, School of Medicine, CNRS FR 3503 GEIST, Limoges, France ; Department of Neurology, Brazzaville University Hospital, Brazzaville, Republic of Congo
| | | | - Sophie Pilleron
- Tropical Neuroepidemiology, Faculty of Medicine, INSERM UMR 1094, 2 rue du Docteur Marcland, 87025 Limoges, France ; Institute of Neuroepidemiology and Tropical Neurology, University Limoges, School of Medicine, CNRS FR 3503 GEIST, Limoges, France
| | - Ileana Desormais
- Tropical Neuroepidemiology, Faculty of Medicine, INSERM UMR 1094, 2 rue du Docteur Marcland, 87025 Limoges, France ; Institute of Neuroepidemiology and Tropical Neurology, University Limoges, School of Medicine, CNRS FR 3503 GEIST, Limoges, France ; Department of CardioVascular Surgery, CHU, Limoges, France
| | - Philippe Lacroix
- Tropical Neuroepidemiology, Faculty of Medicine, INSERM UMR 1094, 2 rue du Docteur Marcland, 87025 Limoges, France ; Institute of Neuroepidemiology and Tropical Neurology, University Limoges, School of Medicine, CNRS FR 3503 GEIST, Limoges, France ; Department of CardioVascular Surgery, CHU, Limoges, France
| | - Victor Aboyans
- Tropical Neuroepidemiology, Faculty of Medicine, INSERM UMR 1094, 2 rue du Docteur Marcland, 87025 Limoges, France ; Institute of Neuroepidemiology and Tropical Neurology, University Limoges, School of Medicine, CNRS FR 3503 GEIST, Limoges, France ; Department of Cardiology, Dupuytren University Hospital, Limoges, France
| | - Pierre Jésus
- Tropical Neuroepidemiology, Faculty of Medicine, INSERM UMR 1094, 2 rue du Docteur Marcland, 87025 Limoges, France ; Institute of Neuroepidemiology and Tropical Neurology, University Limoges, School of Medicine, CNRS FR 3503 GEIST, Limoges, France ; Department of Nutrition, CHU, Limoges, France
| | - Jean-Claude Desport
- Tropical Neuroepidemiology, Faculty of Medicine, INSERM UMR 1094, 2 rue du Docteur Marcland, 87025 Limoges, France ; Institute of Neuroepidemiology and Tropical Neurology, University Limoges, School of Medicine, CNRS FR 3503 GEIST, Limoges, France ; Department of Nutrition, CHU, Limoges, France
| | - Achille E Tchalla
- EA 6310, Disability, Activity, Aging, Autonomy and the Environment (HAVAE), Limoges, France
| | - Benoît Marin
- Tropical Neuroepidemiology, Faculty of Medicine, INSERM UMR 1094, 2 rue du Docteur Marcland, 87025 Limoges, France ; Institute of Neuroepidemiology and Tropical Neurology, University Limoges, School of Medicine, CNRS FR 3503 GEIST, Limoges, France ; CHU, Department of Medical Information & Evaluation, Clinical Research and Biostatistic Unit, Limoges, France
| | | | - Jean-Pierre Clément
- Tropical Neuroepidemiology, Faculty of Medicine, INSERM UMR 1094, 2 rue du Docteur Marcland, 87025 Limoges, France ; Institute of Neuroepidemiology and Tropical Neurology, University Limoges, School of Medicine, CNRS FR 3503 GEIST, Limoges, France ; Hospital and University Federation of Adult and Geriatric Psychiatry, Limoges, France
| | | | - Pierre-Marie Preux
- Tropical Neuroepidemiology, Faculty of Medicine, INSERM UMR 1094, 2 rue du Docteur Marcland, 87025 Limoges, France ; Institute of Neuroepidemiology and Tropical Neurology, University Limoges, School of Medicine, CNRS FR 3503 GEIST, Limoges, France ; CHU, Department of Medical Information & Evaluation, Clinical Research and Biostatistic Unit, Limoges, France
| | | |
Collapse
|
3182
|
Allen M, Kachadoorian M, Quicksall Z, Zou F, Chai HS, Younkin C, Crook JE, Pankratz VS, Carrasquillo MM, Krishnan S, Nguyen T, Ma L, Malphrus K, Lincoln S, Bisceglio G, Kolbert CP, Jen J, Mukherjee S, Kauwe JK, Crane PK, Haines JL, Mayeux R, Pericak-Vance MA, Farrer LA, Schellenberg GD, Parisi JE, Petersen RC, Graff-Radford NR, Dickson DW, Younkin SG, Ertekin-Taner N. Association of MAPT haplotypes with Alzheimer's disease risk and MAPT brain gene expression levels. ALZHEIMERS RESEARCH & THERAPY 2014; 6:39. [PMID: 25324900 PMCID: PMC4198935 DOI: 10.1186/alzrt268] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/28/2014] [Indexed: 01/01/2023]
Abstract
Introduction MAPT encodes for tau, the predominant component of neurofibrillary tangles that are neuropathological hallmarks of Alzheimer’s disease (AD). Genetic association of MAPT variants with late-onset AD (LOAD) risk has been inconsistent, although insufficient power and incomplete assessment of MAPT haplotypes may account for this. Methods We examined the association of MAPT haplotypes with LOAD risk in more than 20,000 subjects (n-cases = 9,814, n-controls = 11,550) from Mayo Clinic (n-cases = 2,052, n-controls = 3,406) and the Alzheimer’s Disease Genetics Consortium (ADGC, n-cases = 7,762, n-controls = 8,144). We also assessed associations with brain MAPT gene expression levels measured in the cerebellum (n = 197) and temporal cortex (n = 202) of LOAD subjects. Six single nucleotide polymorphisms (SNPs) which tag MAPT haplotypes with frequencies greater than 1% were evaluated. Results H2-haplotype tagging rs8070723-G allele associated with reduced risk of LOAD (odds ratio, OR = 0.90, 95% confidence interval, CI = 0.85-0.95, p = 5.2E-05) with consistent results in the Mayo (OR = 0.81, p = 7.0E-04) and ADGC (OR = 0.89, p = 1.26E-04) cohorts. rs3785883-A allele was also nominally significantly associated with LOAD risk (OR = 1.06, 95% CI = 1.01-1.13, p = 0.034). Haplotype analysis revealed significant global association with LOAD risk in the combined cohort (p = 0.033), with significant association of the H2 haplotype with reduced risk of LOAD as expected (p = 1.53E-04) and suggestive association with additional haplotypes. MAPT SNPs and haplotypes also associated with brain MAPT levels in the cerebellum and temporal cortex of AD subjects with the strongest associations observed for the H2 haplotype and reduced brain MAPT levels (β = -0.16 to -0.20, p = 1.0E-03 to 3.0E-03). Conclusions These results confirm the previously reported MAPT H2 associations with LOAD risk in two large series, that this haplotype has the strongest effect on brain MAPT expression amongst those tested and identify additional haplotypes with suggestive associations, which require replication in independent series. These biologically congruent results provide compelling evidence to screen the MAPT region for regulatory variants which confer LOAD risk by influencing its brain gene expression.
Collapse
Affiliation(s)
- Mariet Allen
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | | | - Zachary Quicksall
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Fanggeng Zou
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - High Seng Chai
- Department of Health Sciences Research, Mayo Clinic Minnesota, Rochester, MN 55905, USA
| | - Curtis Younkin
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Julia E Crook
- Department of Health Sciences Research, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - V Shane Pankratz
- Department of Health Sciences Research, Mayo Clinic Minnesota, Rochester, MN 55905, USA
| | | | - Siddharth Krishnan
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Thuy Nguyen
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Li Ma
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Kimberly Malphrus
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Sarah Lincoln
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Gina Bisceglio
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | | | - Jin Jen
- Medical Genome Facility, Mayo Clinic Minnesota, Rochester, MN 55905, USA
| | | | - John K Kauwe
- Departments of Biology, Neuroscience, Brigham Young University, Provo, UT 84602, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle 98104, WA, USA
| | - Jonathan L Haines
- Department of Molecular Physiology and Biophysics, and the Vanderbilt Center for Human Genetics Research, Vanderbilt University, Nashville, TN, USA ; Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Richard Mayeux
- Gertrude H. Sergievsky Center, Department of Neurology, and Taub Institute on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Margaret A Pericak-Vance
- The John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - Lindsay A Farrer
- Departments of Biostatistics, Medicine (Genetics Program), Ophthalmology, Neurology, and Epidemiology, Boston University, Boston, MA, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Joseph E Parisi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ronald C Petersen
- Department of Neurology, Mayo Clinic Minnesota, Rochester, MN 55905, USA
| | - Neill R Graff-Radford
- Department of Neurology, Mayo Clinic Florida, 4500 San Pablo Road, Birdsall 3, Jacksonville, FL 32224, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Steven G Younkin
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA ; Department of Neurology, Mayo Clinic Florida, 4500 San Pablo Road, Birdsall 3, Jacksonville, FL 32224, USA
| |
Collapse
|
3183
|
Ferrari R, Hernandez DG, Nalls MA, Rohrer JD, Ramasamy A, Kwok JBJ, Dobson-Stone C, Brooks WS, Schofield PR, Halliday GM, Hodges JR, Piguet O, Bartley L, Thompson E, Haan E, Hernández I, Ruiz A, Boada M, Borroni B, Padovani A, Cruchaga C, Cairns NJ, Benussi L, Binetti G, Ghidoni R, Forloni G, Galimberti D, Fenoglio C, Serpente M, Scarpini E, Clarimón J, Lleó A, Blesa R, Waldö ML, Nilsson K, Nilsson C, Mackenzie IRA, Hsiung GYR, Mann DMA, Grafman J, Morris CM, Attems J, Griffiths TD, McKeith IG, Thomas AJ, Pietrini P, Huey ED, Wassermann EM, Baborie A, Jaros E, Tierney MC, Pastor P, Razquin C, Ortega-Cubero S, Alonso E, Perneczky R, Diehl-Schmid J, Alexopoulos P, Kurz A, Rainero I, Rubino E, Pinessi L, Rogaeva E, St George-Hyslop P, Rossi G, Tagliavini F, Giaccone G, Rowe JB, Schlachetzki JCM, Uphill J, Collinge J, Mead S, Danek A, Van Deerlin VM, Grossman M, Trojanowski JQ, van der Zee J, Deschamps W, Van Langenhove T, Cruts M, Van Broeckhoven C, Cappa SF, Le Ber I, Hannequin D, Golfier V, Vercelletto M, Brice A, Nacmias B, Sorbi S, Bagnoli S, Piaceri I, Nielsen JE, Hjermind LE, Riemenschneider M, Mayhaus M, Ibach B, Gasparoni G, Pichler S, Gu W, Rossor MN, et alFerrari R, Hernandez DG, Nalls MA, Rohrer JD, Ramasamy A, Kwok JBJ, Dobson-Stone C, Brooks WS, Schofield PR, Halliday GM, Hodges JR, Piguet O, Bartley L, Thompson E, Haan E, Hernández I, Ruiz A, Boada M, Borroni B, Padovani A, Cruchaga C, Cairns NJ, Benussi L, Binetti G, Ghidoni R, Forloni G, Galimberti D, Fenoglio C, Serpente M, Scarpini E, Clarimón J, Lleó A, Blesa R, Waldö ML, Nilsson K, Nilsson C, Mackenzie IRA, Hsiung GYR, Mann DMA, Grafman J, Morris CM, Attems J, Griffiths TD, McKeith IG, Thomas AJ, Pietrini P, Huey ED, Wassermann EM, Baborie A, Jaros E, Tierney MC, Pastor P, Razquin C, Ortega-Cubero S, Alonso E, Perneczky R, Diehl-Schmid J, Alexopoulos P, Kurz A, Rainero I, Rubino E, Pinessi L, Rogaeva E, St George-Hyslop P, Rossi G, Tagliavini F, Giaccone G, Rowe JB, Schlachetzki JCM, Uphill J, Collinge J, Mead S, Danek A, Van Deerlin VM, Grossman M, Trojanowski JQ, van der Zee J, Deschamps W, Van Langenhove T, Cruts M, Van Broeckhoven C, Cappa SF, Le Ber I, Hannequin D, Golfier V, Vercelletto M, Brice A, Nacmias B, Sorbi S, Bagnoli S, Piaceri I, Nielsen JE, Hjermind LE, Riemenschneider M, Mayhaus M, Ibach B, Gasparoni G, Pichler S, Gu W, Rossor MN, Fox NC, Warren JD, Spillantini MG, Morris HR, Rizzu P, Heutink P, Snowden JS, Rollinson S, Richardson A, Gerhard A, Bruni AC, Maletta R, Frangipane F, Cupidi C, Bernardi L, Anfossi M, Gallo M, Conidi ME, Smirne N, Rademakers R, Baker M, Dickson DW, Graff-Radford NR, Petersen RC, Knopman D, Josephs KA, Boeve BF, Parisi JE, Seeley WW, Miller BL, Karydas AM, Rosen H, van Swieten JC, Dopper EGP, Seelaar H, Pijnenburg YAL, Scheltens P, Logroscino G, Capozzo R, Novelli V, Puca AA, Franceschi M, Postiglione A, Milan G, Sorrentino P, Kristiansen M, Chiang HH, Graff C, Pasquier F, Rollin A, Deramecourt V, Lebert F, Kapogiannis D, Ferrucci L, Pickering-Brown S, Singleton AB, Hardy J, Momeni P. Frontotemporal dementia and its subtypes: a genome-wide association study. Lancet Neurol 2014; 13:686-99. [PMID: 24943344 PMCID: PMC4112126 DOI: 10.1016/s1474-4422(14)70065-1] [Show More Authors] [Citation(s) in RCA: 282] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Frontotemporal dementia (FTD) is a complex disorder characterised by a broad range of clinical manifestations, differential pathological signatures, and genetic variability. Mutations in three genes-MAPT, GRN, and C9orf72--have been associated with FTD. We sought to identify novel genetic risk loci associated with the disorder. METHODS We did a two-stage genome-wide association study on clinical FTD, analysing samples from 3526 patients with FTD and 9402 healthy controls. To reduce genetic heterogeneity, all participants were of European ancestry. In the discovery phase (samples from 2154 patients with FTD and 4308 controls), we did separate association analyses for each FTD subtype (behavioural variant FTD, semantic dementia, progressive non-fluent aphasia, and FTD overlapping with motor neuron disease [FTD-MND]), followed by a meta-analysis of the entire dataset. We carried forward replication of the novel suggestive loci in an independent sample series (samples from 1372 patients and 5094 controls) and then did joint phase and brain expression and methylation quantitative trait loci analyses for the associated (p<5 × 10(-8)) single-nucleotide polymorphisms. FINDINGS We identified novel associations exceeding the genome-wide significance threshold (p<5 × 10(-8)). Combined (joint) analyses of discovery and replication phases showed genome-wide significant association at 6p21.3, HLA locus (immune system), for rs9268877 (p=1·05 × 10(-8); odds ratio=1·204 [95% CI 1·11-1·30]), rs9268856 (p=5·51 × 10(-9); 0·809 [0·76-0·86]) and rs1980493 (p value=1·57 × 10(-8), 0·775 [0·69-0·86]) in the entire cohort. We also identified a potential novel locus at 11q14, encompassing RAB38/CTSC (the transcripts of which are related to lysosomal biology), for the behavioural FTD subtype for which joint analyses showed suggestive association for rs302668 (p=2·44 × 10(-7); 0·814 [0·71-0·92]). Analysis of expression and methylation quantitative trait loci data suggested that these loci might affect expression and methylation in cis. INTERPRETATION Our findings suggest that immune system processes (link to 6p21.3) and possibly lysosomal and autophagy pathways (link to 11q14) are potentially involved in FTD. Our findings need to be replicated to better define the association of the newly identified loci with disease and to shed light on the pathomechanisms contributing to FTD. FUNDING The National Institute of Neurological Disorders and Stroke and National Institute on Aging, the Wellcome/MRC Centre on Parkinson's disease, Alzheimer's Research UK, and Texas Tech University Health Sciences Center.
Collapse
Affiliation(s)
- Raffaele Ferrari
- Laboratory of Neurogenetics, Department of Internal Medicine, Texas Tech University Health Science Center, Lubbock, Texas, USA; Reta Lila Weston Research Laboratories, Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Dena G Hernandez
- Reta Lila Weston Research Laboratories, Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Michael A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan D Rohrer
- Reta Lila Weston Research Laboratories, Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK; Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Adaikalavan Ramasamy
- Reta Lila Weston Research Laboratories, Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK; Department of Medical and Molecular Genetics, King's College London, Guy's Hospital, London, UK
| | - John B J Kwok
- Neuroscience Research Australia, Sydney, NSW, Australia; University of New South Wales, Sydney, NSW, Australia
| | - Carol Dobson-Stone
- Neuroscience Research Australia, Sydney, NSW, Australia; University of New South Wales, Sydney, NSW, Australia
| | - William S Brooks
- Neuroscience Research Australia, Sydney, NSW, Australia; University of New South Wales, Sydney, NSW, Australia
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia; University of New South Wales, Sydney, NSW, Australia
| | - Glenda M Halliday
- Neuroscience Research Australia, Sydney, NSW, Australia; University of New South Wales, Sydney, NSW, Australia
| | - John R Hodges
- Neuroscience Research Australia, Sydney, NSW, Australia; University of New South Wales, Sydney, NSW, Australia
| | - Olivier Piguet
- Neuroscience Research Australia, Sydney, NSW, Australia; University of New South Wales, Sydney, NSW, Australia
| | | | - Elizabeth Thompson
- South Australian Clinical Genetics Service, SA Pathology at Women's and Children's Hospital, North Adelaide, SA, Australia; Department of Paediatrics, University of Adelaide, Adelaide, SA, Australia
| | - Eric Haan
- South Australian Clinical Genetics Service, SA Pathology at Women's and Children's Hospital, North Adelaide, SA, Australia; Department of Paediatrics, University of Adelaide, Adelaide, SA, Australia
| | - Isabel Hernández
- Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Agustín Ruiz
- Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Mercè Boada
- Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain; Hospital Universitari Vall d'Hebron-Institut de Recerca, Universitat Autonoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | | | | | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, USA; Hope Center, Washington University School of Medicine, St Louis, Missouri, USA
| | - Nigel J Cairns
- Hope Center, Washington University School of Medicine, St Louis, Missouri, USA; Department of Pathology and Immunology, Washington University, St Louis, Missouri, USA
| | - Luisa Benussi
- NeuroBioGen Lab-Memory Clinic, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Giuliano Binetti
- NeuroBioGen Lab-Memory Clinic, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- Proteomics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Gianluigi Forloni
- Biology of Neurodegenerative Disorders, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | - Daniela Galimberti
- University of Milan, Milan, Italy; Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- University of Milan, Milan, Italy; Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Serpente
- University of Milan, Milan, Italy; Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Elio Scarpini
- University of Milan, Milan, Italy; Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Jordi Clarimón
- Memory Unit, Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Center for Networker Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alberto Lleó
- Memory Unit, Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Center for Networker Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Rafael Blesa
- Memory Unit, Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Center for Networker Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Maria Landqvist Waldö
- Unit of Geriatric Psychiatry, Department of Clinical Sciences, Lund University, Sweden
| | - Karin Nilsson
- Unit of Geriatric Psychiatry, Department of Clinical Sciences, Lund University, Sweden
| | - Christer Nilsson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Sweden
| | - Ian R A Mackenzie
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Ging-Yuek R Hsiung
- Division of Neurology, University of British Columbia, Vancouver, Canada
| | - David M A Mann
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Salford Royal Hospital, Stott Lane, Salford, UK
| | - Jordan Grafman
- Rehabilitation Institute of Chicago, Departments of Physical Medicine and Rehabilitation, Psychiatry, and Cognitive Neurology and Alzheimer's Disease Center, IL, USA; Feinberg School of Medicine, Northwestern University, IL, USA; Department of Psychology, Weinberg College of Arts and Sciences, Northwestern University, IL, USA
| | - Christopher M Morris
- Rehabilitation Institute of Chicago, Departments of Physical Medicine and Rehabilitation, Psychiatry, and Cognitive Neurology and Alzheimer's Disease Center, IL, USA; Feinberg School of Medicine, Northwestern University, IL, USA; Newcastle Brain Tissue Resource, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK; Newcastle University, Institute for Ageing and Health, Campus for Ageing and Vitality, Newcastle upon Tyne, UK; Institute of Neuroscience, Newcastle University Medical School, Newcastle upon Tyne, UK
| | - Johannes Attems
- Rehabilitation Institute of Chicago, Departments of Physical Medicine and Rehabilitation, Psychiatry, and Cognitive Neurology and Alzheimer's Disease Center, IL, USA; Feinberg School of Medicine, Northwestern University, IL, USA; Newcastle Brain Tissue Resource, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK; Newcastle University, Institute for Ageing and Health, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Timothy D Griffiths
- Rehabilitation Institute of Chicago, Departments of Physical Medicine and Rehabilitation, Psychiatry, and Cognitive Neurology and Alzheimer's Disease Center, IL, USA; Feinberg School of Medicine, Northwestern University, IL, USA; Newcastle Brain Tissue Resource, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK; Institute of Neuroscience, Newcastle University Medical School, Newcastle upon Tyne, UK
| | - Ian G McKeith
- Biomedical Research Building, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Alan J Thomas
- Newcastle University, Institute for Ageing and Health, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - P Pietrini
- Clinical Psychology Branch, Pisa University Hospital, Pisa, Italy; Laboratory of Clinical Biochemistry and Molecular Biology, University of Pisa, Pisa, Italy
| | - Edward D Huey
- Taub Institute, Departments of Psychiatry and Neurology, Columbia University, New York, NY, USA 10032
| | - Eric M Wassermann
- Behavioral Neurology Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Atik Baborie
- Neuropathology Department, Walton Centre FT, Liverpool, UK
| | - Evelyn Jaros
- Newcastle University, Institute for Ageing and Health, Campus for Ageing and Vitality, Newcastle upon Tyne, UK; Neuropathology/Cellular Pathology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Michael C Tierney
- Behavioral Neurology Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Pau Pastor
- Center for Networker Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, Universidad de Navarra, Pamplona, Spain; Department of Neurology, Clínica Universidad de Navarra, University of Navarra School of Medicine, Pamplona, Spain
| | - Cristina Razquin
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, Universidad de Navarra, Pamplona, Spain
| | - Sara Ortega-Cubero
- Center for Networker Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, Universidad de Navarra, Pamplona, Spain
| | - Elena Alonso
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, Universidad de Navarra, Pamplona, Spain
| | - Robert Perneczky
- Neuroepidemiology and Ageing Research Unit, School of Public Health, Faculty of Medicine, The Imperial College of Science, Technology and Medicine, London, UK; West London Cognitive Disorders Treatment and Research Unit, West London Mental Health Trust, London TW8 8 DS, UK; Department of Psychiatry and Psychotherapy, Technische Universität München, Munich, Germany
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, Technische Universität München, Munich, Germany
| | - Panagiotis Alexopoulos
- Department of Psychiatry and Psychotherapy, Technische Universität München, Munich, Germany
| | - Alexander Kurz
- Department of Psychiatry and Psychotherapy, Technische Universität München, Munich, Germany
| | - Innocenzo Rainero
- Neurology I, Department of Neuroscience, University of Torino, Italy; AO Città della Salute e della Scienza di Torino, Italy
| | - Elisa Rubino
- Neurology I, Department of Neuroscience, University of Torino, Italy; AO Città della Salute e della Scienza di Torino, Italy
| | - Lorenzo Pinessi
- Neurology I, Department of Neuroscience, University of Torino, Italy; AO Città della Salute e della Scienza di Torino, Italy
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Peter St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Cambridge Institute for Medical Research and the Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Giacomina Rossi
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Fabrizio Tagliavini
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Giorgio Giaccone
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - James B Rowe
- Cambridge University Department of Clinical Neurosciences, Cambridge CB2 0SZ, UK; MRC Cognition and Brain Sciences Unit, Cambridge, UK; Behavioural and Clinical Neuroscience Institute, Cambridge, UK
| | - Johannes C M Schlachetzki
- Department of Psychiatry and Psychotherapy, University of Freiburg Medical School, Germany; Department of Molecular Neurology, University Hospital Erlangen, Erlangen, Germany
| | - James Uphill
- MRC Prion Unit, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - John Collinge
- MRC Prion Unit, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Simon Mead
- MRC Prion Unit, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Vivianna M Van Deerlin
- University of Pennsylvania Perelman School of Medicine, Department of Neurology and Penn Frontotemporal Degeneration Center, Philadelphia, PA, USA
| | - Murray Grossman
- University of Pennsylvania Perelman School of Medicine, Department of Neurology and Penn Frontotemporal Degeneration Center, Philadelphia, PA, USA
| | - John Q Trojanowski
- University of Pennsylvania Perelman School of Medicine, Department of Neurology and Penn Frontotemporal Degeneration Center, Philadelphia, PA, USA
| | - Julie van der Zee
- Neurodegenerative Brain Diseases group, Department of Molecular Genetics, VIB, Antwerp, Belgium; Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - William Deschamps
- Neurodegenerative Brain Diseases group, Department of Molecular Genetics, VIB, Antwerp, Belgium; Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Tim Van Langenhove
- Neurodegenerative Brain Diseases group, Department of Molecular Genetics, VIB, Antwerp, Belgium; Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Marc Cruts
- Neurodegenerative Brain Diseases group, Department of Molecular Genetics, VIB, Antwerp, Belgium; Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases group, Department of Molecular Genetics, VIB, Antwerp, Belgium; Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Stefano F Cappa
- Neurorehabilitation Unit, Deptartment Of Clinical Neuroscience, Vita-Salute University and San Raffaele Scientific Institute, Milan, Italy
| | - Isabelle Le Ber
- Inserm, UMR_S975, CRICM, F-75013; UPMC Univ Paris 06, UMR_S975, F-75013; and CNRS UMR 7225, F-75013, Paris, France; AP-HP, Hôpital de la Salpêtrière, Département de Neurologie-Centre de Références des Démences Rares, F-75013, Paris, France
| | - Didier Hannequin
- Service de Neurologie, Inserm U1079, CNR-MAJ, Rouen University Hospital, France
| | | | | | - Alexis Brice
- Inserm, UMR_S975, CRICM, F-75013; UPMC Univ Paris 06, UMR_S975, F-75013; and CNRS UMR 7225, F-75013, Paris, France; AP-HP, Hôpital de la Salpêtrière, Département de Neurologie-Centre de Références des Démences Rares, F-75013, Paris, France
| | - Benedetta Nacmias
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) University of Florence, Florence, Italy
| | - Sandro Sorbi
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) University of Florence, Florence, Italy
| | - Silvia Bagnoli
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) University of Florence, Florence, Italy
| | - Irene Piaceri
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) University of Florence, Florence, Italy
| | - Jørgen E Nielsen
- Danish Dementia Research Centre, Neurogenetics Clinic, Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Denmark; Department of Cellular and Molecular Medicine, Section of Neurogenetics, The Panum Institute, University of Copenhagen, Denmark
| | - Lena E Hjermind
- Danish Dementia Research Centre, Neurogenetics Clinic, Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Denmark; Department of Cellular and Molecular Medicine, Section of Neurogenetics, The Panum Institute, University of Copenhagen, Denmark
| | - Matthias Riemenschneider
- Saarland University Hospital, Department for Psychiatry and Psychotherapy, Homburg/Saar, Germany; Saarland University, Laboratory for Neurogenetics, Kirrberger, Homburg/Saar, Germany
| | - Manuel Mayhaus
- Saarland University, Laboratory for Neurogenetics, Kirrberger, Homburg/Saar, Germany
| | - Bernd Ibach
- University Regensburg, Department of Psychiatry, Psychotherapy and Psychosomatics, Universitätsstr 84, Regensburg, Germany
| | - Gilles Gasparoni
- Saarland University, Laboratory for Neurogenetics, Kirrberger, Homburg/Saar, Germany
| | - Sabrina Pichler
- Saarland University, Laboratory for Neurogenetics, Kirrberger, Homburg/Saar, Germany
| | - Wei Gu
- Saarland University, Laboratory for Neurogenetics, Kirrberger, Homburg/Saar, Germany; Luxembourg Centre For Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg
| | - Martin N Rossor
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Jason D Warren
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Maria Grazia Spillantini
- University of Cambridge, Department of Clinical Neurosciences, John Van Geest Brain Repair Centre, Cambridge, UK
| | - Huw R Morris
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, School of Medicine, Cardiff, UK
| | - Patrizia Rizzu
- German Center of Neurodegenerative Diseases-Tübingen, Tübingen, Germany
| | - Peter Heutink
- German Center of Neurodegenerative Diseases-Tübingen, Tübingen, Germany
| | - Julie S Snowden
- Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - Sara Rollinson
- Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - Anna Richardson
- Salford Royal Foundation Trust, Faculty of Medical and Human Sciences, University of Manchester, UK
| | - Alexander Gerhard
- Institute of Brain, Behaviour and Mental Health, The University of Manchester, Withington, Manchester, UK
| | - Amalia C Bruni
- Regional Neurogenetic Centre, ASPCZ, Lamezia Terme, Italy
| | | | | | - Chiara Cupidi
- Regional Neurogenetic Centre, ASPCZ, Lamezia Terme, Italy
| | - Livia Bernardi
- Regional Neurogenetic Centre, ASPCZ, Lamezia Terme, Italy
| | - Maria Anfossi
- Regional Neurogenetic Centre, ASPCZ, Lamezia Terme, Italy
| | - Maura Gallo
- Regional Neurogenetic Centre, ASPCZ, Lamezia Terme, Italy
| | | | | | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Matt Baker
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | | | | | - David Knopman
- Department of Neurology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Keith A Josephs
- Department of Neurology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Joseph E Parisi
- Department of Pathology, Mayo Clinic Rochester, Rochester, MN, USA
| | - William W Seeley
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Bruce L Miller
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Anna M Karydas
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Howard Rosen
- Department of Neurology, University of California, San Francisco, CA, USA
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands; Department of Medical Genetics, VU University Medical Centre, Amsterdam, The Netherlands
| | - Elise G P Dopper
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Centre and Department of Neurology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Centre and Department of Neurology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Giancarlo Logroscino
- Department of Basic Medical Sciences, Neurosciences and Sense Organs of the Aldo Moro, University of Bari, Italy
| | - Rosa Capozzo
- Department of Basic Medical Sciences, Neurosciences and Sense Organs of the Aldo Moro, University of Bari, Italy
| | - Valeria Novelli
- Department of Molecular Cardiology, IRCCS Fondazione S Maugeri, Pavia, Italy
| | - Annibale A Puca
- Cardiovascular Research Unit, IRCCS Multimedica, Milan, Italy; Department of Medicine and Surgery, University of Salerno, Baronissi (SA), Italy
| | | | - Alfredo Postiglione
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Graziella Milan
- Geriatric Center Frullone-ASL Napoli 1 Centro, Naples, Italy
| | | | | | - Huei-Hsin Chiang
- Karolinska Institutet, Department NVS, KI-Alzheimer Disease Research Center, Stockholm, Sweden; Department of Geriatric Medicine, Genetics Unit, Karolinska Universtiy Hospital, Stockholm
| | - Caroline Graff
- Karolinska Institutet, Department NVS, KI-Alzheimer Disease Research Center, Stockholm, Sweden; Department of Geriatric Medicine, Genetics Unit, Karolinska Universtiy Hospital, Stockholm
| | | | | | | | | | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Luigi Ferrucci
- Clinical Research Branch, National Institute on Aging, Baltimore, MD, USA
| | - Stuart Pickering-Brown
- Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - John Hardy
- Reta Lila Weston Research Laboratories, Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK.
| | - Parastoo Momeni
- Laboratory of Neurogenetics, Department of Internal Medicine, Texas Tech University Health Science Center, Lubbock, Texas, USA
| |
Collapse
|
3184
|
Ma J, Yu JT, Tan L. MS4A Cluster in Alzheimer's Disease. Mol Neurobiol 2014; 51:1240-8. [PMID: 24981432 DOI: 10.1007/s12035-014-8800-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 06/22/2014] [Indexed: 01/13/2023]
Abstract
Several variants within membrane-spanning 4-domains subfamily A (MS4A) gene cluster have recently been implicated the association of Alzheimer's disease (AD) by serial recent genome-wide association studies (GWAS). As cell membrane proteins, MS4A family members are found to participate in the regulation of calcium signaling which have been widely discussed in neurodegeneration and AD. Besides, although the MS4A family members are poorly characterized, an important role in immunity has already been identified for several members of this cluster (such as MS4A1, MS4A2, and MS4A4B), indicating the possible involvement of MS4A gene cluster in AD pathogenesis. In this article, we briefly summarize the structure, localization, and function of MS4A gene cluster, review recent genetic and expression findings concerning the association of MS4A gene cluster with AD pathogenesis, and also speculate the possible roles of MS4A gene cluster in this disease. Based on the contributing effects of MS4A gene cluster in AD pathogenesis, targeting MS4A gene cluster might provide new opportunities for AD treatment.
Collapse
Affiliation(s)
- Jing Ma
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | | | | |
Collapse
|
3185
|
Tan MS, Yu JT, Jiang T, Zhu XC, Guan HS, Tan L. Genetic variation in BIN1 gene and Alzheimer's disease risk in Han Chinese individuals. Neurobiol Aging 2014; 35:1781.e1-8. [DOI: 10.1016/j.neurobiolaging.2014.01.151] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 12/30/2013] [Accepted: 01/30/2014] [Indexed: 12/20/2022]
|
3186
|
Corbo RM, Gambina G, Broggio E, Scarabino D, Scacchi R. Association study of two steroid biosynthesis genes (COMT and CYP17) with Alzheimer's disease in the Italian population. J Neurol Sci 2014; 344:149-53. [PMID: 25012481 DOI: 10.1016/j.jns.2014.06.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/11/2014] [Accepted: 06/23/2014] [Indexed: 01/01/2023]
Abstract
The greater predisposition of women to Alzheimer's disease (AD), owing to the decrease in postmenopausal estrogen, may be influenced by polymorphic variation in genes regulating estrogen metabolism (e.g., COMT) and estrogen biosynthesis (e.g., CYP17). In order to better understand how the estrogen pathway genetic variation might affect AD onset, we conducted a case-control study of two single nucleotide polymorphisms (SNPs) of these two genes (COMT rs4680 and CYP17 rs743572) in a sample of AD patients of Italian origin. The COMT allele and genotype were found associated neither with AD onset nor with parameters of AD severity, such as cognitive impairment, age at onset, or disease duration. In contrast, CYP17 was found to affect the age at disease onset mainly in males and, as compared with noncarriers, people carrying the A2 (C) allele had a 2.2-fold increased risk for AD. These findings suggest that the CYP17 A2 allele influences AD susceptibility in a sex-specific way by acting not only on AD risk but also on the age at disease onset, an important parameter of AD severity.
Collapse
Affiliation(s)
- Rosa Maria Corbo
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University, Rome, Italy; CNR Institute of Cellular Biology and Neurobiology, Via E. Ramerini 32, 00015 Monterotondo Scalo, Rome, Italy.
| | - Giuseppe Gambina
- Alzheimer's Disease Center, Department of Neuroscience, University and Hospital of Verona, Verona, Italy
| | - Elisabetta Broggio
- Alzheimer's Disease Center, Department of Neuroscience, University and Hospital of Verona, Verona, Italy
| | - Daniela Scarabino
- CNR Institute of Cellular Biology and Neurobiology, Via E. Ramerini 32, 00015 Monterotondo Scalo, Rome, Italy
| | - Renato Scacchi
- CNR Institute of Molecular Biology and Pathology, c/o Department of Biology and Biotechnology "Charles Darwin", Sapienza University, Rome, Italy
| |
Collapse
|
3187
|
|
3188
|
|
3189
|
Sassi C, Guerreiro R, Gibbs R, Ding J, Lupton MK, Troakes C, Al-Sarraj S, Niblock M, Gallo JM, Adnan J, Killick R, Brown KS, Medway C, Lord J, Turton J, Bras J, Morgan K, Powell JF, Singleton A, Hardy J. Investigating the role of rare coding variability in Mendelian dementia genes (APP, PSEN1, PSEN2, GRN, MAPT, and PRNP) in late-onset Alzheimer's disease. Neurobiol Aging 2014; 35:2881.e1-2881.e6. [PMID: 25104557 PMCID: PMC4236585 DOI: 10.1016/j.neurobiolaging.2014.06.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 06/06/2014] [Accepted: 06/07/2014] [Indexed: 10/31/2022]
Abstract
The overlapping clinical and neuropathologic features between late-onset apparently sporadic Alzheimer's disease (LOAD), familial Alzheimer's disease (FAD), and other neurodegenerative dementias (frontotemporal dementia, corticobasal degeneration, progressive supranuclear palsy, and Creutzfeldt-Jakob disease) raise the question of whether shared genetic risk factors may explain the similar phenotype among these disparate disorders. To investigate this intriguing hypothesis, we analyzed rare coding variability in 6 Mendelian dementia genes (APP, PSEN1, PSEN2, GRN, MAPT, and PRNP), in 141 LOAD patients and 179 elderly controls, neuropathologically proven, from the UK. In our cohort, 14 LOAD cases (10%) and 11 controls (6%) carry at least 1 rare variant in the genes studied. We report a novel variant in PSEN1 (p.I168T) and a rare variant in PSEN2 (p.A237V), absent in controls and both likely pathogenic. Our findings support previous studies, suggesting that (1) rare coding variability in PSEN1 and PSEN2 may influence the susceptibility for LOAD and (2) GRN, MAPT, and PRNP are not major contributors to LOAD. Thus, genetic screening is pivotal for the clinical differential diagnosis of these neurodegenerative dementias.
Collapse
Affiliation(s)
- Celeste Sassi
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, UK; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| | - Rita Guerreiro
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, UK; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Raphael Gibbs
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, UK; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Jinhui Ding
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | | - Claire Troakes
- King's College London Institute of Psychiatry, London, UK
| | - Safa Al-Sarraj
- King's College London Institute of Psychiatry, London, UK
| | | | | | - Jihad Adnan
- King's College London Institute of Psychiatry, London, UK
| | | | - Kristelle S Brown
- Translation Cell Sciences-Human Genetics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, UK
| | - Christopher Medway
- Translation Cell Sciences-Human Genetics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, UK
| | - Jenny Lord
- Translation Cell Sciences-Human Genetics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, UK
| | - James Turton
- Translation Cell Sciences-Human Genetics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, UK
| | - Jose Bras
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, UK
| | | | - Kevin Morgan
- Translation Cell Sciences-Human Genetics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, UK
| | - John F Powell
- King's College London Institute of Psychiatry, London, UK
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, UK
| |
Collapse
|
3190
|
González F, Zhu Z, Shi ZD, Lelli K, Verma N, Li QV, Huangfu D. An iCRISPR platform for rapid, multiplexable, and inducible genome editing in human pluripotent stem cells. Cell Stem Cell 2014; 15:215-226. [PMID: 24931489 DOI: 10.1016/j.stem.2014.05.018] [Citation(s) in RCA: 367] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 05/06/2014] [Accepted: 05/28/2014] [Indexed: 12/26/2022]
Abstract
Human pluripotent stem cells (hPSCs) offer a unique platform for elucidating the genes and molecular pathways that underlie complex traits and diseases. To realize this promise, methods for rapid and controllable genetic manipulations are urgently needed. By combining two newly developed gene-editing tools, the TALEN and CRISPR/Cas systems, we have developed a genome-engineering platform in hPSCs, which we named iCRISPR. iCRISPR enabled rapid and highly efficient generation of biallelic knockout hPSCs for loss-of-function studies, as well as homozygous knockin hPSCs with specific nucleotide alterations for precise modeling of disease conditions. We further demonstrate efficient one-step generation of double- and triple-gene knockout hPSC lines, as well as stage-specific inducible gene knockout during hPSC differentiation. Thus the iCRISPR platform is uniquely suited for dissection of complex genetic interactions and pleiotropic gene functions in human disease studies and has the potential to support high-throughput genetic analysis in hPSCs.
Collapse
Affiliation(s)
- Federico González
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10065, USA
| | - Zengrong Zhu
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10065, USA
| | - Zhong-Dong Shi
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10065, USA
| | - Katherine Lelli
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10065, USA
| | - Nipun Verma
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10065, USA.,Weill Graduate School of Medical Sciences at Cornell University/The Rockefeller University/Sloan-Kettering Institute Tri-Institutional M.D.-Ph.D. Program, 1300 York Avenue, New York, NY 10065, USA
| | - Qing V Li
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10065, USA.,Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Danwei Huangfu
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10065, USA
| |
Collapse
|
3191
|
Escott-Price V, Bellenguez C, Wang LS, Choi SH, Harold D, Jones L, Holmans P, Gerrish A, Vedernikov A, Richards A, DeStefano AL, Lambert JC, Ibrahim-Verbaas CA, Naj AC, Sims R, Jun G, Bis JC, Beecham GW, Grenier-Boley B, Russo G, Thornton-Wells TA, Denning N, Smith AV, Chouraki V, Thomas C, Ikram MA, Zelenika D, Vardarajan BN, Kamatani Y, Lin CF, Schmidt H, Kunkle B, Dunstan ML, Vronskaya M, the United Kingdom Brain Expression Consortium, Johnson AD, Ruiz A, Bihoreau MT, Reitz C, Pasquier F, Hollingworth P, Hanon O, Fitzpatrick AL, Buxbaum JD, Campion D, Crane PK, Baldwin C, Becker T, Gudnason V, Cruchaga C, Craig D, Amin N, Berr C, Lopez OL, De Jager PL, Deramecourt V, Johnston JA, Evans D, Lovestone S, Letenneur L, Hernández I, Rubinsztein DC, Eiriksdottir G, Sleegers K, Goate AM, Fiévet N, Huentelman MJ, Gill M, Brown K, Kamboh MI, Keller L, Barberger-Gateau P, McGuinness B, Larson EB, Myers AJ, Dufouil C, Todd S, Wallon D, Love S, Rogaeva E, Gallacher J, George-Hyslop PS, Clarimon J, Lleo A, Bayer A, Tsuang DW, Yu L, Tsolaki M, Bossù P, Spalletta G, Proitsi P, Collinge J, Sorbi S, Garcia FS, Fox NC, Hardy J, Naranjo MCD, Bosco P, Clarke R, Brayne C, et alEscott-Price V, Bellenguez C, Wang LS, Choi SH, Harold D, Jones L, Holmans P, Gerrish A, Vedernikov A, Richards A, DeStefano AL, Lambert JC, Ibrahim-Verbaas CA, Naj AC, Sims R, Jun G, Bis JC, Beecham GW, Grenier-Boley B, Russo G, Thornton-Wells TA, Denning N, Smith AV, Chouraki V, Thomas C, Ikram MA, Zelenika D, Vardarajan BN, Kamatani Y, Lin CF, Schmidt H, Kunkle B, Dunstan ML, Vronskaya M, the United Kingdom Brain Expression Consortium, Johnson AD, Ruiz A, Bihoreau MT, Reitz C, Pasquier F, Hollingworth P, Hanon O, Fitzpatrick AL, Buxbaum JD, Campion D, Crane PK, Baldwin C, Becker T, Gudnason V, Cruchaga C, Craig D, Amin N, Berr C, Lopez OL, De Jager PL, Deramecourt V, Johnston JA, Evans D, Lovestone S, Letenneur L, Hernández I, Rubinsztein DC, Eiriksdottir G, Sleegers K, Goate AM, Fiévet N, Huentelman MJ, Gill M, Brown K, Kamboh MI, Keller L, Barberger-Gateau P, McGuinness B, Larson EB, Myers AJ, Dufouil C, Todd S, Wallon D, Love S, Rogaeva E, Gallacher J, George-Hyslop PS, Clarimon J, Lleo A, Bayer A, Tsuang DW, Yu L, Tsolaki M, Bossù P, Spalletta G, Proitsi P, Collinge J, Sorbi S, Garcia FS, Fox NC, Hardy J, Naranjo MCD, Bosco P, Clarke R, Brayne C, Galimberti D, Scarpini E, Bonuccelli U, Mancuso M, Siciliano G, Moebus S, Mecocci P, Zompo MD, Maier W, Hampel H, Pilotto A, Frank-García A, Panza F, Solfrizzi V, Caffarra P, Nacmias B, Perry W, Mayhaus M, Lannfelt L, Hakonarson H, Pichler S, Carrasquillo MM, Ingelsson M, Beekly D, Alvarez V, Zou F, Valladares O, Younkin SG, Coto E, Hamilton-Nelson KL, Gu W, Razquin C, Pastor P, Mateo I, Owen MJ, Faber KM, Jonsson PV, Combarros O, O'Donovan MC, Cantwell LB, Soininen H, Blacker D, Mead S, Mosley TH, Bennett DA, Harris TB, Fratiglioni L, Holmes C, de Bruijn RFAG, Passmore P, Montine TJ, Bettens K, Rotter JI, Brice A, Morgan K, Foroud TM, Kukull WA, Hannequin D, Powell JF, Nalls MA, Ritchie K, Lunetta KL, Kauwe JSK, Boerwinkle E, Riemenschneider M, Boada M, Hiltunen M, Martin ER, Schmidt R, Rujescu D, Dartigues JF, Mayeux R, Tzourio C, Hofman A, Nöthen MM, Graff C, Psaty BM, Haines JL, Lathrop M, Pericak-Vance MA, Launer LJ, Van Broeckhoven C, Farrer LA, van Duijn CM, Ramirez A, Seshadri S, Schellenberg GD, Amouyel P, Williams J. Gene-wide analysis detects two new susceptibility genes for Alzheimer's disease. PLoS One 2014; 9:e94661. [PMID: 24922517 PMCID: PMC4055488 DOI: 10.1371/journal.pone.0094661] [Show More Authors] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/17/2014] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Alzheimer's disease is a common debilitating dementia with known heritability, for which 20 late onset susceptibility loci have been identified, but more remain to be discovered. This study sought to identify new susceptibility genes, using an alternative gene-wide analytical approach which tests for patterns of association within genes, in the powerful genome-wide association dataset of the International Genomics of Alzheimer's Project Consortium, comprising over 7 m genotypes from 25,580 Alzheimer's cases and 48,466 controls. PRINCIPAL FINDINGS In addition to earlier reported genes, we detected genome-wide significant loci on chromosomes 8 (TP53INP1, p = 1.4×10-6) and 14 (IGHV1-67 p = 7.9×10-8) which indexed novel susceptibility loci. SIGNIFICANCE The additional genes identified in this study, have an array of functions previously implicated in Alzheimer's disease, including aspects of energy metabolism, protein degradation and the immune system and add further weight to these pathways as potential therapeutic targets in Alzheimer's disease.
Collapse
Affiliation(s)
- Valentina Escott-Price
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Céline Bellenguez
- Inserm U744, Lille, France
- Université Lille 2, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Seung-Hoan Choi
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Denise Harold
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Lesley Jones
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Peter Holmans
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Amy Gerrish
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Alexey Vedernikov
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Alexander Richards
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Anita L. DeStefano
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Jean-Charles Lambert
- Inserm U744, Lille, France
- Université Lille 2, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Carla A. Ibrahim-Verbaas
- Department of Epidemiology and Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Adam C. Naj
- Department of Biostatistics and Epidemiology and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Rebecca Sims
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Gyungah Jun
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Gary W. Beecham
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, Florida, United States of America
| | - Benjamin Grenier-Boley
- Inserm U744, Lille, France
- Université Lille 2, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Giancarlo Russo
- Functional Genomics Center Zurich, ETH/University of Zurich, Zurich, Switzerland
| | - Tricia A. Thornton-Wells
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Nicola Denning
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Albert V. Smith
- University of Iceland, Faculty of Medicine, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | - Vincent Chouraki
- Inserm U744, Lille, France
- Université Lille 2, Lille, France
- Institut Pasteur de Lille, Lille, France
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Charlene Thomas
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - M. Arfan Ikram
- Departments of Epidemiology, Neurology and Radiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Netherlands Consortium for Healthy Aging, Leiden, The Netherlands
| | - Diana Zelenika
- Centre National de Genotypage, Institut Genomique, Commissariat à l'énergie Atomique, Evry, France
| | - Badri N. Vardarajan
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
- Taub Institute on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University New York, New York, United States of America
- Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, New York, United States of America
| | | | - Chiao-Feng Lin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Helena Schmidt
- Institute for Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Brian Kunkle
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Melanie L. Dunstan
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Maria Vronskaya
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | | | - Andrew D. Johnson
- NHLBI Cardiovascular Epidemiology and Human Genomics Branch, The Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Agustin Ruiz
- Memory Clinic of Fundació ACE. Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Marie-Thérèse Bihoreau
- Centre National de Genotypage, Institut Genomique, Commissariat à l'énergie Atomique, Evry, France
| | - Christiane Reitz
- Taub Institute on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University New York, New York, United States of America
- Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, New York, United States of America
| | - Florence Pasquier
- Université Lille 2, Lille, France
- CNR-MAJ, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Paul Hollingworth
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Olivier Hanon
- University Paris Descartes, Sorbonne Paris V, Broca Hospital, Geriatrics department, Paris, France
| | - Annette L. Fitzpatrick
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Departments of Epidemiology and Global Health, University of Washington, Seattle, Washington, United States of America
| | - Joseph D. Buxbaum
- Department of Neuroscience, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America
- Departments of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Dominique Campion
- CNR-MAJ, Inserm U1079, Rouen University Hospital, 76031 France, Rouen, France
| | - Paul K. Crane
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Clinton Baldwin
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Tim Becker
- German Center for Neurodegenerative Diseases (DZNE), Bonn, and Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn, Germany
| | - Vilmundur Gudnason
- University of Iceland, Faculty of Medicine, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | - Carlos Cruchaga
- Department of Psychiatry and Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - David Craig
- Ageing Group, Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Claudine Berr
- INSERM U1061, Faculty of Medicine, Hôpital La Colombière, Montpellier, France
| | - Oscar L. Lopez
- Departments of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Philip L. De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Department of Neurology & Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute, Boston, Massachusetts, United States of America
| | - Vincent Deramecourt
- Université Lille 2, Lille, France
- CNR-MAJ, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Janet A. Johnston
- Ageing Group, Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Denis Evans
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Simon Lovestone
- King's College London, Institute of Psychiatry, Department of Neuroscience, De Crespigny Park, Denmark Hill, London, United Kingom
| | - Luc Letenneur
- Inserm U897, Victor Segalen University, F-33076, Bordeaux, France
| | - Isabel Hernández
- Memory Clinic of Fundació ACE. Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - David C. Rubinsztein
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | | | - Kristel Sleegers
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, Antwerp, Belgium
- Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Alison M. Goate
- Department of Psychiatry and Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Nathalie Fiévet
- Inserm U744, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Matthew J. Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Michael Gill
- Discipline of Psychiatry, Trinity College, Dublin, Ireland
| | - Kristelle Brown
- Institute of Genetics, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - M. Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Alzheimer's Disease Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Lina Keller
- Aging Reasearch Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Pascale Barberger-Gateau
- King's College London, Institute of Psychiatry, Department of Neuroscience, De Crespigny Park, Denmark Hill, London, United Kingom
| | - Bernadette McGuinness
- Ageing Group, Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Eric B. Larson
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Group Health Research Institute, Group Health, Seattle, Washington, United States of America
| | - Amanda J. Myers
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Carole Dufouil
- Inserm U897, Victor Segalen University, F-33076, Bordeaux, France
| | - Stephen Todd
- Ageing Group, Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - David Wallon
- CNR-MAJ, Inserm U1079, Rouen University Hospital, 76031 France, Rouen, France
| | - Seth Love
- University of Bristol Institute of Clinical Neurosciences, School of Clinical Sciences, Frenchay Hospital, Bristol, United Kingdom
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
| | - John Gallacher
- Institute of Primary Care and Public Health, Cardiff University, Neuadd Meirionnydd, University Hospital of Wales, Heath Park, Cardiff, United Kingdom
| | - Peter St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
- Cambridge Institute for Medical Research and Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Jordi Clarimon
- Neurology Department. IIB Sant Pau. Sant Pau Hospital. Universitat Autònoma de Barcelona, Barcelona, Spain
- Center for Networker Biomedical Research in Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Alberto Lleo
- Neurology Department. IIB Sant Pau. Sant Pau Hospital. Universitat Autònoma de Barcelona, Barcelona, Spain
- Center for Networker Biomedical Research in Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Anthony Bayer
- Institute of Primary Care and Public Health, Cardiff University, Neuadd Meirionnydd, University Hospital of Wales, Heath Park, Cardiff, United Kingdom
| | - Debby W. Tsuang
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, United States of America
| | - Lei Yu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Magda Tsolaki
- 3rd Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paola Bossù
- Clinical and Behavioral Neurology, Fondazione Santa Lucia, Roma, Italy
| | | | - Petra Proitsi
- King's College London, Institute of Psychiatry, Department of Neuroscience, De Crespigny Park, Denmark Hill, London, United Kingom
| | - John Collinge
- MRC Prion Unit, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Sandro Sorbi
- NEUROFARBA Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- Centro di Ricerca, Trasferimento e Alta Formazione DENOTHE, University of Florence, Florence, Italy
| | | | - Nick C. Fox
- Dementia Research Center, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - John Hardy
- Department of Molecular Neuroscience and Reta Lilla Weston Laboratories, Institute of Neurology, London, United Kingdom
| | | | - Paolo Bosco
- IRCCS Associazione Oasi Maria SS, Troina, Italy
| | - Robert Clarke
- Oxford Healthy Aging Project (OHAP), Clinical Trial Service Unit, University of Oxford, Oxford, United Kingdom
| | - Carol Brayne
- Cognitive Function and Ageing Study (CFAS), Institute of Public Health, University of Cambridge, Cambridge, United Kingdom
| | - Daniela Galimberti
- University of Milan, Fondazione Cà Granda, IRCCS Ospedale Policlinico, Milan, Italy
| | - Elio Scarpini
- University of Milan, Fondazione Cà Granda, IRCCS Ospedale Policlinico, Milan, Italy
| | | | | | | | - Susanne Moebus
- Urban Epidemiology, Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Patrizia Mecocci
- Section of Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, University of Perugia, Perugia, Italy
| | - Maria Del Zompo
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Wolfgang Maier
- Department of Psychiatry and Psychotherapy, University of Bonn, Germany and German Center for Neurodegenerative Diseases (DZNE, Bonn), Bonn, Germany
| | - Harald Hampel
- Department of Psychiatry, University of Frankfurt am Main, Frankfurt am Main, Germany (H.H.)
- Department of Psychiatry, Ludwig-Maximilians University, Munich, Germany
| | - Alberto Pilotto
- Gerontology and Geriatrics Research Laboratory, I.R.C.C.S. Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Ana Frank-García
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM); Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria “Hospital la Paz” (IdIPaz), Madrid, Spain
| | - Francesco Panza
- Department of Geriatrics,Center for Aging Brain,University of Bari, Bari, Italy
| | - Vincenzo Solfrizzi
- Department of Geriatrics,Center for Aging Brain,University of Bari, Bari, Italy
| | - Paolo Caffarra
- Department of Neuroscience-University of Parma, Parma, Italy
- Center for Cognitive Disorders AUSL, Parma, Italy
| | - Benedetta Nacmias
- NEUROFARBA Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- Centro di Ricerca, Trasferimento e Alta Formazione DENOTHE, University of Florence, Florence, Italy
| | - William Perry
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, Florida, United States of America
| | - Manuel Mayhaus
- Department Of Psychiatry, University Hospital, Saarland, Germany
| | - Lars Lannfelt
- Department of Public Health/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Sabrina Pichler
- Department Of Psychiatry, University Hospital, Saarland, Germany
| | | | - Martin Ingelsson
- Department of Public Health/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Duane Beekly
- National Alzheimer's Coordinating Center, University of Washington, Seattle, Washington, United States of America
| | | | - Fanggeng Zou
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Otto Valladares
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Steven G. Younkin
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, United States of America
| | | | - Kara L. Hamilton-Nelson
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Wei Gu
- Department of Psychiatry, University Hospital, Saarland, Germany
| | - Cristina Razquin
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, University of Navarra School of Medicine, Pamplona, Spain
| | - Pau Pastor
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, University of Navarra School of Medicine, Pamplona, Spain
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Ignacio Mateo
- Neurology Service and CIBERNED, "Marqués de Valdecilla" University Hospital (University of Cantabria and IFIMAV), Santander, Spain
| | - Michael J. Owen
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Kelley M. Faber
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, United States of America
| | - Palmi V. Jonsson
- University of Iceland, Faculty of Medicine, Reykjavik, Iceland
- Landspitali University Hospital, Reykjavik, Iceland
| | - Onofre Combarros
- Neurology Service and CIBERNED, "Marqués de Valdecilla" University Hospital (University of Cantabria and IFIMAV), Santander, Spain
| | - Michael C. O'Donovan
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| | - Laura B. Cantwell
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Hilkka Soininen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
- Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Deborah Blacker
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Department of Psychiatry, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Simon Mead
- MRC Prion Unit, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Thomas H. Mosley
- Department of Medicine (Geriatrics), University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - David A. Bennett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, United States of America
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Tamara B. Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute of Health, Bethesda, Maryland, United States of America
| | - Laura Fratiglioni
- Aging Research Center, Department Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Department Geriatric Medicine, Genetics Unit, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Clive Holmes
- Division of Clinical Neurosciences, School of Medicine, University of Southampton, Southampton, United Kingdom
| | - Renee F. A. G. de Bruijn
- Departments of Neurology and Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Peter Passmore
- Ageing Group, Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Thomas J. Montine
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Karolien Bettens
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, Antwerp, Belgium
- Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Alexis Brice
- INSERM UMR_S975-CNRS UMR 7225, Université Pierre et Marie Curie, Centre de recherche de l'Institut du Cerveau et de la Moëlle épinière-CRICM, Hôpital de la Salpêtrière, Paris France
- AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Kevin Morgan
- Institute of Genetics, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Tatiana M. Foroud
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, United States of America
| | - Walter A. Kukull
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Didier Hannequin
- CNR-MAJ, Inserm U1079, Rouen University Hospital, 76031 France, Rouen, France
| | - John F. Powell
- King's College London, Institute of Psychiatry, Department of Neuroscience, De Crespigny Park, Denmark Hill, London, United Kingom
| | - Michael A. Nalls
- Laboratory of Neurogenetics, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Karen Ritchie
- INSERM U1061, Faculty of Medicine, Hôpital La Colombière, Montpellier, France
- Imperial College, London, United Kingdom
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - John S. K. Kauwe
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Eric Boerwinkle
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Human Genetics Center and Div. of Epidemiology, University of Texas Health Sciences Center at Houston, Houston, Texas, United States of America
| | | | - Mercè Boada
- Memory Clinic of Fundació ACE. Institut Català de Neurociències Aplicades, Barcelona, Spain
- Hospital Universitari Vall d'Hebron - Institut de Recerca, Universitat Autònoma de Barcelona. (VHIR-UAB), Barcelona, Spain
| | - Mikko Hiltunen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
- Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Eden R. Martin
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, Florida, United States of America
| | | | - Dan Rujescu
- Department of Psychiatry, Ludwig-Maximilians University, Munich, Germany
| | - Jean-François Dartigues
- Inserm U897, Victor Segalen University, F-33076, Bordeaux, France
- Centre de Mémoire de Ressources et de Recherche de Bordeaux, CHU de Bordeaux, Bordeaux, France
| | - Richard Mayeux
- Taub Institute on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University New York, New York, United States of America
- Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, New York, United States of America
| | | | - Albert Hofman
- Departments of Epidemiology, Neurology and Radiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Netherlands Consortium for Healthy Aging, Leiden, The Netherlands
| | - Markus M. Nöthen
- Institute of Human Genetics, Department of Genomics, Life and Brain Center, University of Bonn, and German Center for Neurodegenerative Diseases (DZNE, Bonn), Bonn, Germany
| | - Caroline Graff
- Department Geriatric Medicine, Genetics Unit, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, KIADRC, Stockholm, Sweden
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington, United States of America
| | - Jonathan L. Haines
- Vanderbilt Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Epidemiology & Biostatistics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Mark Lathrop
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Fondation Jean Dausset- CEPH, Paris, France
- McGill University and Génome Québec Innovation Centre, Montreal, Canada
| | - Margaret A. Pericak-Vance
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, Florida, United States of America
| | - Lenore J. Launer
- Laboratory of Epidemiology, Demography, and Biometry, National Institute of Health, Bethesda, Maryland, United States of America
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, Antwerp, Belgium
- Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Lindsay A. Farrer
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, United States of America
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Cornelia M. van Duijn
- Netherlands Consortium for Healthy Aging, Leiden, The Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Center for Medical Systems Biology, Leiden, The Netherlands
| | - Alfredo Ramirez
- Department of Psychiatry and Psychotherapy and Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- The Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Philippe Amouyel
- Inserm U744, Lille, France
- Université Lille 2, Lille, France
- Institut Pasteur de Lille, Lille, France
- CNR-MAJ, Centre Hospitalier Régional Universitaire de Lille, Lille, France
- Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Julie Williams
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
3192
|
Malnar M, Hecimovic S, Mattsson N, Zetterberg H. Bidirectional links between Alzheimer's disease and Niemann-Pick type C disease. Neurobiol Dis 2014; 72 Pt A:37-47. [PMID: 24907492 DOI: 10.1016/j.nbd.2014.05.033] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 05/17/2014] [Accepted: 05/27/2014] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) and Niemann-Pick type C (NPC) disease are progressive neurodegenerative diseases with very different epidemiology and etiology. AD is a common cause of dementia with a complex polyfactorial etiology, including both genetic and environmental risk factors, while NPC is a very rare autosomal recessive disease. However, the diseases share some disease-related molecular pathways, including abnormal cholesterol metabolism, and involvement of amyloid-β (Aβ) and tau pathology. Here we review recent studies on these pathological traits, focusing on studies of Aβ and tau pathology in NPC, and the importance of the NPC1 gene in AD. Further studies of similarities and differences between AD and NPC may be useful to increase the understanding of both these devastating neurological diseases.
Collapse
Affiliation(s)
- Martina Malnar
- Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Silva Hecimovic
- Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia.
| | - Niklas Mattsson
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (CIND), San Francisco VA Medical Center, San Francisco, CA, USA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Gothenburg, Sweden; UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
| |
Collapse
|
3193
|
Benitez BA, Jin SC, Guerreiro R, Graham R, Lord J, Harold D, Sims R, Lambert JC, Gibbs JR, Bras J, Sassi C, Harari O, Bertelsen S, Lupton MK, Powell J, Bellenguez C, Brown K, Medway C, Haddick PCG, van der Brug MP, Bhangale T, Ortmann W, Behrens T, Mayeux R, Pericak-Vance MA, Farrer LA, Schellenberg GD, Haines JL, Turton J, Braae A, Barber I, Fagan AM, Holtzman DM, Morris JC, The 3C Study Group, the EADI consortium, the Alzheimer's Disease Genetic Consortium (ADGC), Alzheimer's Disease Neuroimaging Initiative (ADNI), the GERAD Consortium, Williams J, Kauwe JS, Amouyel P, Morgan K, Singleton A, Hardy J, Goate AM, Cruchaga C, Singleton A, Hardy J, Goate AM, Cruchaga C. Missense variant in TREML2 protects against Alzheimer's disease. Neurobiol Aging 2014; 35:1510.e19-26. [PMID: 24439484 PMCID: PMC3961557 DOI: 10.1016/j.neurobiolaging.2013.12.010] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 12/09/2013] [Accepted: 12/13/2013] [Indexed: 01/22/2023]
Abstract
TREM and TREM-like receptors are a structurally similar protein family encoded by genes clustered on chromosome 6p21.11. Recent studies have identified a rare coding variant (p.R47H) in TREM2 that confers a high risk for Alzheimer's disease (AD). In addition, common single nucleotide polymorphisms in this genomic region are associated with cerebrospinal fluid biomarkers for AD and a common intergenic variant found near the TREML2 gene has been identified to be protective for AD. However, little is known about the functional variant underlying the latter association or its relationship with the p.R47H. Here, we report comprehensive analyses using whole-exome sequencing data, cerebrospinal fluid biomarker analyses, meta-analyses (16,254 cases and 20,052 controls) and cell-based functional studies to support the role of the TREML2 coding missense variant p.S144G (rs3747742) as a potential driver of the meta-analysis AD-associated genome-wide association studies signal. Additionally, we demonstrate that the protective role of TREML2 in AD is independent of the role of TREM2 gene as a risk factor for AD.
Collapse
Affiliation(s)
- Bruno A. Benitez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Sheng Chih Jin
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Rita Guerreiro
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK,Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Rob Graham
- Diagnostic Discovery Department, Genentech Inc, South San Francisco, CA, USA
| | - Jenny Lord
- Human Genetics, School of Molecular Medical Sciences, University of Nottingham, Nottingham, UK
| | - Denise Harold
- Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Rebecca Sims
- Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Jean-Charles Lambert
- Inserm, Lille, France,Universite Lille 2, Lille, France,Institut Pasteur de Lille, Lille, France
| | - J. Raphael Gibbs
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK,Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Jose Bras
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Celeste Sassi
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK,Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Oscar Harari
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah Bertelsen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | - John Powell
- Institute of Psychiatry, King's College London, London, UK
| | - Celine Bellenguez
- Inserm, Lille, France,Universite Lille 2, Lille, France,Institut Pasteur de Lille, Lille, France
| | - Kristelle Brown
- Human Genetics, School of Molecular Medical Sciences, University of Nottingham, Nottingham, UK
| | - Christopher Medway
- Human Genetics, School of Molecular Medical Sciences, University of Nottingham, Nottingham, UK
| | - Patrick CG. Haddick
- Diagnostic Discovery Department, Genentech Inc, South San Francisco, CA, USA
| | | | - Tushar Bhangale
- Department of Bioinformatics and Computational Biology, Genentech Inc, South San Francisco, CA, USA
| | - Ward Ortmann
- Human Genetics Department, Genentech Inc, South San Francisco, CA, USA
| | - Tim Behrens
- Human Genetics Department, Genentech Inc, South San Francisco, CA, USA
| | - Richard Mayeux
- Department of Neurology, Taub Institute on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA,Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
| | - Margaret A. Pericak-Vance
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA,Dr John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - Lindsay A. Farrer
- Department of Medicine, Boston University Schools of Medicine and Public Health, Boston, MA, USA,Department of Biomedical Genetics, Boston University Schools of Medicine and Public Health, Boston, MA, USA,Department of Neurology, Boston University Schools of Medicine and Public Health, Boston, MA, USA,Department of Ophthalmology, Boston University Schools of Medicine and Public Health, Boston, MA, USA,Department of Epidemiology, Boston University Schools of Medicine and Public Health, Boston, MA, USA,Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, MA, USA
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan L. Haines
- Department of Molecular Physiology and Biophysics, Vanderbilt Center for Human Genetics Research, Vanderbilt University, Nashville, TN, USA
| | - Jim Turton
- Human Genetics, School of Molecular Medical Sciences, University of Nottingham, Nottingham, UK
| | - Anne Braae
- Human Genetics, School of Molecular Medical Sciences, University of Nottingham, Nottingham, UK
| | - Imelda Barber
- Human Genetics, School of Molecular Medical Sciences, University of Nottingham, Nottingham, UK
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Julie Williams
- Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - John S.K. Kauwe
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Philippe Amouyel
- Inserm, Lille, France,Universite Lille 2, Lille, France,Institut Pasteur de Lille, Lille, France
| | - Kevin Morgan
- Human Genetics, School of Molecular Medical Sciences, University of Nottingham, Nottingham, UK
| | - Andy Singleton
- Human Genetics Department, Genentech Inc, South San Francisco, CA, USA
| | - John Hardy
- Human Genetics Department, Genentech Inc, South San Francisco, CA, USA
| | - Alison M. Goate
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA,Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA,Alternate corresponding author at: Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO 63110, USA. Tel.: +314 286 0546; fax: +314 747 2983.
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA; Universite Lille 2, Lille, France.
| | - Andy Singleton
- Human Genetics Department, Genentech Inc, South San Francisco, CA, USA
| | - John Hardy
- Human Genetics Department, Genentech Inc, South San Francisco, CA, USA
| | - Alison M Goate
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA; Universite Lille 2, Lille, France.
| |
Collapse
|
3194
|
Hinney A, Albayrak O, Antel J, Volckmar AL, Sims R, Chapman J, Harold D, Gerrish A, Heid IM, Winkler TW, Scherag A, Wiltfang J, Williams J, Hebebrand J. Genetic variation at the CELF1 (CUGBP, elav-like family member 1 gene) locus is genome-wide associated with Alzheimer's disease and obesity. Am J Med Genet B Neuropsychiatr Genet 2014; 165B:283-93. [PMID: 24788522 DOI: 10.1002/ajmg.b.32234] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/10/2014] [Indexed: 01/01/2023]
Abstract
Deviations from normal body weight are observed prior to and after the onset of Alzheimer's disease (AD). Midlife obesity confers increased AD risk in later life, whereas late-life obesity is associated with decreased AD risk. The role of underweight and weight loss for AD risk is controversial. Based on the hypothesis of shared genetic variants for both obesity and AD, we analyzed the variants identified for AD or obesity from genome-wide association meta-analyses of the GERAD (AD, cases = 6,688, controls = 13,685) and GIANT (body mass index [BMI] as measure of obesity, n = 123,865) consortia. Our cross-disorder analysis of genome-wide significant 39 obesity SNPs and 23 AD SNPs in these two large data sets revealed that: (1) The AD SNP rs10838725 (pAD = 1.1 × 10(-08)) at the locus CELF1 is also genome-wide significant for obesity (pBMI = 7.35 × 10(-09) ). (2) Four additional AD risk SNPs were nominally associated with obesity (rs17125944 at FERMT2, pBMI = 4.03 × 10(-05), pBMI corr = 2.50 × 10(-03) ; rs3851179 at PICALM; pBMI = 0.002, rs2075650 at TOMM40/APOE, pBMI = 0.024, rs3865444 at CD33, pBMI = 0.024). (3) SNPs at two of the obesity risk loci (rs4836133 downstream of ZNF608; pAD = 0.002 and at rs713586 downstream of RBJ/DNAJC27; pAD = 0.018) were nominally associated with AD risk. Additionally, among the SNPs used for confirmation in both studies the AD risk allele of rs1858973, with an AD association just below genome-wide significance (pAD = 7.20 × 10(-07)), was also associated with obesity (SNP at IQCK/GPRC5B; pBMI = 5.21 × 10(-06) ; pcorr = 3.24 × 10(-04)). Our first GWAS based cross-disorder analysis for AD and obesity suggests that rs10838725 at the locus CELF1 might be relevant for both disorders.
Collapse
Affiliation(s)
- Anke Hinney
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, Universitätsklinikum Essen, University of Duisburg-Essen, Essen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3195
|
Knowles TPJ, Vendruscolo M, Dobson CM. The amyloid state and its association with protein misfolding diseases. Nat Rev Mol Cell Biol 2014; 15:384-96. [PMID: 24854788 DOI: 10.1038/nrm3810] [Citation(s) in RCA: 1727] [Impact Index Per Article: 157.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The phenomenon of protein aggregation and amyloid formation has become the subject of rapidly increasing research activities across a wide range of scientific disciplines. Such activities have been stimulated by the association of amyloid deposition with a range of debilitating medical disorders, from Alzheimer's disease to type II diabetes, many of which are major threats to human health and welfare in the modern world. It has become clear, however, that the ability to form the amyloid state is more general than previously imagined, and that its study can provide unique insights into the nature of the functional forms of peptides and proteins, as well as understanding the means by which protein homeostasis can be maintained and protein metastasis avoided.
Collapse
Affiliation(s)
- Tuomas P J Knowles
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Michele Vendruscolo
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| |
Collapse
|
3196
|
Logue MW, Schu M, Vardarajan BN, Farrell J, Lunetta KL, Jun G, Baldwin CT, DeAngelis MM, Farrer LA. Search for age-related macular degeneration risk variants in Alzheimer disease genes and pathways. Neurobiol Aging 2014; 35:1510.e7-18. [PMID: 24439028 PMCID: PMC3961547 DOI: 10.1016/j.neurobiolaging.2013.12.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 11/22/2013] [Accepted: 12/12/2013] [Indexed: 12/17/2022]
Abstract
Several lines of inquiry point to overlapping molecular mechanisms between late-onset Alzheimer disease (AD) and age-related macular degeneration (AMD). We evaluated summarized results from large genome-wide association studies for AD and AMD to test the hypothesis that AD susceptibility loci are also associated with AMD. We observed association of both disorders with genes in a region of chromosome 7, including PILRA and ZCWPW1 (peak AMD SNP rs7792525, minor allele frequency [MAF] = 19%, odds ratio [OR] = 1.14, p = 2.34 × 10(-6)), and with ABCA7 (peak AMD SNP rs3752228, MAF = 0.054, OR = 1.22, p = 0.00012). Next, we evaluated association of AMD with genes in AD-related pathways identified by canonical pathway analysis of AD-associated genes. Significant associations were observed with multiple previously identified AMD risk loci and 2 novel genes: HGS (peak SNP rs8070488, MAF = 0.23, OR = 0.91, p = 7.52 × 10(-5)), which plays a role in the clathrin-mediated endocytosis signaling pathway, and TNF (peak SNP rs2071590, MAF = 0.34, OR = 0.89, p = 1.17 × 10(-5)), which is a member of the atherosclerosis signaling and the LXR/RXR activation pathways. Our results suggest that AMD and AD share genetic mechanisms.
Collapse
Affiliation(s)
- Mark W. Logue
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA.,Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA
| | - Matthew Schu
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA
| | - Badri N. Vardarajan
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA
| | - John Farrell
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA
| | - Gyungah Jun
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA.,Department of Ophthalmology, Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA.,Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA
| | - Clinton T. Baldwin
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA
| | | | - Lindsay A. Farrer
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA.,Department of Neurology, Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA.,Department of Ophthalmology, Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA.,Department of Epidemiology, Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA.,Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, MA, 02118, USA.,Corresponding Author: Dr. Lindsay A. Farrer, Boston University School of Medicine, Biomedical Genetics L320, 72 East Concord Street, Boston, MA 02118, Tel: (617) 638-5393, Fax: (617) 638-4275,
| |
Collapse
|
3197
|
Medland SE, Jahanshad N, Neale BM, Thompson PM. Whole-genome analyses of whole-brain data: working within an expanded search space. Nat Neurosci 2014; 17:791-800. [PMID: 24866045 PMCID: PMC4300949 DOI: 10.1038/nn.3718] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 04/03/2014] [Indexed: 02/06/2023]
Abstract
Large-scale comparisons of patients and healthy controls have unearthed genetic risk factors associated with a range of neurological and psychiatric illnesses. Meanwhile, brain imaging studies are increasing in size and scope, revealing disease and genetic effects on brain structure and function, and implicating neural pathways and causal mechanisms. With the advent of global neuroimaging consortia, imaging studies are now well powered to discover genetic variants that reliably affect the brain. Genetic analyses of brain measures from tens of thousands of people are being extended to test genetic associations with signals at millions of locations in the brain, and connectome-wide, genome-wide scans can jointly screen brain circuits and genomes; these analyses and others present new statistical challenges. There is a growing need for the community to establish and enforce standards in this developing field to ensure robust findings. Here we discuss how neuroimagers and geneticists have formed alliances to discover how genetic factors affect the brain. The field is rapidly advancing with ultra-high-resolution imaging and whole-genome sequencing. We recommend a rigorous approach to neuroimaging genomics that capitalizes on its recent successes and ensures the reliability of future discoveries.
Collapse
Affiliation(s)
- Sarah E Medland
- Quantitative Genetics, Queensland Institute of Medical Research Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Neda Jahanshad
- Imaging Genetics Center, Institute for Neuroimaging & Informatics, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- Department of Neurology, University of Southern California, Los Angeles, California, USA
| | - Benjamin M Neale
- Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
- Psychiatric and Neurodevelopmental Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Paul M Thompson
- Imaging Genetics Center, Institute for Neuroimaging & Informatics, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- Imaging Genetics Center, Institute for Neuroimaging & Informatics, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- Department of Neurology, University of Southern California, Los Angeles, California, USA
- Department of Psychiatry, University of Southern California, Los Angeles, California, USA
- Department of Engineering, University of Southern California, Los Angeles, California, USA
- Department of Radiology, University of Southern California, Los Angeles, California, USA
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
- Department of Ophthalmology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
3198
|
Shen N, Chen B, Jiang Y, Feng R, Liao M, Zhang L, Li F, Ma G, Chen Z, Zhao B, Li K, Liu G. An Updated Analysis with 85,939 Samples Confirms the Association Between CR1 rs6656401 Polymorphism and Alzheimer's Disease. Mol Neurobiol 2014; 51:1017-23. [PMID: 24878768 DOI: 10.1007/s12035-014-8761-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 05/22/2014] [Indexed: 01/29/2023]
Abstract
The complement receptor 1 (CR1) rs6656401 polymorphism was first identified to be associated with Alzheimer's disease (AD) in European ancestry. However, the following studies reported weak or no significant association in Chinese, Japanese, Korean, African-American, Polish, and Canadian populations. We think that these negative results may have been caused by either relatively small sample sizes compared with those used for the previous genome-wide association studies (GWAS) in European ancestry or the genetic heterogeneity of the rs6656401 polymorphism in different populations. Here, we reevaluated this association using the relatively large-scale samples from previous 24 studies (N = 85,939, 30,100 cases and 55,839 controls) by searching the PubMed, AlzGene, and Google Scholar databases. Using additive model, we did not identify significant heterogeneity among the 24 studies. We observed significant association between the rs6656401 polymorphism and AD in pooled populations (P = 1.82E-26, odds ratio (OR) = 1.18, 95 % confidence interval (CI) 1.15-1.22). In subgroup analysis, we identified significant results in East Asian population with P = 5.00E-04, OR = 1.31, 95 % CI 1.13-1.52. To our knowledge, this is the first meta-analysis to investigate the association between rs6656401 polymorphism and AD in East Asian, African-American, Canadian, and European populations. Our analysis further supports previous findings that the CR1 rs6656401 polymorphism contributes to AD susceptibility. We believe that our findings will be very useful for future genetic studies on AD.
Collapse
Affiliation(s)
- Ning Shen
- Department of Physiology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3199
|
Zhao QF, Yu JT, Tan MS, Tan L. ABCA7 in Alzheimer's Disease. Mol Neurobiol 2014; 51:1008-16. [PMID: 24878767 DOI: 10.1007/s12035-014-8759-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 05/21/2014] [Indexed: 12/20/2022]
Abstract
ATP-binding cassette A7 (ABCA7) gene has recently been identified as a strong genetic locus associated with late-onset Alzheimer's disease (LOAD) through genome-wide association studies (GWASs). ABCA7 is a member of the ATP-binding cassette (ABC) transporter gene superfamily, which codes for 49 ABC proteins, divided into 7 subfamilies (coded A-G). As a multispan transmembrane protein, ABCA7 is most abundantly expressed in the microglial cells in the brain. The levels of ABCA7 have been detected to be increased in the Alzheimer's disease (AD) brain, which positively correlated with amyloid plaque burden and disease severity. Emerging data suggests that ABCA7 could be associated with AD via various pathways, possibly including amyloid-β (Aβ) accumulation, lipid metabolism, and phagocytosis. In this review, we summarize the known functions of ABCA7 and discuss the single-nucleotide polymorphisms (SNPs) related to LOAD, as well as their potential physiological effects. Finally, given the contributions of ABCA7 to AD pathogenesis, targeting ABCA7 might provide novel opportunities for AD therapy.
Collapse
Affiliation(s)
- Qing-Fei Zhao
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | | | | | | |
Collapse
|
3200
|
Gratten J, Wray NR, Keller MC, Visscher PM. Large-scale genomics unveils the genetic architecture of psychiatric disorders. Nat Neurosci 2014; 17:782-90. [PMID: 24866044 DOI: 10.1038/nn.3708] [Citation(s) in RCA: 252] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/27/2014] [Indexed: 12/11/2022]
Abstract
Family study results are consistent with genetic effects making substantial contributions to risk of psychiatric disorders such as schizophrenia, yet robust identification of specific genetic variants that explain variation in population risk had been disappointing until the advent of technologies that assay the entire genome in large samples. We highlight recent progress that has led to a better understanding of the number of risk variants in the population and the interaction of allele frequency and effect size. The emerging genetic architecture implies a large number of contributing loci (that is, a high genome-wide mutational target) and suggests that genetic risk of psychiatric disorders involves the combined effects of many common variants of small effect, as well as rare and de novo variants of large effect. The capture of a substantial proportion of genetic risk facilitates new study designs to investigate the combined effects of genes and the environment.
Collapse
Affiliation(s)
- Jacob Gratten
- 1] Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia. [2] These authors contributed equally to this work
| | - Naomi R Wray
- 1] Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia. [2] These authors contributed equally to this work
| | - Matthew C Keller
- 1] Department of Psychology and Neuroscience, University of Colorado, Boulder, Colorado, USA. [2] These authors contributed equally to this work
| | - Peter M Visscher
- 1] Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia. [2] The Translational Research Institute, University of Queensland Diamantina Institute, Brisbane, Queensland, Australia. [3] These authors contributed equally to this work
| |
Collapse
|