3101
|
Excess of rare coding variants in PLD3 in late- but not early-onset Alzheimer's disease. Hum Genome Var 2015; 2:14028. [PMID: 27081517 PMCID: PMC4785568 DOI: 10.1038/hgv.2014.28] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 09/28/2014] [Accepted: 10/28/2014] [Indexed: 11/25/2022] Open
Abstract
Recently, mutations in phospholipase D3 (PLD3) were reported in late-onset Alzheimer's disease (AD). By screening the coding regions of PLD3 for variants in a European cohort of 1,089 AD cases, 182 individuals with frontotemporal lobar degeneration and 1,456 controls, we identified 32 variants with a minor allele frequency <5% and observed an excess of rare variants in individuals with late- but not early-onset AD (P=0.034, χ2-test; odds ratio=1.46).
Collapse
|
3102
|
Ferencz B, Gerritsen L. Genetics and underlying pathology of dementia. Neuropsychol Rev 2015; 25:113-24. [PMID: 25567624 DOI: 10.1007/s11065-014-9276-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 12/21/2014] [Indexed: 12/14/2022]
Abstract
As the population steadily ages, dementia, in all its forms, remains a great societal challenge. Yet, our knowledge of their etiology remains rather limited. To this end, genetic studies can give us insight into the underlying mechanisms that lead to the development of dementia, potentially facilitating treatments in the future. In this review we cover the most recent genetic risk factors associated with the onset of the four most common dementia types today, including Alzheimer's disease (AD), Vascular Dementia (VaD), Frontotemporal Lobar Degeneration (FTLD) and Lewy Body Dementia (LBD). Moreover, we discuss the overlap in major underlying pathologies of dementia derived from their genetic associations. While all four dementia types appear to involve genes associated with tau-pathology and neuroinflammation only LBD, AD and VaD appear to involve amyloid genes while LBD and FTLD share alpha synuclein genes. Together these findings suggest that some of the dementias may exist along a spectrum and demonstrates the necessity to conduct large-scale studies pinpointing the etiology of the dementias and potential gene and environment interactions that may influence their development.
Collapse
Affiliation(s)
- Beata Ferencz
- Aging Research Center (ARC), Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | | |
Collapse
|
3103
|
Chauhan G, Adams HHH, Bis JC, Weinstein G, Yu L, Töglhofer AM, Smith AV, van der Lee SJ, Gottesman RF, Thomson R, Wang J, Yang Q, Niessen WJ, Lopez OL, Becker JT, Phan TG, Beare RJ, Arfanakis K, Fleischman D, Vernooij MW, Mazoyer B, Schmidt H, Srikanth V, Knopman DS, Jack CR, Amouyel P, Hofman A, DeCarli C, Tzourio C, van Duijn CM, Bennett DA, Schmidt R, Longstreth WT, Mosley TH, Fornage M, Launer LJ, Seshadri S, Ikram MA, Debette S. Association of Alzheimer's disease GWAS loci with MRI markers of brain aging. Neurobiol Aging 2015; 36:1765.e7-1765.e16. [PMID: 25670335 DOI: 10.1016/j.neurobiolaging.2014.12.028] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 12/22/2014] [Accepted: 12/26/2014] [Indexed: 10/24/2022]
Abstract
Whether novel risk variants of Alzheimer's disease (AD) identified through genome-wide association studies also influence magnetic resonance imaging-based intermediate phenotypes of AD in the general population is unclear. We studied association of 24 AD risk loci with intracranial volume, total brain volume, hippocampal volume (HV), white matter hyperintensity burden, and brain infarcts in a meta-analysis of genetic association studies from large population-based samples (N = 8175-11,550). In single-SNP based tests, AD risk allele of APOE (rs2075650) was associated with smaller HV (p = 0.0054) and CD33 (rs3865444) with smaller intracranial volume (p = 0.0058). In gene-based tests, there was associations of HLA-DRB1 with total brain volume (p = 0.0006) and BIN1 with HV (p = 0.00089). A weighted AD genetic risk score was associated with smaller HV (beta ± SE = -0.047 ± 0.013, p = 0.00041), even after excluding the APOE locus (p = 0.029). However, only association of AD genetic risk score with HV, including APOE, was significant after multiple testing correction (including number of independent phenotypes tested). These results suggest that novel AD genetic risk variants may contribute to structural brain aging in nondemented older community persons.
Collapse
Affiliation(s)
- Ganesh Chauhan
- INSERM U897, University of Bordeaux, Bordeaux, France; University of Bordeaux, Bordeaux, FranceINSERM U897, University of Bordeaux, Bordeaux, France
| | - Hieab H H Adams
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Radiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Galit Weinstein
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA; The Framingham Heart Study, Boston, MA, USA
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Anna Maria Töglhofer
- Institute of Molecular Biology and Biochemistry, Centre for Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Albert Vernon Smith
- Icelandic Heart Association, Iceland; Department of Medicine, University of Iceland, Reykjavik, Iceland
| | - Sven J van der Lee
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Rebecca F Gottesman
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, USA; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | - Russell Thomson
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania, Australia
| | - Jing Wang
- The Framingham Heart Study, Boston, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Qiong Yang
- The Framingham Heart Study, Boston, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Wiro J Niessen
- Department of Radiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - James T Becker
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Psychology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Thanh G Phan
- Stroke and Ageing Research Centre, Southern Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Richard J Beare
- Stroke and Ageing Research Centre, Southern Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia; Developmental Imaging Group, Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Konstantinos Arfanakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Debra Fleischman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Radiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Bernard Mazoyer
- Groupe d'Imagerie Neurofonctionnelle, UMR5296, CNRS, CEA, Université de Bordeaux, Bordeaux, France
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry, Centre for Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Velandai Srikanth
- Stroke and Ageing Research Centre, Southern Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia; Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania, Australia
| | - David S Knopman
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Philippe Amouyel
- Department of Epidemiology and Public Health, Pasteur Institute of Lille, Lille, France; INSERM, U744, Lille, France; Université Lille 2, Lille, France
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Charles DeCarli
- Department of Neurology, University of California at Davis, Davis, CA, USA
| | | | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands; Netherlands Consortium for Healthy Aging, Leiden, the Netherlands; Center for Medical Systems Biology, Leiden, the Netherlands
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Reinhold Schmidt
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University of Graz, Austria
| | - William T Longstreth
- Departments of Neurology and Epidemiology, University of Washington, Seattle, WA, USA
| | - Thomas H Mosley
- Department of Medicine-Geriatrics/Gerontology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Myriam Fornage
- The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA; The Framingham Heart Study, Boston, MA, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Radiology, Erasmus Medical Center, Rotterdam, the Netherlands; Netherlands Consortium for Healthy Aging, Leiden, the Netherlands
| | - Stephanie Debette
- INSERM U897, University of Bordeaux, Bordeaux, France; University of Bordeaux, Bordeaux, FranceINSERM U897, University of Bordeaux, Bordeaux, France; Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Department of Neurology, Bordeaux University Hospital, Bordeaux, France.
| |
Collapse
|
3104
|
Nelson PT, Wang WX, Partch AB, Monsell SE, Valladares O, Ellingson SR, Wilfred BR, Naj AC, Wang LS, Kukull WA, Fardo DW. Reassessment of risk genotypes (GRN, TMEM106B, and ABCC9 variants) associated with hippocampal sclerosis of aging pathology. J Neuropathol Exp Neurol 2015; 74:75-84. [PMID: 25470345 PMCID: PMC4270894 DOI: 10.1097/nen.0000000000000151] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hippocampal sclerosis of aging (HS-Aging) is a common high-morbidity neurodegenerative condition in elderly persons. To understand the risk factors for HS-Aging, we analyzed data from the Alzheimer's Disease Genetics Consortium and correlated the data with clinical and pathologic information from the National Alzheimer's Coordinating Center database. Overall, 268 research volunteers with HS-Aging and 2,957 controls were included; detailed neuropathologic data were available for all. The study focused on single-nucleotide polymorphisms previously associated with HS-Aging risk: rs5848 (GRN), rs1990622 (TMEM106B), and rs704180 (ABCC9). Analyses of a subsample that was not previously evaluated (51 HS-Aging cases and 561 controls) replicated the associations of previously identified HS-Aging risk alleles. To test for evidence of gene-gene interactions and genotype-phenotype relationships, pooled data were analyzed. The risk for HS-Aging diagnosis associated with these genetic polymorphisms was not secondary to an association with either Alzheimer disease or dementia with Lewy body neuropathologic changes. The presence of multiple risk genotypes was associated with a trend for additive risk for HS-Aging pathology. We conclude that multiple genes play important roles in HS-Aging, which is a distinctive neurodegenerative disease of aging.
Collapse
Affiliation(s)
- Peter T. Nelson
- University of Kentucky, Department of Pathology and Sanders-Brown Center on Aging, Lexington, Kentucky (PTN)
| | - Wang-Xia Wang
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, Kentucky (W-XW, BRW)
| | - Amanda B. Partch
- University of Pennsylvania, Department of Pathology and Laboratory Medicine, Philadelphia, Pennsylvania (ABP, OV, L-SW)
| | - Sarah E. Monsell
- University of Washington, National Alzheimer's Coordinating Center, Seattle, Washington (SEM)
| | - Otto Valladares
- University of Pennsylvania, Department of Pathology and Laboratory Medicine, Philadelphia, Pennsylvania (ABP, OV, L-SW)
| | - Sally R. Ellingson
- University of Kentucky, Division of Biomedical Informatics, College of Public Health, Lexington, Kentucky (SRE)
| | - Bernard R. Wilfred
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, Kentucky (W-XW, BRW)
| | - Adam C. Naj
- University of Pennsylvania, Department of Biostatistics and Epidemiology, Perelman School of Medicine, Philadelphia, Pennsylvania (ACN)
| | - Li-San Wang
- University of Pennsylvania, Department of Pathology and Laboratory Medicine, Philadelphia, Pennsylvania (ABP, OV, L-SW)
| | - Walter A. Kukull
- University of Washington, Department of Epidemiology, Seattle, Washington (WAK)
| | - David W. Fardo
- University of Kentucky, Department of Biostatistics and Sanders-Brown Center on Aging, Lexington, Kentucky (DWF)
| |
Collapse
|
3105
|
Darst BF, Koscik RL, Hermann BP, La Rue A, Sager MA, Johnson SC, Engelman CD. Heritability of cognitive traits among siblings with a parental history of Alzheimer's disease. J Alzheimers Dis 2015; 45:1149-55. [PMID: 25649654 PMCID: PMC4398625 DOI: 10.3233/jad-142658] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cognitive decline is one of the hallmark features of Alzheimer's disease, but many studies struggle to find strong associations between cognitive function and genetic variants. In order to identify which aspects of cognition are more likely to have a strong genetic component, we assessed the heritability of various cognitive functions related to Alzheimer's disease in 303 initially asymptomatic middle-aged adult siblings with a parental history of Alzheimer's disease from the Wisconsin Registry for Alzheimer's Prevention. Participants underwent extensive cognitive testing, and six cognitive factors were identified via factor analysis. Working Memory and Visual Learning & Memory had the highest heritability (52% and 41%, respectively). Inclusion of APOE allele counts did not notably change heritability estimates, indicating that there are likely additional genetic variants contributing to cognition. These findings suggest that future genetic studies should focus on the cognitive domains of Working Memory and Visual Learning & Memory.
Collapse
Affiliation(s)
- Burcu F. Darst
- Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Rebecca L. Koscik
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Bruce P. Hermann
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Asenath La Rue
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Mark A. Sager
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Alzheimer’s Diseases Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Memorial VA Hospital, Madison, WI, USA
| | - Corinne D. Engelman
- Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
3106
|
Castro-Chavira SA, Fernandez T, Nicolini H, Diaz-Cintra S, Prado-Alcala RA. Genetic markers in biological fluids for aging-related major neurocognitive disorder. Curr Alzheimer Res 2015; 12:200-9. [PMID: 25731625 PMCID: PMC4443795 DOI: 10.2174/1567205012666150302155138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 01/18/2015] [Accepted: 01/20/2015] [Indexed: 12/11/2022]
Abstract
Aging-related major neurocognitive disorder (NCD), formerly named dementia, comprises of the different acquired diseases whose primary deficit is impairment in cognitive functions such as complex attention, executive function, learning and memory, language, perceptual/motor skills, and social cognition, and that are related to specific brain regions and/or networks. According to its etiology, the most common subtypes of major NCDs are due to Alzheimer' s disease (AD), vascular disease (VaD), Lewy body disease (LBD), and frontotemporal lobar degeneration (FTLD). These pathologies are frequently present in mixed forms, i.e., AD plus VaD or AD plus LBD, thus diagnosed as due to multiple etiologies. In this paper, the definitions, criteria, pathologies, subtypes and genetic markers for the most common age-related major NCD subtypes are summarized. The current diagnostic criteria consider cognitive decline leading to major NCD or dementia as a progressive degenerative process with an underlying neuropathology that begins before the manifestation of symptoms. Biomarkers associated with this asymptomatic phase are being developed as accurate risk factor and biomarker assessments are fundamental to provide timely treatment since no treatments to prevent or cure NCD yet exist. Biological fluid assessment represents a safer, cheaper and less invasive method compared to contrast imaging studies to predict NCD appearance. Genetic factors particularly have a key role not only in predicting development of the disease but also the age of onset as well as the presentation of comorbidities that may contribute to the disease pathology and trigger synergistic mechanisms which may, in turn, accelerate the neurodegenerative process and its resultant behavioral and functional disorders.
Collapse
Affiliation(s)
| | | | | | | | - R A Prado-Alcala
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Campus UNAM Juriquilla, Universidad Nacional Autonoma de Mexico, Boulevard Universitario # 3001, Juriquilla, Queretaro. C. P. 76230, Queretaro, Mexico.
| |
Collapse
|
3107
|
Finelli D, Rollinson S, Harris J, Jones M, Richardson A, Gerhard A, Snowden J, Mann D, Pickering-Brown S. TREM2 analysis and increased risk of Alzheimer's disease. Neurobiol Aging 2015; 36:546.e9-13. [DOI: 10.1016/j.neurobiolaging.2014.08.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 08/01/2014] [Indexed: 11/24/2022]
|
3108
|
Ďurmanová V, Shawkatová I, Javor J, Párnická Z, Čopíková-Cundráková D, Turčáni P, Lisá I, Gajdošechová B, Buc M, Bucová M. VLA4 Gene Polymorphism and Susceptibility to Multiple Sclerosis in Slovaks. Folia Biol (Praha) 2015; 61:8-13. [PMID: 25958306 DOI: 10.14712/fb2015061010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Multiple sclerosis (MS) is an inflammatory autoimmune disease occurring in genetically sensitive individuals. As migration of immune cells into the CNS is facilitated by the Very Late Antigen 4 (VLA-4) integrin molecule, the VLA4 gene may be considered as a plausible candidate genetic risk factor for susceptibility to MS. Therefore, the objective of our study was to investigate the association between two genetic polymorphisms located in the VLA4 gene and the risk of multiple sclerosis. One hundred seventeen MS patients and 165 control subjects from Slovakia were genotyped for VLA4 gene SNP polymorphisms at positions 269 (C/A) and 3061 (A/G). The same study cohorts were also genotyped for the rs3135388 polymorphism tagging the HLA-DRB1*15:01 allele, which is a known genetic factor associated with susceptibility to develop MS in many populations. Our findings show for the first time that the rs3135388 polymorphism is a strong risk factor for MS in the Slovak population. Investigation of the VLA4 gene polymorphisms revealed a significantly higher frequency of the 3061AG genotype in MS patients compared to the controls (P ≤ 0.05). We suggest that the 3061AG polymorphic variant is an independent genetic risk factor for MS development in our population as it was significantly associated with this disease. The association was also confirmed after applying multivariate logistic-regression analysis adjusted for gender, age and HLA-DRB1*15:01 positivity as possible influencing factors.
Collapse
Affiliation(s)
- V Ďurmanová
- Institute of Immunology, School of Medicine, Comenius University in Bratislava, Slovakia
| | - I Shawkatová
- Institute of Immunology, School of Medicine, Comenius University in Bratislava, Slovakia
| | - J Javor
- Institute of Immunology, School of Medicine, Comenius University in Bratislava, Slovakia
| | - Z Párnická
- Institute of Immunology, School of Medicine, Comenius University in Bratislava, Slovakia
| | - D Čopíková-Cundráková
- 1st Department of Neurology, School of Medicine, Comenius University in Bratislava and University Hospital, Bratislava, Slovakia
| | - P Turčáni
- 1st Department of Neurology, School of Medicine, Comenius University in Bratislava and University Hospital, Bratislava, Slovakia
| | - I Lisá
- 2nd Department of Neurology, School of Medicine, Comenius University in Bratislava and University Hospital, Bratislava, Slovakia
| | - B Gajdošechová
- Institute of Immunology, School of Medicine, Comenius University in Bratislava, Slovakia
| | - M Buc
- Institute of Immunology, School of Medicine, Comenius University in Bratislava, Slovakia
| | - M Bucová
- Institute of Immunology, School of Medicine, Comenius University in Bratislava, Slovakia
| |
Collapse
|
3109
|
Karch CM, Goate AM. Alzheimer's disease risk genes and mechanisms of disease pathogenesis. Biol Psychiatry 2015; 77:43-51. [PMID: 24951455 PMCID: PMC4234692 DOI: 10.1016/j.biopsych.2014.05.006] [Citation(s) in RCA: 948] [Impact Index Per Article: 94.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 04/30/2014] [Accepted: 05/05/2014] [Indexed: 01/18/2023]
Abstract
We review the genetic risk factors for late-onset Alzheimer's disease (AD) and their role in AD pathogenesis. More recent advances in understanding of the human genome-technologic advances in methods to analyze millions of polymorphisms in thousands of subjects-have revealed new genes associated with AD risk, including ABCA7, BIN1, CASS4, CD33, CD2AP, CELF1, CLU, CR1, DSG2, EPHA1, FERMT2, HLA-DRB5-DBR1, INPP5D, MS4A, MEF2C, NME8, PICALM, PTK2B, SLC24H4-RIN3, SORL1, and ZCWPW1. Emerging technologies to analyze the entire genome in large data sets have also revealed coding variants that increase AD risk: PLD3 and TREM2. We review the relationship between these AD risk genes and the cellular and neuropathologic features of AD. Understanding the mechanisms underlying the association of these genes with risk for disease will provide the most meaningful targets for therapeutic development to date.
Collapse
Affiliation(s)
| | - Alison M. Goate
- Corresponding author Contact information: Department of Psychiatry, Washington University School of Medicine, 425 S. Euclid Ave, Campus Box 8134, St. Louis, MO 63110, phone: 314-362-8691, fax: 314-747-2983,
| |
Collapse
|
3110
|
Abstract
Localized protein synthesis is a mechanism by which morphologically polarized cells react in a spatially confined and temporally acute manner to changes in their environment. During the development of the nervous system intra-axonal protein synthesis is crucial for the establishment of neuronal connections. In contrast, mature axons have long been considered as translationally inactive but upon nerve injury or under neurodegenerative conditions specific subsets of mRNAs are recruited into axons and locally translated. Intra-axonally synthesized proteins can have pathogenic or restorative and regenerative functions, and thus targeting the axonal translatome might have therapeutic value, for example in the treatment of spinal cord injury or Alzheimer's disease. In the case of Alzheimer's disease the local synthesis of the stress response transcription factor activating transcription factor 4 mediates the long-range retrograde spread of pathology across the brain, and inhibition of local Atf4 translation downstream of the integrated stress response might interfere with this spread. Several molecular tools and approaches have been developed to target specifically the axonal translatome by either overexposing proteins locally in axons or, conversely, knocking down selectively axonally localized mRNAs. Many questions about axonal translation remain to be answered, especially with regard to the mechanisms establishing specificity but, nevertheless, targeting the axonal translatome is a promising novel avenue to pursue in the development for future therapies for various neurological conditions.
Collapse
Affiliation(s)
- Jimena Baleriola
- />The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, 650 W. 168th St., New York, NY USA
| | - Ulrich Hengst
- />The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, 650 W. 168th St., New York, NY USA
- />Department of Pathology and Cell Biology, Columbia University, 650 W. 168th St., New York, NY USA
| |
Collapse
|
3111
|
Chaudhry M, Wang X, Bamne MN, Hasnain S, Demirci F, Lopez OL, Kamboh MI. Genetic variation in imprinted genes is associated with risk of late-onset Alzheimer's disease. J Alzheimers Dis 2015; 44:989-94. [PMID: 25391383 PMCID: PMC4324355 DOI: 10.3233/jad-142106] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epigenetic changes including genomic imprinting may affect risk of late-onset Alzheimer's disease (LOAD). There are >100 known imprinted genes and most of them are expressed in human brain. In this study, we examined the association of single nucleotide polymorphisms (SNPs) in 93 imprinted genes with LOAD risk in 1291 LOAD cases and 958 cognitively normal controls. We performed single-site, gene-based, and haplotype analyses. Single-site analysis showed 14 significant associations at p < 0.01. The most significant SNP (rs11770199; p = 0.0003) in single-site analysis was located on chromosome 7 in the GRB10 gene. Gene-based analyses revealed four significant associations in the WT1, ZC3H12C, DLGAP2, and GPR1 genes at p < 0.05. The haplotype analysis also revealed significant associations with three genes (ZC3H12C, DLGAP2, and GPR1). These findings suggest a possible role of imprinted genes in AD pathogenesis that show specific expression in the brain.
Collapse
Affiliation(s)
- Mamoonah Chaudhry
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan
| | - Xingbin Wang
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mikhil N. Bamne
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shahida Hasnain
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan
- The Women University Multan, Multan, Pakistan
| | - F.Yesim Demirci
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Alzheimer’s Disease Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - M. Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Alzheimer’s Disease Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
3112
|
Bennett DA, Yu L, Yang J, Srivastava GP, Aubin C, De Jager PL. Epigenomics of Alzheimer's disease. Transl Res 2015; 165:200-20. [PMID: 24905038 PMCID: PMC4233194 DOI: 10.1016/j.trsl.2014.05.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 05/08/2014] [Accepted: 05/10/2014] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a large and growing public health problem. It is characterized by the accumulation of amyloid β peptides and abnormally phosphorylated tau proteins that are associated with cognitive decline and dementia. Much has been learned about the genomics of AD from linkage analyses and, more recently, genome-wide association studies. Several but not all aspects of the genomic landscape are involved in amyloid β metabolism. The moderate concordance of disease among twins suggests other factors, potentially epigenomic factors, are related to AD. We are at the earliest stages of examining the relation of the epigenome to the clinical and pathologic phenotypes that characterize AD. Our literature review suggests that there is some evidence of age-related changes in human brain methylation. Unfortunately, studies of AD have been relatively small with limited coverage of methylation sites and microRNA, let alone other epigenomic marks. We are in the midst of 2 large studies of human brains including coverage of more than 420,000 autosomal cytosine-guanine dinucleotides with the Illumina Infinium HumanMethylation450 BeadArray, and histone acetylation with chromatin immunoprecipitation sequencing. We present descriptive data to help inform other researchers what to expect from these approaches to better design and power their studies. We then discuss future directions to inform on the epigenomic architecture of AD.
Collapse
Affiliation(s)
- David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Ill.
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Ill
| | - Jingyun Yang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Ill
| | - Gyan P Srivastava
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Program in Medical and Population Genetics, Broad Institute, Cambridge, Mass
| | - Cristin Aubin
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Program in Medical and Population Genetics, Broad Institute, Cambridge, Mass
| | - Philip L De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Program in Medical and Population Genetics, Broad Institute, Cambridge, Mass
| |
Collapse
|
3113
|
Exome and Whole Genome Sequencing in Aging and Longevity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 847:127-39. [DOI: 10.1007/978-1-4939-2404-2_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
3114
|
Pouget JG, Gonçalves VF, Nurmi EL, P Laughlin C, Mallya KS, McCracken JT, Aman MG, McDougle CJ, Scahill L, Misener VL, Tiwari AK, Zai CC, Brandl EJ, Felsky D, Leung AQ, Lieberman JA, Meltzer HY, Potkin SG, Niedling C, Steimer W, Leucht S, Knight J, Müller DJ, Kennedy JL. Investigation of TSPO variants in schizophrenia and antipsychotic treatment outcomes. Pharmacogenomics 2015; 16:5-22. [DOI: 10.2217/pgs.14.158] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: TSPO is a neuroinflammatory biomarker and emerging therapeutic target in psychiatric disorders. We evaluated whether TSPO polymorphisms contribute to interindividual variability in schizophrenia, antipsychotic efficacy and antipsychotic-induced weight gain. Patients & methods: We analyzed TSPO polymorphisms in 670 schizophrenia cases and 775 healthy controls. Gene–gene interactions between TSPO and other mitochondrial membrane protein-encoding genes (VDAC1 and ANT1) were explored. Positive findings were evaluated in two independent samples (Munich, n = 300; RUPP, n = 119). Results: TSPO rs6971 was independently associated with antipsychotic-induced weight gain in the discovery (puncor = 0.04) and RUPP samples (p = 3.00 × 10-3), and interacted with ANT1 rs10024068 in the discovery (p = 1.15 × 10-3) and RUPP samples (p = 2.76 × 10-4). Conclusion: Our findings highlight TSPO as a candidate for future investigations of antipsychotic-induced weight gain, and support the involvement of mitochondrial membrane components in this serious treatment side effect. Original submitted 20 August 2014; Revision submitted 3 November 2014
Collapse
Affiliation(s)
- Jennie G Pouget
- • Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
- • Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- • Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Vanessa F Gonçalves
- • Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
- • Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Erika L Nurmi
- • Department of Psychiatry & Biobehavioral Sciences, UCLA Semel Institute, Los Angeles, CA, USA
| | - Christopher P Laughlin
- • Department of Psychiatry & Biobehavioral Sciences, UCLA Semel Institute, Los Angeles, CA, USA
| | - Karyn S Mallya
- • Department of Psychiatry & Biobehavioral Sciences, UCLA Semel Institute, Los Angeles, CA, USA
| | - James T McCracken
- • Department of Psychiatry & Biobehavioral Sciences, UCLA Semel Institute, Los Angeles, CA, USA
| | - Michael G Aman
- • Department of Psychiatry, Ohio State University, OH, USA
| | | | | | - Virginia L Misener
- • Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
| | - Arun K Tiwari
- • Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
| | - Clement C Zai
- • Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
- • Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Eva J Brandl
- • Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
- • Department of Psychiatry & Psychotherapy, Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Daniel Felsky
- • Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
- • Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- • Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Amy Q Leung
- • Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
| | - Jeffrey A Lieberman
- • Department of Psychiatry, College of Physicians & Surgeons, Columbia University, NY, USA
- • New York State Psychiatric Institute, New York, NY, USA
| | - Herbert Y Meltzer
- • Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Steven G Potkin
- • Brain Imaging Centre, Irvine Hall, University of California, Irvine, CA, USA
| | - Charlotte Niedling
- • Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, TU-München, Germany
| | - Werner Steimer
- • Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, TU-München, Germany
| | - Stefan Leucht
- • Psychiatrische Klinik und Poliklinik, Klinikum rechts der Isar, TU-München, Germany
| | - Jo Knight
- • Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
- • Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- • Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- • Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Daniel J Müller
- • Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
- • Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- • Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- • Pharmacogenetics Research Clinic, Centre for Addiction & Mental Health, Toronto, ON, Canada
| | - James L Kennedy
- • Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
- • Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- • Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3115
|
Abstract
The small, calcium-sensor protein, calmodulin, is ubiquitously expressed and central to cell function in all cell types. Here the literature linking calmodulin to Alzheimer's disease is reviewed. Several experimentally-verified calmodulin-binding proteins are involved in the formation of amyloid-β plaques including amyloid-β protein precursor, β-secretase, presenilin-1, and ADAM10. Many others possess potential calmodulin-binding domains that remain to be verified. Three calmodulin binding proteins are associated with the formation of neurofibrillary tangles: two kinases (CaMKII, CDK5) and one protein phosphatase (PP2B or calcineurin). Many of the genes recently identified by genome wide association studies and other studies encode proteins that contain putative calmodulin-binding domains but only a couple (e.g., APOE, BIN1) have been experimentally confirmed as calmodulin binding proteins. At least two receptors involved in calcium metabolism and linked to Alzheimer's disease (mAchR; NMDAR) have also been identified as calmodulin-binding proteins. In addition to this, many proteins that are involved in other cellular events intimately associated with Alzheimer's disease including calcium channel function, cholesterol metabolism, neuroinflammation, endocytosis, cell cycle events, and apoptosis have been tentatively or experimentally verified as calmodulin binding proteins. The use of calmodulin as a potential biomarker and as a therapeutic target is discussed.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto at Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Kristeen Eshak
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Michael A. Myre
- Center for Human Genetic Research, Richard B. Simches Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3116
|
Płóciennik A, Prendecki M, Zuba E, Siudzinski M, Dorszewska J. Activated Caspase-3 and Neurodegeneration and Synaptic Plasticity in Alzheimer’s Disease. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/aad.2015.43007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
3117
|
Stepanov VA, Bocharova AV, Marusin AV, Zhukova NG, Alifirova VM, Zhukova IA. Replicative association analysis of genetic markers of cognitive traits with Alzheimer’s disease in the Russian population. Mol Biol 2014. [DOI: 10.1134/s0026893314060168] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
3118
|
Monastero R, Caruso C, Vasto S. Alzheimer's disease and infections, where we stand and where we go. IMMUNITY & AGEING 2014; 11:26. [PMID: 25535510 PMCID: PMC4273443 DOI: 10.1186/s12979-014-0026-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 12/08/2014] [Indexed: 11/29/2022]
Affiliation(s)
- Roberto Monastero
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Calogero Caruso
- Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
| | - Sonya Vasto
- National Center for Research, Institute of Biomedicine and Molecular Immunology, Palermo, Italy ; Department of Science and Biological, Chemical and Pharmaceutical Technologies, Institute of Biomedicine and Molecular Immunology, Palermo, Italy
| |
Collapse
|
3119
|
Ramirez A, van der Flier WM, Herold C, Ramonet D, Heilmann S, Lewczuk P, Popp J, Lacour A, Drichel D, Louwersheimer E, Kummer MP, Cruchaga C, Hoffmann P, Teunissen C, Holstege H, Kornhuber J, Peters O, Naj AC, Chouraki V, Bellenguez C, Gerrish A, International Genomics of Alzheimer's Project (IGAP), Alzheimer's Disease Neuroimaging Initiative (ADNI), Heun R, Frölich L, Hüll M, Buscemi L, Herms S, Kölsch H, Scheltens P, Breteler MM, Rüther E, Wiltfang J, Goate A, Jessen F, Maier W, Heneka MT, Becker T, Nöthen MM. SUCLG2 identified as both a determinator of CSF Aβ1-42 levels and an attenuator of cognitive decline in Alzheimer's disease. Hum Mol Genet 2014; 23:6644-58. [PMID: 25027320 PMCID: PMC4240204 DOI: 10.1093/hmg/ddu372] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 06/27/2014] [Accepted: 07/09/2014] [Indexed: 01/17/2023] Open
Abstract
Cerebrospinal fluid amyloid-beta 1-42 (Aβ1-42) and phosphorylated Tau at position 181 (pTau181) are biomarkers of Alzheimer's disease (AD). We performed an analysis and meta-analysis of genome-wide association study data on Aβ1-42 and pTau181 in AD dementia patients followed by independent replication. An association was found between Aβ1-42 level and a single-nucleotide polymorphism in SUCLG2 (rs62256378) (P = 2.5×10(-12)). An interaction between APOE genotype and rs62256378 was detected (P = 9.5 × 10(-5)), with the strongest effect being observed in APOE-ε4 noncarriers. Clinically, rs62256378 was associated with rate of cognitive decline in AD dementia patients (P = 3.1 × 10(-3)). Functional microglia experiments showed that SUCLG2 was involved in clearance of Aβ1-42.
Collapse
Affiliation(s)
- Alfredo Ramirez
- Department of Psychiatry and Psychotherapy, Institute of Human Genetics,
| | - Wiesje M van der Flier
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, 1081 HZ, Amsterdam, The Netherlands, Department of Epidemiology & Biostatistics, VU University Medical Center, 1007 MB, Amsterdam, The Netherlands
| | - Christine Herold
- German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | | | - Stefanie Heilmann
- Institute of Human Genetics, Department of Genomics, Life & Brain Center
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | | | - André Lacour
- German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Dmitriy Drichel
- German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Eva Louwersheimer
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, 1081 HZ, Amsterdam, The Netherlands, Department of Epidemiology & Biostatistics, VU University Medical Center, 1007 MB, Amsterdam, The Netherlands
| | - Markus P Kummer
- Clinical Neuroscience Unit, Department of Neurology, German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Carlos Cruchaga
- Department of Psychiatry, Hope Center for Neurological Disorders, School of Medicine
| | - Per Hoffmann
- Institute of Human Genetics, Department of Genomics, Life & Brain Center, Division of Medical Genetics, University Hospital and Department of Biomedicine, University of Basel, CH-4058, Basel, Switzerland
| | - Charlotte Teunissen
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, 1081 HZ, Amsterdam, The Netherlands, Department of Epidemiology & Biostatistics, VU University Medical Center, 1007 MB, Amsterdam, The Netherlands
| | - Henne Holstege
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, 1081 HZ, Amsterdam, The Netherlands, Department of Epidemiology & Biostatistics, VU University Medical Center, 1007 MB, Amsterdam, The Netherlands
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Oliver Peters
- Department of Psychiatry, Charité, 14050, Berlin, Germany
| | - Adam C Naj
- Center for Clinical Epidemiology & Biostatistics, University of Pennsylvania, PA 19104, Philadelphia, USA
| | - Vincent Chouraki
- Department of Neurology, Boston University School of Medicine, MA 02118, Boston, USA, The Framingham Heart Study, MA 01702, Framingham, USA
| | - Céline Bellenguez
- Inserm, U744, Lille 59000, France, Université Lille 2, Lille 59000, France, Institut Pasteur de Lille, Lille 59000, France
| | - Amy Gerrish
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | | | | | | | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159, Mannheim, Germany
| | - Michael Hüll
- Centre for Geriatric Medicine and Section of Gerontopsychiatry and Neuropsychology, Medical School, University of Freiburg, 79106, Freiburg, Germany
| | - Lara Buscemi
- Department of Fundamental Neurosciences, UNIL, 1005 Lausanne, Switzerland and
| | - Stefan Herms
- Institute of Human Genetics, Department of Genomics, Life & Brain Center, Division of Medical Genetics, University Hospital and Department of Biomedicine, University of Basel, CH-4058, Basel, Switzerland
| | | | - Philip Scheltens
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, 1081 HZ, Amsterdam, The Netherlands, Department of Epidemiology & Biostatistics, VU University Medical Center, 1007 MB, Amsterdam, The Netherlands
| | - Monique M Breteler
- German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Eckart Rüther
- Department of Psychiatry and Psychotherapy, University of Göttingen, 37075 Göttingen, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University of Göttingen, 37075 Göttingen, Germany
| | - Alison Goate
- Department of Psychiatry, Department of Genetics, Washington University, St. Louis, MO 63110, USA
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Wolfgang Maier
- Department of Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Michael T Heneka
- Clinical Neuroscience Unit, Department of Neurology, German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Tim Becker
- Institute for Medical Biometry, Informatics, and Epidemiology, University of Bonn, 53127, Bonn, Germany, German Center for Neurodegenerative Diseases (DZNE), 53175, Bonn, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, Department of Genomics, Life & Brain Center
| |
Collapse
|
3120
|
Jaffe AE, Shin J, Collado-Torres L, Leek JT, Tao R, Li C, Gao Y, Jia Y, Maher BJ, Hyde TM, Kleinman JE, Weinberger DR. Developmental regulation of human cortex transcription and its clinical relevance at single base resolution. Nat Neurosci 2014; 18:154-161. [PMID: 25501035 PMCID: PMC4281298 DOI: 10.1038/nn.3898] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 11/14/2014] [Indexed: 02/08/2023]
Abstract
Transcriptome analysis of human brain provides fundamental insight about development and disease, but largely relies on existing annotation. We sequenced transcriptomes of 72 prefrontal cortex samples across six life stages, and identified 50,650 differentially expression regions (DERs) associated with developmental and aging, agnostic of annotation. While many DERs annotated to non-exonic sequence (41.1%), most were similarly regulated in cytosolic mRNA extracted from independent samples. The DERs were developmentally conserved across 16 brain regions and within the developing mouse cortex, and were expressed in diverse cell and tissue types. The DERs were further enriched for active chromatin marks and clinical risk for neurodevelopmental disorders like schizophrenia. Lastly, we demonstrate quantitatively that these DERs associate with a changing neuronal phenotype related to differentiation and maturation. These data highlight conserved molecular signatures of transcriptional dynamics across brain development, some potential clinical relevance and the incomplete annotation of the human brain transcriptome.
Collapse
Affiliation(s)
- Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore MD 21205.,Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore MD 21205.,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore MD 21205
| | - Jooheon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore MD 21205
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore MD 21205.,Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore MD 21205
| | - Jeffrey T Leek
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore MD 21205.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore MD 21205
| | - Ran Tao
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore MD 21205
| | - Chao Li
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore MD 21205
| | - Yuan Gao
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore MD 21205
| | - Yankai Jia
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore MD 21205
| | - Brady J Maher
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore MD 21205.,Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore MD 21205.,Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore MD 21205.,Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore MD 21205.,Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205.,Department of Biological Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore MD 21205
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore MD 21205.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore MD 21205.,Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore MD 21205.,Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| |
Collapse
|
3121
|
Freude K, Pires C, Hyttel P, Hall VJ. Induced Pluripotent Stem Cells Derived from Alzheimer's Disease Patients: The Promise, the Hope and the Path Ahead. J Clin Med 2014; 3:1402-36. [PMID: 26237610 PMCID: PMC4470192 DOI: 10.3390/jcm3041402] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 11/12/2014] [Accepted: 11/14/2014] [Indexed: 02/07/2023] Open
Abstract
The future hope of generated induced pluripotent stem cells (iPS cells) from Alzheimer’s disease patients is multifold. Firstly, they may help to uncover novel mechanisms of the disease, which could lead to the development of new and unprecedented drugs for patients and secondly, they could also be directly used for screening and testing of potential new compounds for drug discovery. In addition, in the case of familial known mutations, these cells could be targeted by use of advanced gene-editing techniques to correct the mutation and be used for future cell transplantation therapies. This review summarizes the work so far in regards to production and characterization of iPS cell lines from both sporadic and familial Alzheimer’s patients and from other iPS cell lines that may help to model the disease. It provides a detailed comparison between published reports and states the present hurdles we face with this new technology. The promise of new gene-editing techniques and accelerated aging models also aim to move this field further by providing better control cell lines for comparisons and potentially better phenotypes, respectively.
Collapse
Affiliation(s)
- Kristine Freude
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegaardsvej 7, Frederiksberg C DK-1870, Denmark.
| | - Carlota Pires
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegaardsvej 7, Frederiksberg C DK-1870, Denmark.
| | - Poul Hyttel
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegaardsvej 7, Frederiksberg C DK-1870, Denmark.
| | - Vanessa Jane Hall
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegaardsvej 7, Frederiksberg C DK-1870, Denmark.
| |
Collapse
|
3122
|
Influence of genetic variants in SORL1 gene on the manifestation of Alzheimer's disease. Neurobiol Aging 2014; 36:1605.e13-20. [PMID: 25659857 DOI: 10.1016/j.neurobiolaging.2014.12.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/12/2014] [Accepted: 12/05/2014] [Indexed: 11/21/2022]
Abstract
We studied the association of SORL1 single-nucleotide polymorphisms genotypes with measures of pathology in patients with probable Alzheimer's disease (AD) using an endophenotype approach. We included (1) 133 patients from the German Dementia Competence Network (71 ± 8 years; 50% females; Mini Mental State Examination [MMSE], 24 ± 3); (2) 83 patients from the Alzheimer's Disease Neuroimaging Initiative (75 ± 8 years; 45% females; MMSE, 24 ± 2); and (3) 452 patients from the Amsterdam Dementia Cohort 66 ± 8 years; 47% females; MMSE, 20 ± 5). As endophenotype markers we used cognitive tests, cerebrospinal fluid (CSF) biomarkers amyloid-beta, total tau (tau), tau phosphorylated at threonine 181, and hippocampal atrophy. We measured 19 SORL1 SNP alleles. Genotype-endophenotype associations were determined by linear regression analyses. There was an association between rs2070045-G allele and increased CSF-tau and more hippocampal atrophy. Additionally, haplotype-based analyses revealed an association between haplotype rs11218340-A/rs3824966-G/rs3824968-A and higher CSF-tau and CSF-tau phosphorylated at threonine 181. In conclusion, we found that SORL1 SNP rs2070045-G allele was related to CSF-tau and hippocampal atrophy, 2 endophenotype markers of AD, suggesting that SORL1 may be implicated in the downstream pathology in AD.
Collapse
|
3123
|
Johansson JU, Woodling NS, Wang Q, Panchal M, Liang X, Trueba-Saiz A, Brown HD, Mhatre SD, Loui T, Andreasson KI. Prostaglandin signaling suppresses beneficial microglial function in Alzheimer's disease models. J Clin Invest 2014; 125:350-64. [PMID: 25485684 DOI: 10.1172/jci77487] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 10/30/2014] [Indexed: 12/25/2022] Open
Abstract
Microglia, the innate immune cells of the CNS, perform critical inflammatory and noninflammatory functions that maintain normal neural function. For example, microglia clear misfolded proteins, elaborate trophic factors, and regulate and terminate toxic inflammation. In Alzheimer's disease (AD), however, beneficial microglial functions become impaired, accelerating synaptic and neuronal loss. Better understanding of the molecular mechanisms that contribute to microglial dysfunction is an important objective for identifying potential strategies to delay progression to AD. The inflammatory cyclooxygenase/prostaglandin E2 (COX/PGE2) pathway has been implicated in preclinical AD development, both in human epidemiology studies and in transgenic rodent models of AD. Here, we evaluated murine models that recapitulate microglial responses to Aβ peptides and determined that microglia-specific deletion of the gene encoding the PGE2 receptor EP2 restores microglial chemotaxis and Aβ clearance, suppresses toxic inflammation, increases cytoprotective insulin-like growth factor 1 (IGF1) signaling, and prevents synaptic injury and memory deficits. Our findings indicate that EP2 signaling suppresses beneficial microglia functions that falter during AD development and suggest that inhibition of the COX/PGE2/EP2 immune pathway has potential as a strategy to restore healthy microglial function and prevent progression to AD.
Collapse
|
3124
|
Association between NME8 locus polymorphism and cognitive decline, cerebrospinal fluid and neuroimaging biomarkers in Alzheimer's disease. PLoS One 2014; 9:e114777. [PMID: 25486118 PMCID: PMC4259473 DOI: 10.1371/journal.pone.0114777] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 11/13/2014] [Indexed: 12/24/2022] Open
Abstract
Recently, a large meta-analysis of five genome wide association studies (GWAS) identified a novel locus (rs2718058) adjacent to NME8 that played a preventive role in Alzheimer's disease (AD). However, this link between the single nucleotide polymorphism (SNP) rs2718058 and the pathology of AD have not been mentioned yet. Therefore, this study assessed the strength of association between the NME8 rs2718058 genotypes and AD-related measures including the cerebrospinal fluid (CSF) amyloid beta, tau, P-tau concentrations, neuroimaging biomarkers and cognitive performance, in a large cohort from Alzheimer's Disease Neuroimaging Initiative (ADNI) database. We used information of a total of 719 individuals, including 211 normal cognition (NC), 346 mild cognitive impairment (MCI) and 162 AD. Although we didn't observe a positive relationship between rs2718058 and AD, it was significantly associated with several AD related endophenotypes. Among the normal cognitively normal participants, the minor allele G carriers showed significantly associated with higher CDRSB score than A allele carriers (P = 0.021). Occipital gyrus atrophy were significantly associated with NME8 genotype status (P = 0.002), with A allele carriers has more atrophy than the minor allele G carriers in AD patients; lateral ventricle (both right and left) cerebral metabolic rate for glucose (CMRgl) were significantly associated with NME8 genotype (P<0.05), with GA genotype had higher metabolism than GG and AA genotypes in MCI group; the atrophic right hippocampus in 18 months is significantly different between the three group, with GG and AA genotypes had more hippocampus atrophy than GA genotypes in the whole group. Together, our results are consistent with the direction of previous research, suggesting that NME8 rs2718058 appears to play a role in lowering the brain neurodegeneration.
Collapse
|
3125
|
Yajima R, Tokutake T, Koyama A, Kasuga K, Tezuka T, Nishizawa M, Ikeuchi T. ApoE-isoform-dependent cellular uptake of amyloid-β is mediated by lipoprotein receptor LR11/SorLA. Biochem Biophys Res Commun 2014; 456:482-8. [PMID: 25482438 DOI: 10.1016/j.bbrc.2014.11.111] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 11/28/2014] [Indexed: 11/30/2022]
Abstract
The formation of senile plaques composed of β-amyloid (Aβ) in the brain is likely the initial event in Alzheimer's disease (AD). Possession of the APOE ε4 allele, the strong genetic factor for AD, facilitates the Aβ deposition from the presymptomatic stage of AD in a gene-dosage-dependent manner. However, the precise mechanism by which apoE isoforms differentially induce the AD pathology is largely unknown. LR11/SorLA is a type I membrane protein that functions as the neuronal lipoprotein endocytic receptor of apoE and the sorting receptor of the amyloid precursor protein (APP) to regulate amyloidogenesis. Recently, LR11/SorLA has been reported to be involved in the lysosomal targeting of extracellular amyloid-β (Aβ) through the binding of Aβ to the vacuolar protein sorting 10 (VPS10) protein domain of LR11/SorLA. Here, we attempted to examine the human-apoE-isoform-dependent effect on the cellular uptake of Aβ through the formation of a complex between an apoE isoform and LR11/SorLA. Cell culture experiments using Neuro2a cells revealed that the cellular uptake of secreted apoE3 and apoE4 was enhanced by the overexpression of LR11/SorLA. In contrast, the cellular uptake of apoE2 was not affected by the expression of LR11/SorLA. Co-immunoprecipitation assay revealed that apoE-isoform-dependent differences were observed in the formation of an apoE-LR11 complex (apoE4>apoE3>apoE2). ApoE-isoform-dependent differences in cellular uptake of FAM-labeled Aβ were further investigated by coculture assay in which donor cells secrete one of the apoE isoforms and recipient cells express FL-LR11. The cellular uptake of extracellular Aβ into the recipient cells was most prominently accentuated when cocultured with the donor cells secreting apoE4 in the medium, followed by apoE3 and apoE2. Taken together, our results provide evidence for the mechanism whereby human-apoE-isoform-dependent differences modulate the cellular uptake of Aβ mediated by LR11/SorLA.
Collapse
Affiliation(s)
- Ryuji Yajima
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takayoshi Tokutake
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akihide Koyama
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan
| | - Kensaku Kasuga
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan; Center for Transdisciplinary Research, Niigata University, Niigata, Japan; Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Toshiyuki Tezuka
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan; Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masatoyo Nishizawa
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan.
| |
Collapse
|
3126
|
Validating GWAS-Identified Risk Loci for Alzheimer’s Disease in Han Chinese Populations. Mol Neurobiol 2014; 53:379-390. [DOI: 10.1007/s12035-014-9015-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 11/18/2014] [Indexed: 10/24/2022]
|
3127
|
Wetzel-Smith MK, Hunkapiller J, Bhangale TR, Srinivasan K, Maloney JA, Atwal JK, Sa SM, Yaylaoglu MB, Foreman O, Ortmann W, Rathore N, Hansen DV, Tessier-Lavigne M, Mayeux R, Pericak-Vance M, Haines J, Farrer LA, Schellenberg GD, Goate A, Behrens TW, Cruchaga C, Watts RJ, Graham RR. A rare mutation in UNC5C predisposes to late-onset Alzheimer's disease and increases neuronal cell death. Nat Med 2014; 20:1452-7. [PMID: 25419706 PMCID: PMC4301587 DOI: 10.1038/nm.3736] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 09/23/2014] [Indexed: 12/23/2022]
Abstract
We have identified a rare coding mutation, T835M (rs137875858), in the UNC5C netrin receptor gene that segregated with disease in an autosomal dominant pattern in two families enriched for late-onset Alzheimer's disease and that was associated with disease across four large case-control cohorts (odds ratio = 2.15, Pmeta = 0.0095). T835M alters a conserved residue in the hinge region of UNC5C, and in vitro studies demonstrate that this mutation leads to increased cell death in human HEK293T cells and in rodent neurons. Furthermore, neurons expressing T835M UNC5C are more susceptible to cell death from multiple neurotoxic stimuli, including β-amyloid (Aβ), glutamate and staurosporine. On the basis of these data and the enriched hippocampal expression of UNC5C in the adult nervous system, we propose that one possible mechanism in which T835M UNC5C contributes to the risk of Alzheimer's disease is by increasing susceptibility to neuronal cell death, particularly in vulnerable regions of the Alzheimer's disease brain.
Collapse
Affiliation(s)
| | - Julie Hunkapiller
- Department of Human Genetics, Genentech, South San Francisco, California, USA
| | - Tushar R Bhangale
- Department of Bioinformatics and Computational Biology, Genentech, South San Francisco, California, USA
| | | | - Janice A Maloney
- Department of Neuroscience, Genentech, South San Francisco, California, USA
| | - Jasvinder K Atwal
- Department of Neuroscience, Genentech, South San Francisco, California, USA
| | - Susan M Sa
- Department of Pathology, Genentech, South San Francisco, California, USA
| | - Murat B Yaylaoglu
- Department of Pathology, Genentech, South San Francisco, California, USA
| | - Oded Foreman
- Department of Pathology, Genentech, South San Francisco, California, USA
| | - Ward Ortmann
- Department of Human Genetics, Genentech, South San Francisco, California, USA
| | - Nisha Rathore
- Department of Human Genetics, Genentech, South San Francisco, California, USA
| | - David V Hansen
- Department of Neuroscience, Genentech, South San Francisco, California, USA
| | - Marc Tessier-Lavigne
- Laboratory of Brain Development and Repair, Rockefeller University, New York, New York, USA
| | - Richard Mayeux
- 1] Department of Neurology, Taub Institute on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA. [2] Gertrude H. Sergievsky Center, Columbia University, New York, New York, USA
| | - Margaret Pericak-Vance
- 1] The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, USA. [2] Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, Florida, USA
| | - Jonathan Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Lindsay A Farrer
- 1] Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA. [2] Department of Neurology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA. [3] Department of Ophthalmology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA. [4] Department of Epidemiology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA. [5] Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Alison Goate
- 1] Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA. [2] Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA. [3] Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Timothy W Behrens
- Department of Human Genetics, Genentech, South San Francisco, California, USA
| | - Carlos Cruchaga
- 1] Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA. [2] Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ryan J Watts
- Department of Neuroscience, Genentech, South San Francisco, California, USA
| | - Robert R Graham
- Department of Human Genetics, Genentech, South San Francisco, California, USA
| |
Collapse
|
3128
|
Alzheimer’s Disease Genetics. Curr Behav Neurosci Rep 2014. [DOI: 10.1007/s40473-014-0026-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
3129
|
Ahmed RM, Paterson RW, Warren JD, Zetterberg H, O'Brien JT, Fox NC, Halliday GM, Schott JM. Biomarkers in dementia: clinical utility and new directions. J Neurol Neurosurg Psychiatry 2014; 85:1426-34. [PMID: 25261571 PMCID: PMC4335455 DOI: 10.1136/jnnp-2014-307662] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 08/27/2014] [Accepted: 09/03/2014] [Indexed: 12/12/2022]
Abstract
Imaging, cerebrospinal fluid (CSF) and blood-based biomarkers have the potential to improve the accuracy by which specific causes of dementia can be diagnosed in vivo, provide insights into the underlying pathophysiology, and may be used as inclusion criteria and outcome measures for clinical trials. While a number of imaging and CSF biomarkers are currently used for each of these purposes, this is an evolving field, with numerous potential biomarkers in varying stages of research and development. We review the currently available biomarkers for the three most common forms of neurodegenerative dementia, and give an overview of research techniques that may in due course make their way into the clinic.
Collapse
Affiliation(s)
- R M Ahmed
- Neuroscience Research Australia and the University of NSW, Sydney, New South Wales, Australia
| | - R W Paterson
- Dementia Research Centre, University College London Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - J D Warren
- Dementia Research Centre, University College London Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - H Zetterberg
- Department of Molecular Neuroscience, University College London Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Institute of Neuroscience and Physiology, Mölndal, Sweden
| | - J T O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - N C Fox
- Dementia Research Centre, University College London Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - G M Halliday
- Neuroscience Research Australia and the University of NSW, Sydney, New South Wales, Australia
| | - J M Schott
- Dementia Research Centre, University College London Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
3130
|
Bras J, Guerreiro R, Darwent L, Parkkinen L, Ansorge O, Escott-Price V, Hernandez DG, Nalls MA, Clark LN, Honig LS, Marder K, Van Der Flier WM, Lemstra A, Scheltens P, Rogaeva E, St George-Hyslop P, Londos E, Zetterberg H, Ortega-Cubero S, Pastor P, Ferman TJ, Graff-Radford NR, Ross OA, Barber I, Braae A, Brown K, Morgan K, Maetzler W, Berg D, Troakes C, Al-Sarraj S, Lashley T, Compta Y, Revesz T, Lees A, Cairns N, Halliday GM, Mann D, Pickering-Brown S, Dickson DW, Singleton A, Hardy J. Genetic analysis implicates APOE, SNCA and suggests lysosomal dysfunction in the etiology of dementia with Lewy bodies. Hum Mol Genet 2014; 23:6139-46. [PMID: 24973356 PMCID: PMC4222357 DOI: 10.1093/hmg/ddu334] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 05/15/2014] [Accepted: 06/24/2014] [Indexed: 01/28/2023] Open
Abstract
Clinical and neuropathological similarities between dementia with Lewy bodies (DLB), Parkinson's and Alzheimer's diseases (PD and AD, respectively) suggest that these disorders may share etiology. To test this hypothesis, we have performed an association study of 54 genomic regions, previously implicated in PD or AD, in a large cohort of DLB cases and controls. The cohort comprised 788 DLB cases and 2624 controls. To minimize the issue of potential misdiagnosis, we have also performed the analysis including only neuropathologically proven DLB cases (667 cases). The results show that the APOE is a strong genetic risk factor for DLB, confirming previous findings, and that the SNCA and SCARB2 loci are also associated after a study-wise Bonferroni correction, although these have a different association profile than the associations reported for the same loci in PD. We have previously shown that the p.N370S variant in GBA is associated with DLB, which, together with the findings at the SCARB2 locus, suggests a role for lysosomal dysfunction in this disease. These results indicate that DLB has a unique genetic risk profile when compared with the two most common neurodegenerative diseases and that the lysosome may play an important role in the etiology of this disorder. We make all these data available.
Collapse
Affiliation(s)
- Jose Bras
- Department of Molecular Neuroscience,
| | | | | | - Laura Parkkinen
- Nuffield Department of Clinical Neurosciences, Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Olaf Ansorge
- Nuffield Department of Clinical Neurosciences, Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Valentina Escott-Price
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Dena G Hernandez
- Laboratory of Neurogenetics, National Institutes on Aging, NIH, Bethesda, USA
| | - Michael A Nalls
- Laboratory of Neurogenetics, National Institutes on Aging, NIH, Bethesda, USA
| | - Lorraine N Clark
- Taub Institute for Alzheimer Disease and the Aging Brain, Department of Pathology and Cell Biology, and
| | - Lawrence S Honig
- Taub Institute for Alzheimer Disease and the Aging Brain, Department of Neurology, Columbia University, New York, NY, USA
| | - Karen Marder
- Taub Institute for Alzheimer Disease and the Aging Brain, Department of Neurology, Columbia University, New York, NY, USA
| | - Wiesje M Van Der Flier
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Afina Lemstra
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Philip Scheltens
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Ekaterina Rogaeva
- Department of Medicine, Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Peter St George-Hyslop
- Department of Medicine, Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada, Cambridge Institute for Medical Research, and Cambridge National Institute of Health Research Biomedical Research Unit in Dementia, University of Cambridge, Cambridge CB2 0XY, UK
| | - Elisabet Londos
- Clinical Memory Research Unit, Institute Clinical Sciences Malmö, Lund University, Sweden
| | - Henrik Zetterberg
- Department of Molecular Neuroscience, Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Sara Ortega-Cubero
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, University of Navarra, Pamplona, Spain, Department of Neurology, Clínica Universidad de Navarra, 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
| | - Pau Pastor
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, University of Navarra, Pamplona, Spain, Department of Neurology, Clínica Universidad de Navarra, 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
| | | | | | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Imelda Barber
- Translation Cell Sciences - Human Genetics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, UK
| | - Anne Braae
- Translation Cell Sciences - Human Genetics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, UK
| | - Kristelle Brown
- Translation Cell Sciences - Human Genetics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, UK
| | - Kevin Morgan
- Translation Cell Sciences - Human Genetics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, UK
| | - Walter Maetzler
- Hertie Institute for Clinical Brain Research, Department of Neurodegeneration, Center of Neurology, University of Tuebingen, and DZNE, German Center for Neurodegenerative Diseases, Tuebingen, Germany
| | - Daniela Berg
- Hertie Institute for Clinical Brain Research, Department of Neurodegeneration, Center of Neurology, University of Tuebingen, and DZNE, German Center for Neurodegenerative Diseases, Tuebingen, Germany
| | - Claire Troakes
- MRC London Neurodegenerative Diseases Brain Bank, Department of Clinical Neuroscience, King's College London, Institute of Psychiatry, London, UK
| | - Safa Al-Sarraj
- MRC London Neurodegenerative Diseases Brain Bank, Department of Clinical Neuroscience, King's College London, Institute of Psychiatry, London, UK
| | - Tammaryn Lashley
- Queen Square Brain Bank, Department of Molecular Neuroscience, and
| | - Yaroslau Compta
- Queen Square Brain Bank, Department of Molecular Neuroscience, and Parkinson's disease and Movement Disorders Unit, Neurology Service, IDIBAPS, CIBERNED, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Tamas Revesz
- Queen Square Brain Bank, Department of Molecular Neuroscience, and
| | - Andrew Lees
- Queen Square Brain Bank, Department of Molecular Neuroscience, and
| | - Nigel Cairns
- Knight Alzheimer's Disease Research Center and Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Glenda M Halliday
- Neuroscience Research Australia, Sydney, Australia, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - David Mann
- Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK and
| | - Stuart Pickering-Brown
- Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK and
| | | | - Andrew Singleton
- Laboratory of Neurogenetics, National Institutes on Aging, NIH, Bethesda, USA
| | - John Hardy
- Reta Lila Weston Research Laboratories, Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK
| |
Collapse
|
3131
|
Knowles EEM, Mathias SR, McKay DR, Sprooten E, Blangero J, Almasy L, Glahn DC. Genome-Wide Analyses of Working-Memory Ability: A Review. Curr Behav Neurosci Rep 2014; 1:224-233. [PMID: 25729637 PMCID: PMC4339023 DOI: 10.1007/s40473-014-0028-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Working memory, a theoretical construct from the field of cognitive psychology, is crucial to everyday life. It refers to the ability to temporarily store and manipulate task-relevant information. The identification of genes for working memory might shed light on the molecular mechanisms of this important cognitive ability and-given the genetic overlap between, for example, schizophrenia risk and working-memory ability-might also reveal important candidate genes for psychiatric illness. A number of genome-wide searches for genes that influence working memory have been conducted in recent years. Interestingly, the results of those searches converge on the mediating role of neuronal excitability in working-memory performance, such that the role of each gene highlighted by genome-wide methods plays a part in ion channel formation and/or dopaminergic signaling in the brain, with either direct or indirect influence on dopamine levels in the prefrontal cortex. This result dovetails with animal models of working memory that highlight the role of dynamic network connectivity, as mediated by dopaminergic signaling, in the dorsolateral prefrontal cortex. Future work, which aims to characterize functional variants influencing working-memory ability, might choose to focus on those genes highlighted in the present review and also those networks in which the genes fall. Confirming gene associations and highlighting functional characterization of those associations might have implications for the understanding of normal variation in working-memory ability and also for the development of drugs for mental illness.
Collapse
Affiliation(s)
- E E M Knowles
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Olin Neuropsychiatric Research Center, Institute of Living, Hartford, Hospital, Hartford, CT, USA
| | - S R Mathias
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Olin Neuropsychiatric Research Center, Institute of Living, Hartford, Hospital, Hartford, CT, USA
| | - D R McKay
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Olin Neuropsychiatric Research Center, Institute of Living, Hartford, Hospital, Hartford, CT, USA
| | - E Sprooten
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Olin Neuropsychiatric Research Center, Institute of Living, Hartford, Hospital, Hartford, CT, USA
| | - John Blangero
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Laura Almasy
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - D C Glahn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Olin Neuropsychiatric Research Center, Institute of Living, Hartford, Hospital, Hartford, CT, USA
| |
Collapse
|
3132
|
Cavedo E, Lista S, Khachaturian Z, Aisen P, Amouyel P, Herholz K, Jack CR, Sperling R, Cummings J, Blennow K, O'Bryant S, Frisoni GB, Khachaturian A, Kivipelto M, Klunk W, Broich K, Andrieu S, de Schotten MT, Mangin JF, Lammertsma AA, Johnson K, Teipel S, Drzezga A, Bokde A, Colliot O, Bakardjian H, Zetterberg H, Dubois B, Vellas B, Schneider LS, Hampel H. The Road Ahead to Cure Alzheimer's Disease: Development of Biological Markers and Neuroimaging Methods for Prevention Trials Across all Stages and Target Populations. J Prev Alzheimers Dis 2014; 1:181-202. [PMID: 26478889 PMCID: PMC4606938 DOI: 10.14283/jpad.2014.32] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Alzheimer's disease (AD) is a slowly progressing non-linear dynamic brain disease in which pathophysiological abnormalities, detectable in vivo by biological markers, precede overt clinical symptoms by many years to decades. Use of these biomarkers for the detection of early and preclinical AD has become of central importance following publication of two international expert working group's revised criteria for the diagnosis of AD dementia, mild cognitive impairment (MCI) due to AD, prodromal AD and preclinical AD. As a consequence of matured research evidence six AD biomarkers are sufficiently validated and partly qualified to be incorporated into operationalized clinical diagnostic criteria and use in primary and secondary prevention trials. These biomarkers fall into two molecular categories: biomarkers of amyloid-beta (Aβ) deposition and plaque formation as well as of tau-protein related hyperphosphorylation and neurodegeneration. Three of the six gold-standard ("core feasible) biomarkers are neuroimaging measures and three are cerebrospinal fluid (CSF) analytes. CSF Aβ1-42 (Aβ1-42), also expressed as Aβ1-42 : Aβ1-40 ratio, T-tau, and P-tau Thr181 & Thr231 proteins have proven diagnostic accuracy and risk enhancement in prodromal MCI and AD dementia. Conversely, having all three biomarkers in the normal range rules out AD. Intermediate conditions require further patient follow-up. Magnetic resonance imaging (MRI) at increasing field strength and resolution allows detecting the evolution of distinct types of structural and functional abnormality pattern throughout early to late AD stages. Anatomical or volumetric MRI is the most widely used technique and provides local and global measures of atrophy. The revised diagnostic criteria for "prodromal AD" and "mild cognitive impairment due to AD" include hippocampal atrophy (as the fourth validated biomarker), which is considered an indicator of regional neuronal injury. Advanced image analysis techniques generate automatic and reproducible measures both in regions of interest, such as the hippocampus and in an exploratory fashion, observer and hypothesis-indedendent, throughout the entire brain. Evolving modalities such as diffusion-tensor imaging (DTI) and advanced tractography as well as resting-state functional MRI provide useful additionally useful measures indicating the degree of fiber tract and neural network disintegration (structural, effective and functional connectivity) that may substantially contribute to early detection and the mapping of progression. These modalities require further standardization and validation. The use of molecular in vivo amyloid imaging agents (the fifth validated biomarker), such as the Pittsburgh Compound-B and markers of neurodegeneration, such as fluoro-2-deoxy-D-glucose (FDG) (as the sixth validated biomarker) support the detection of early AD pathological processes and associated neurodegeneration. How to use, interpret, and disclose biomarker results drives the need for optimized standardization. Multimodal AD biomarkers do not evolve in an identical manner but rather in a sequential but temporally overlapping fashion. Models of the temporal evolution of AD biomarkers can take the form of plots of biomarker severity (degree of abnormality) versus time. AD biomarkers can be combined to increase accuracy or risk. A list of genetic risk factors is increasingly included in secondary prevention trials to stratify and select individuals at genetic risk of AD. Although most of these biomarker candidates are not yet qualified and approved by regulatory authorities for their intended use in drug trials, they are nonetheless applied in ongoing clinical studies for the following functions: (i) inclusion/exclusion criteria, (ii) patient stratification, (iii) evaluation of treatment effect, (iv) drug target engagement, and (v) safety. Moreover, novel promising hypothesis-driven, as well as exploratory biochemical, genetic, electrophysiological, and neuroimaging markers for use in clinical trials are being developed. The current state-of-the-art and future perspectives on both biological and neuroimaging derived biomarker discovery and development as well as the intended application in prevention trials is outlined in the present publication.
Collapse
Affiliation(s)
- E Cavedo
- Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A) Hôpital de la Pitié-Salpétrière & Institut du Cerveau et de la Moelle épinière (ICM), UMR S 1127, Hôpital de la Pitié-Salpétrière Paris & CATI multicenter neuroimaging platform, France; Laboratory of Epidemiology, Neuroimaging and Telemedicine, IRCCS San Giovanni di Dio Fatebenefratelli Brescia, Italy
| | - S Lista
- AXA Research Fund & UPMC Chair; Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A) Hôpital de la Pitié-Salpétrière & Inserm U1127 Institut du Cerveau et de la Moelle épinière (ICM), Hôpital de la Pitié-Salpétrière Paris, France
| | - Z Khachaturian
- The Campaign to Prevent Alzheimer's Disease by 2020 (PAD2020), Potomac, MD, USA
| | - P Aisen
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - P Amouyel
- Inserm, U744, Lille, 59000, France; Université Lille 2, Lille, 59000, France; Institut Pasteur de Lille, Lille, 59000, France; Centre Hospitalier Régional Universitaire de Lille, Lille, 59000, France
| | - K Herholz
- Institute of Brain, Behaviour and Mental Health, University of Manchester, UK
| | - C R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - R Sperling
- Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - J Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, 888 West Bonneville Avenue, Las Vegas, Nevada 89106, USA
| | - K Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - S O'Bryant
- Department of Internal Medicine, Institute for Aging & Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - G B Frisoni
- IRCCS Istituto Centro S. Giovanni di Dio Fatebenefratelli, Brescia, Italy; University Hospitals and University of Geneva, Geneva, Switzerland
| | | | - M Kivipelto
- Karolinska Institutet Alzheimer Research Center, NVS, Stockholm, Sweden
| | - W Klunk
- Department of Psychiatry, University of Pittsburgh School of Medicine, USA; Department of Neurology, University of Pittsburgh School of Medicine, USA
| | - K Broich
- Federal Institute of Drugs and Medical Devices (BfArM), Bonn, Germany
| | - S Andrieu
- Inserm UMR1027, Université de Toulouse III Paul Sabatier, Toulouse, France; Public health department, CHU de Toulouse
| | - M Thiebaut de Schotten
- Natbrainlab, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, King's College London, London, UK; Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière (ICM), UMRS 1127 Paris, France; Inserm, U 1127, Paris, France; CNRS, UMR 7225, Paris, France
| | - J-F Mangin
- CEA UNATI, Neurospin, CEA Gif-sur-Yvette, France & CATI multicenter neuroimaging platform
| | - A A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - K Johnson
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - S Teipel
- Department of Psychosomatic Medicine, University of Rostock, and DZNE, German Center for Neurodegenerative Diseases, Rostock, Germany
| | - A Drzezga
- Department of Nuclear Medicine, University Hospital of Cologne, Cologne Germany
| | - A Bokde
- Cognitive Systems Group, Discipline of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - O Colliot
- Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013, Paris, France; Institut du Cerveau et de la Moelle épinière, ICM, Inserm, U1127, F-75013, Paris, France; CNRS, UMR 7225 ICM, 75013, Paris, France; Inria, Aramis project-team, Centre de Recherche Paris-Rocquencourt, France
| | - H Bakardjian
- Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpétrière University Hospital, Paris, France; IHU-A-ICM - Paris Institute of Translational Neurosciences, Paris, France
| | - H Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology, Queen Square, London, UK
| | - B Dubois
- Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A) Hôpital de la Pitié-Salpétrière & Inserm U1127 Institut du Cerveau et de la Moelle épinière (ICM), Hôpital de la Pitié-Salpétrière Paris, France
| | - B Vellas
- Inserm UMR1027, University of Toulouse, Toulouse, France
| | - L S Schneider
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - H Hampel
- AXA Research Fund & UPMC Chair; Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A) Hôpital de la Pitié-Salpétrière & Inserm U1127 Institut du Cerveau et de la Moelle épinière (ICM), Hôpital de la Pitié-Salpétrière Paris, France
| |
Collapse
|
3133
|
Bai Z, Han G, Xie B, Wang J, Song F, Peng X, Lei H. AlzBase: an Integrative Database for Gene Dysregulation in Alzheimer's Disease. Mol Neurobiol 2014; 53:310-319. [PMID: 25432889 DOI: 10.1007/s12035-014-9011-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 11/14/2014] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) affects a significant portion of elderly people worldwide. Although the amyloid-β (Aβ) cascade hypothesis has been the prevailing theory for the molecular mechanism of AD in the past few decades, treatment strategies targeting the Aβ cascade have not demonstrated effectiveness as yet. Thus, elucidating the spatial and temporal evolution of the molecular pathways in AD remains to be a daunting task. To facilitate novel discoveries in this filed, here, we have integrated information from multiple sources for the better understanding of gene functions in AD pathogenesis. Several categories of information have been collected, including (1) gene dysregulation in AD and closely related processes/diseases such as aging and neurological disorders, (2) correlation of gene dysregulation with AD severity, (3) a wealth of annotations on the functional and regulatory information, and (4) network connections for gene-gene relationship. In addition, we have also provided a comprehensive summary for the top ranked genes in AlzBase. By evaluating the information curated in AlzBase, researchers can prioritize genes from their own research and generate novel hypothesis regarding the molecular mechanism of AD. To demonstrate the utility of AlzBase, we examined the genes from the genetic studies of AD. It revealed links between the upstream genetic variations and downstream endo-phenotype and suggested several genes with higher priority. This integrative database is freely available on the web at http://alz.big.ac.cn/alzBase .
Collapse
Affiliation(s)
- 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
| | - Guangchun Han
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bin Xie
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, 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
| | - 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
| | - Xing Peng
- 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
|
3134
|
Li X, Melief E, Postupna N, Montine KS, Keene CD, Montine TJ. Prostaglandin E2 receptor subtype 2 regulation of scavenger receptor CD36 modulates microglial Aβ42 phagocytosis. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:230-9. [PMID: 25452117 DOI: 10.1016/j.ajpath.2014.09.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 09/14/2014] [Accepted: 09/18/2014] [Indexed: 12/18/2022]
Abstract
Recent studies underline the potential relevance of microglial innate immune activation in Alzheimer disease. Primary mouse microglia that lack prostaglandin E2 receptor subtype 2 (EP2) show decreased innate immune-mediated neurotoxicity and increased amyloid β (Aβ) peptide phagocytosis, features that were replicated in vivo. Here, we tested the hypothesis that scavenger receptor CD36 is an effector of EP2-regulated Aβ phagocytosis. CD36 expression was 143-fold greater in mouse primary microglia than in primary astrocytes. Three different means of suppressing EP2 signaling increased and an agonist of EP2 decreased CD36 expression in primary wild-type microglia. Activation of Toll-like receptor (TLR) 3, TLR4, and TLR7, but not TLR2 or TLR9, reduced primary microglial CD36 transcription and cell surface CD36 protein and reduced Aβ42 phagocytosis as well. At each step, the effects of innate immune activation on CD36 were reversed by at least 50% by an EP2 antagonist, and this partial rescue of microglia Aβ42 phagocytosis was largely mediated by CD36 activity. Finally, we showed in hippocampus of wild-type mice that innate immune activation suppressed CD36 expression by an EP2-dependent mechanism. Taken together with results of others that found brain clearance of Aβ peptides and behavioral improvements mediated by CD36 in mice, regulation of CD36-mediated Aβ phagocytosis by suppression of EP2 signaling may provide a new approach to suppressing some aspects of Alzheimer disease pathogenesis.
Collapse
Affiliation(s)
- Xianwu Li
- Department of Pathology, University of Washington, Seattle, Washington.
| | - Erica Melief
- Department of Pathology, University of Washington, Seattle, Washington
| | - Nadia Postupna
- Department of Pathology, University of Washington, Seattle, Washington
| | | | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington
| | - Thomas J Montine
- Department of Pathology, University of Washington, Seattle, Washington
| |
Collapse
|
3135
|
de Bruijn RFAG, Ikram MA. Cardiovascular risk factors and future risk of Alzheimer's disease. BMC Med 2014; 12:130. [PMID: 25385322 PMCID: PMC4226863 DOI: 10.1186/s12916-014-0130-5] [Citation(s) in RCA: 242] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/15/2014] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder in elderly people, but there are still no curative options. Senile plaques and neurofibrillary tangles are considered hallmarks of AD, but cerebrovascular pathology is also common. In this review, we summarize findings on cardiovascular disease (CVD) and risk factors in the etiology of AD. Firstly, we discuss the association of clinical CVD (such as stroke and heart disease) and AD. Secondly, we summarize the relation between imaging makers of pre-clinical vascular disease and AD. Lastly, we discuss the association of cardiovascular risk factors and AD. We discuss both established cardiovascular risk factors and emerging putative risk factors, which exert their effect partly via CVD.
Collapse
Affiliation(s)
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Wytemaweg 80, Rotterdam, 3015, CN, the Netherlands.
| |
Collapse
|
3136
|
Wilkins HM, Carl SM, Greenlief ACS, Festoff BW, Swerdlow RH. Bioenergetic dysfunction and inflammation in Alzheimer's disease: a possible connection. Front Aging Neurosci 2014; 6:311. [PMID: 25426068 PMCID: PMC4226164 DOI: 10.3389/fnagi.2014.00311] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/23/2014] [Indexed: 11/29/2022] Open
Abstract
Inflammation is observed in Alzheimer’s disease (AD) subject brains. Inflammation-relevant genes are increasingly implicated in AD genetic studies, and inflammatory cytokines to some extent even function as peripheral biomarkers. What underlies AD inflammation is unclear, but no “foreign” agent has been implicated. This suggests that internally produced damage-associated molecular pattern (DAMPs) molecules may drive inflammation in AD. A more complete characterization and understanding of AD-relevant DAMPs could advance our understanding of AD and suggest novel therapeutic strategies. In this review, we consider the possibility that mitochondria, intracellular organelles that resemble bacteria in many ways, trigger and maintain chronic inflammation in AD subjects. Data supporting the possible nexus between AD-associated bioenergetic dysfunction are discussed.
Collapse
Affiliation(s)
- Heather M Wilkins
- Department of Neurology, University of Kansas Medical Center , Kansas City, KS , USA ; University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center , Kansas City, KS , USA
| | - Steven M Carl
- University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center , Kansas City, KS , USA
| | - Alison C S Greenlief
- University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center , Kansas City, KS , USA
| | - Barry W Festoff
- Department of Neurology, University of Kansas Medical Center , Kansas City, KS , USA ; Department of Pharmacology, University of Kansas Medical Center , Kansas City, KS , USA ; Department of Molecular and Integrative Physiology, University of Kansas Medical Center , Kansas City, KS , USA ; pHLOGISTIX Neurodiagnostics , Lenexa, KS , USA
| | - Russell H Swerdlow
- Department of Neurology, University of Kansas Medical Center , Kansas City, KS , USA ; University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center , Kansas City, KS , USA ; Department of Molecular and Integrative Physiology, University of Kansas Medical Center , Kansas City, KS , USA ; Department of Biochemistry and Molecular Biology, University of Kansas Medical Center , Kansas City, KS , USA
| |
Collapse
|
3137
|
Taymans JM, Baekelandt V. Phosphatases of α-synuclein, LRRK2, and tau: important players in the phosphorylation-dependent pathology of Parkinsonism. Front Genet 2014; 5:382. [PMID: 25426138 PMCID: PMC4224088 DOI: 10.3389/fgene.2014.00382] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 10/17/2014] [Indexed: 12/20/2022] Open
Abstract
An important challenge in the field of Parkinson’s disease (PD) is to develop disease modifying therapies capable of stalling or even halting disease progression. Coupled to this challenge is the need to identify disease biomarkers, in order to identify pre-symptomatic hallmarks of disease and monitor disease progression. The answer to these challenges lies in the elucidation of the molecular causes underlying PD, for which important leads are disease genes identified in studies investigating the underlying genetic causes of PD. LRRK2 and α-syn have been both linked to familial forms of PD as well as associated to sporadic PD. Another gene, microtubule associated protein tau (MAPT), has been genetically linked to a dominant form of frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) and genome-wide association studies report a strong association between MAPT and sporadic PD. Interestingly, LRRK2, α-syn, and tau are all phosphorylated proteins, and their phosphorylation patterns are linked to disease. In this review, we provide an overview of the evidence linking LRRK2, α-syn, and tau phosphorylation to PD pathology and focus on studies which have identified phosphatases responsible for dephosphorylation of pathology-related phosphorylations. We also discuss how the LRRK2, α-syn, and tau phosphatases may point to separate or cross-talking pathological pathways in PD. Finally, we will discuss how the study of phosphatases of dominant Parkinsonism proteins opens perspectives for targeting pathological phosphorylation events.
Collapse
Affiliation(s)
- Jean-Marc Taymans
- Department of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven Leuven, Belgium
| | - Veerle Baekelandt
- Department of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven Leuven, Belgium
| |
Collapse
|
3138
|
Liu G, Bao X, Jiang Y, Liao M, Jiang Q, Feng R, Zhang L, Ma G, Chen Z, Wang G, Wang R, Zhao B, Li K. Identifying the Association Between Alzheimer's Disease and Parkinson's Disease Using Genome-Wide Association Studies and Protein-Protein Interaction Network. Mol Neurobiol 2014; 52:1629-1636. [PMID: 25370933 DOI: 10.1007/s12035-014-8946-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 10/20/2014] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the first and second most common neurodegenerative diseases in the elderly. Shared clinical and pathological features have been reported. Recent large-scale genome-wide association studies (GWAS) have been conducted and reported a number of AD and PD variants. Until now, the underlying genetic mechanisms for all these newly identified PD variants as well as the association between AD and PD are still unclear exactly. We think that PD variants may contribute to AD and PD by influence on brain gene expression. Here, we conducted a systems analysis using (1) AD and PD variants (P < 5.00E-08) identified by the published GWAS; (2) four brain expression GWAS datasets using expression quantitative trait loci from the cerebellum and temporal cortex; (3) large-scale AD GWAS from the Alzheimer Disease Genetics Consortium (ADGC); (4) a protein-protein interaction network. Our results indicated that PD variants around the 17q21 were associated with gene expression and suggestive AD risk. We also identified significant interaction among AD and PD susceptibility genes. We believe that our findings may explain the underlying genetic mechanisms for newly identified PD variants in PD and AD, as well as the association between AD and PD, which may be very useful for future genetic studies for both diseases.
Collapse
Affiliation(s)
- Guiyou Liu
- Institute of Neurology, Guangdong Medical College, Zhanjiang, 524001, China.,Genome Analysis Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Xiqi Dao 32, Tianjin Airport Economic Area, Tianjin, 300308, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Mingzhi Liao
- College of Life Science, Northwest A&F University, Yangling, Shaanxi, China
| | - Qinghua Jiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Rennan Feng
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Liangcai Zhang
- Department of Statistics, Rice University, Houston, TX, USA
| | - Guoda Ma
- Institute of Neurology, Guangdong Medical College, Zhanjiang, 524001, China
| | - Zugen Chen
- Department of Human Genetics, University of California at Los Angeles, Los Angeles, CA, USA
| | - Guangyu Wang
- Department of Oncology, The First Hospital of Harbin, Harbin, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Bin Zhao
- Institute of Neurology, Guangdong Medical College, Zhanjiang, 524001, China.
| | - Keshen Li
- Institute of Neurology, Guangdong Medical College, Zhanjiang, 524001, China.
| |
Collapse
|
3139
|
Pankevich DE, Altevogt BM, Dunlop J, Gage FH, Hyman SE. Improving and accelerating drug development for nervous system disorders. Neuron 2014; 84:546-53. [PMID: 25442933 PMCID: PMC4254615 DOI: 10.1016/j.neuron.2014.10.007] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Advances in the neurosciences have placed the field in the position where it is poised to significantly reduce the burden of nervous system disorders. However, drug discovery, development, and translation for nervous system disorders still pose many unique challenges. The key scientific challenges can be summarized as follows: mechanisms of disease, target identification and validation, predictive models, biomarkers for patient stratification and as endpoints for clinical trials, clear regulatory pathways, reliability and reproducibility of published data, and data sharing and collaboration. To accelerate nervous system drug development, the Institute of Medicine's Forum on Neuroscience and Nervous System Disorders has hosted a series of public workshops that brought together representatives of industry, government (including both research funding and regulatory agencies), academia, and patient groups to discuss these challenges and offer potential strategies to improve the translational neuroscience.
Collapse
Affiliation(s)
- Diana E Pankevich
- Forum on Neuroscience and Nervous System Disorders, Institute of Medicine, Washington, DC 20001, USA
| | - Bruce M Altevogt
- Forum on Neuroscience and Nervous System Disorders, Institute of Medicine, Washington, DC 20001, USA.
| | - John Dunlop
- Innovative Medicines and Early Development Unit, AstraZeneca Neuroscience, Cambridge, MA 02139, USA
| | - Fred H Gage
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Steve E Hyman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| |
Collapse
|
3140
|
Yaghmoor F, Noorsaeed A, Alsaggaf S, Aljohani W, Scholtzova H, Boutajangout A, Wisniewski T. The Role of TREM2 in Alzheimer's Disease and Other Neurological Disorders. JOURNAL OF ALZHEIMER'S DISEASE & PARKINSONISM 2014; 4:160. [PMID: 25664220 PMCID: PMC4317331 DOI: 10.4172/2161-0460.1000160] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide. Late-onset AD (LOAD), is the most common form of Alzheimer's disease, representing about >95% of cases and early-onset AD represents <5% of cases. Several risk factors have been discovered that are associated with AD, with advancing age being the most prominent. Other environmental risk factors include diabetes mellitus, level of physical activity, educational status, hypertension and head injury. The most well known genetic risk factor for LOAD is inheritance of the apolipoprotein (apo) E4 allele. Recently, rare variants of TREM2 have been reported as a significant risk factor for LOAD, comparable to inheritance of apoE4. In this review we will focus on the role(s) of TREM2 in AD as well as in other neurodegenerative disorders.
Collapse
Affiliation(s)
- Faris Yaghmoor
- Departments of Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, USA
| | - Ahmed Noorsaeed
- Pathology, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, USA
| | - Samar Alsaggaf
- Departments of Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, USA
| | - Waleed Aljohani
- Departments of Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, USA
| | - Henrieta Scholtzova
- Departments of Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, USA
| | - Allal Boutajangout
- Departments of Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, USA
- Psychiatry, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, USA
- Physiology and Neuroscience, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY10016, USA
- King Abdulaziz University, School of Medicine, Jeddah, Saudi Arabia
| | - Thomas Wisniewski
- Departments of Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, USA
- Pathology, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, USA
- Psychiatry, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, USA
| |
Collapse
|
3141
|
Lord J, Lu AJ, Cruchaga C. Identification of rare variants in Alzheimer's disease. Front Genet 2014; 5:369. [PMID: 25389433 PMCID: PMC4211559 DOI: 10.3389/fgene.2014.00369] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 10/03/2014] [Indexed: 12/21/2022] Open
Abstract
Much progress has been made in recent years in identifying genes involved in the risk of developing Alzheimer's disease (AD), the most common form of dementia. Yet despite the identification of over 20 disease associated loci, mainly through genome wide association studies (GWAS), a large proportion of the genetic component of the disorder remains unexplained. Recent evidence from the AD field, as with other complex diseases, suggests a large proportion of this "missing heritability" may be due to rare variants of moderate to large effect size, but the methodologies to detect such variants are still in their infancy. The latest studies in the field have been focused on the identification of coding variation associated with AD risk, through whole-exome or whole-genome sequencing. Such variants are expected to have larger effect sizes than GWAS loci, and are easier to functionally characterize, and develop cellular and animal models for. This review explores the issues involved in detecting rare variant associations in the context of AD, highlighting some successful approaches utilized to date.
Collapse
Affiliation(s)
- Jenny Lord
- Department of Psychiatry, Washington University School of MedicineSt. Louis, MO, USA
| | - Alexander J. Lu
- Department of Psychiatry, Washington University School of MedicineSt. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of MedicineSt. Louis, MO, USA
- Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University School of MedicineSt. Louis, MO, USA
| |
Collapse
|
3142
|
Kauwe JSK, Bailey MH, Ridge PG, Perry R, Wadsworth ME, Hoyt KL, Staley LA, Karch CM, Harari O, Cruchaga C, Ainscough BJ, Bales K, Pickering EH, Bertelsen S, the Alzheimer's Disease Neuroimaging Initiative, Fagan AM, Holtzman DM, Morris JC, Goate AM. Genome-wide association study of CSF levels of 59 alzheimer's disease candidate proteins: significant associations with proteins involved in amyloid processing and inflammation. PLoS Genet 2014; 10:e1004758. [PMID: 25340798 PMCID: PMC4207667 DOI: 10.1371/journal.pgen.1004758] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 09/16/2014] [Indexed: 01/25/2023] Open
Abstract
Cerebrospinal fluid (CSF) 42 amino acid species of amyloid beta (Aβ42) and tau levels are strongly correlated with the presence of Alzheimer's disease (AD) neuropathology including amyloid plaques and neurodegeneration and have been successfully used as endophenotypes for genetic studies of AD. Additional CSF analytes may also serve as useful endophenotypes that capture other aspects of AD pathophysiology. Here we have conducted a genome-wide association study of CSF levels of 59 AD-related analytes. All analytes were measured using the Rules Based Medicine Human DiscoveryMAP Panel, which includes analytes relevant to several disease-related processes. Data from two independently collected and measured datasets, the Knight Alzheimer's Disease Research Center (ADRC) and Alzheimer's Disease Neuroimaging Initiative (ADNI), were analyzed separately, and combined results were obtained using meta-analysis. We identified genetic associations with CSF levels of 5 proteins (Angiotensin-converting enzyme (ACE), Chemokine (C-C motif) ligand 2 (CCL2), Chemokine (C-C motif) ligand 4 (CCL4), Interleukin 6 receptor (IL6R) and Matrix metalloproteinase-3 (MMP3)) with study-wide significant p-values (p<1.46×10−10) and significant, consistent evidence for association in both the Knight ADRC and the ADNI samples. These proteins are involved in amyloid processing and pro-inflammatory signaling. SNPs associated with ACE, IL6R and MMP3 protein levels are located within the coding regions of the corresponding structural gene. The SNPs associated with CSF levels of CCL4 and CCL2 are located in known chemokine binding proteins. The genetic associations reported here are novel and suggest mechanisms for genetic control of CSF and plasma levels of these disease-related proteins. Significant SNPs in ACE and MMP3 also showed association with AD risk. Our findings suggest that these proteins/pathways may be valuable therapeutic targets for AD. Robust associations in cognitively normal individuals suggest that these SNPs also influence regulation of these proteins more generally and may therefore be relevant to other diseases. The use of quantitative endophenotypes from cerebrospinal fluid has led to the identification of several genetic variants that alter risk or rate of progression of Alzheimer's disease. Here we have analyzed the levels of 58 disease-related proteins in the cerebrospinal fluid for association with millions of variants across the human genome. We have identified significant, replicable associations with 5 analytes, Angiotensin-converting enzyme, Chemokine (C-C motif) ligand 2, Chemokine (C-C motif) ligand 4, Interleukin 6 receptor and Matrix metalloproteinase-3. Our results suggest that these variants play a regulatory role in the respective protein levels and are relevant to the inflammatory and amyloid processing pathways. Variants in associated with ACE and those associated with MMP3 levels also show association with risk for Alzheimer's disease in the expected directions. These associations are consistent in cerebrospinal fluid and plasma and in samples with only cognitively normal individuals suggesting that they are relevant in the regulation of these protein levels beyond the context of Alzheimer's disease.
Collapse
Affiliation(s)
- John S. K. Kauwe
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Matthew H. Bailey
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Perry G. Ridge
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Rachel Perry
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Mark E. Wadsworth
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Kaitlyn L. Hoyt
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Lyndsay A. Staley
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Celeste M. Karch
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Oscar Harari
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Benjamin J. Ainscough
- The Genome Institute, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Kelly Bales
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut, United States of America
| | - Eve H. Pickering
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut, United States of America
| | - Sarah Bertelsen
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, United States of America
| | | | - Anne M. Fagan
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - David M. Holtzman
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Developmental Biology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - John C. Morris
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Alison M. Goate
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
3143
|
Bi R, Zhang W, Yu D, Li X, Wang HZ, Hu QX, Zhang C, Lu W, Ni J, Fang Y, Li T, Yao YG. Mitochondrial DNA haplogroup B5 confers genetic susceptibility to Alzheimer's disease in Han Chinese. Neurobiol Aging 2014; 36:1604.e7-16. [PMID: 25457022 DOI: 10.1016/j.neurobiolaging.2014.10.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 09/07/2014] [Accepted: 10/10/2014] [Indexed: 02/05/2023]
Abstract
Mitochondrial dysfunction has been widely reported in psychiatric and neurodegenerative diseases. We aimed to investigate the association between matrilineal structures of Han Chinese populations and Alzheimer's disease (AD) by a 2-stage case-control study: A total of 341 AD patients and 435 normal individuals from Southwest China were analyzed for mitochondrial DNA sequence variations and were classified into respective haplogroups. A total of 371 AD patients and 470 normal individuals from East China, as validation samples, were genotyped for the variants defining the risk haplogroup. Haplogroup B5 had a significantly higher frequency in AD patients (7.33%) than in control subjects (3.68%) from Southwest China, and we found a similar pattern of higher frequency of B5 in patients in the case-control sample from East China. In the combined population, association of haplogroup B5 with AD risk was strengthened (p = 0.02; odds ratio = 1.74; 95% confidence interval = 1.10-2.76). In lymphoblastoid cell lines belonging to haplogroup B5a, we observed significantly increased reactive oxygen species and decreased mitochondrial mass. Hela cells with stable expression of the MT-ATP6 gene with B5-defining variant m.8584G>A also showed a significantly decreased mitochondrial function. Taken together, our results indicated that haplogroup B5 conferred genetic susceptibility to AD in Han Chinese, and this effect was most likely mediated by ancient variant m.8584G>A. The predisposing effect of B5 to AD is consistent with the ancestral-susceptibility model of complex diseases.
Collapse
Affiliation(s)
- Rui Bi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Wen Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Dandan Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Xiao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Hui-Zhen Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China; School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Qiu-Xiang Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chen Zhang
- Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong, University School of Medicine, Shanghai, China
| | - Weihong Lu
- Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong, University School of Medicine, Shanghai, China
| | - Jianliang Ni
- First Geriatric Department, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yiru Fang
- Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong, University School of Medicine, Shanghai, China
| | - Tao Li
- The Mental Health Center & Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China.
| |
Collapse
|
3144
|
The role of the retromer complex in aging-related neurodegeneration: a molecular and genomic review. Mol Genet Genomics 2014; 290:413-27. [PMID: 25332075 DOI: 10.1007/s00438-014-0939-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 10/10/2014] [Indexed: 10/24/2022]
Abstract
The retromer coat complex is a vital component of the intracellular trafficking mechanism sorting cargo from the endosomes to the trans-Golgi network or to the cell surface. In recent years, genes encoding components of the retromer coat complex and members of the vacuolar protein sorting 10 (Vps10) family of receptors, which play pleiotropic functions in protein trafficking and intracellular/intercellular signaling in neuronal and non-neuronal cells and are primary cargos of the retromer complex, have been implicated as genetic risk factors for sporadic and autosomal dominant forms of several neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and frontotemporal lobar degeneration. In addition to their functions in protein trafficking, the members of the Vps10 receptor family (sortilin, SorL1, SorCS1, SorCS2, and SorCS3) modulate neurotrophic signaling pathways. Both sortilin and SorCS2 act as cell surface receptors to mediate acute responses to proneurotrophins. In addition, sortilin can modulate the intracellular response to brain-derived neurotrophic factor (BDNF) by direct control of BDNF levels and regulating anterograde trafficking of Trk receptors to the synapse. This review article summarizes the emerging data from this rapidly growing field of intracellular trafficking signaling in the pathogenesis of neurodegeneration.
Collapse
|
3145
|
Homogeneous case subgroups increase power in genetic association studies. Eur J Hum Genet 2014; 23:863-9. [PMID: 25271086 DOI: 10.1038/ejhg.2014.194] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/16/2014] [Accepted: 08/20/2014] [Indexed: 12/12/2022] Open
Abstract
Genome-wide association studies of clinically defined cases against controls have transformed our understanding of the genetic causes of many diseases. However, there are limitations to the simple clinical definitions used in these studies, and GWAS analyses are beginning to explore more refined phenotypes in subgroups of the existing data sets. These analyses are often performed ad hoc without considering the power requirements to justify such analyses. Here we derive expressions for the relative power of such subgroup analyses and determine the genotypic relative risks (GRRs) required to achieve equivalent power to a full analysis for relevant scenarios. We show that only modest increases in GRRs may be required to offset the reduction in power from analysing fewer cases, implying that analyses of more genetically homogenous case subgroups may have the potential to identify further associations. We find that, for lower genotypic relative risks in the full sample, subgroup analyses of more homogeneous cases have relatively more power than for higher index genotypic relative risks and that this effect is stronger for rare as opposed to common variants. As GWA studies are likely to have now identified the majority of SNPs with stronger effects, these results strongly advocate a renewed effort to identify phenotypically homogeneous disease groups, in which power to detect genetic variants with small effects will be greater. These results suggest that analysis of case subsets could be a powerful strategy to uncover some of the hidden heritability for common complex disorders, particularly in identifying rarer variants of modest effect.
Collapse
|
3146
|
2014 Report on the Milestones for the US National Plan to Address Alzheimer's Disease. Alzheimers Dement 2014; 10:S430-52. [DOI: 10.1016/j.jalz.2014.08.103] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
3147
|
Small SA. Isolating pathogenic mechanisms embedded within the hippocampal circuit through regional vulnerability. Neuron 2014; 84:32-39. [PMID: 25277453 PMCID: PMC4185396 DOI: 10.1016/j.neuron.2014.08.030] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Some of the most common and devastating disorders of the brain target the hippocampal formation. The hippocampal formation is a complex circuit of interconnected regions, and it is assumed that clues into the causes of these disorders are embedded within the circuit. Neuroimaging tools have been optimized to interrogate the malfunctioning hippocampal circuit, and by applying these tools to patients in the earliest stages of disease and to animal models, patterns of regional vulnerability have been established for Alzheimer's disease, schizophrenia, and cognitive aging. More recently, studies have begun deciphering the cellular and molecular reasons underlying regional dysfunction. Collectively, this information clarifies the pathophysiology of these disorders and informs on therapeutic strategies.
Collapse
Affiliation(s)
- Scott A Small
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Departments of Neurology, Radiology, and Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| |
Collapse
|
3148
|
Abstract
The analyses of genetic factors contributing to Alzheimer's disease (AD) and other dementias have evolved at the same pace as genetic and genomic technologies are developed and improved. The identification of the first genes involved in AD arose from family-based studies, but risk factors have mainly been identified by studies comparing groups of patients with groups of controls. The best outcomes have been heavily associated with the capacity of interrogating genetic variability at the genome level without any particular a priori hypothesis. In this review we assess the role of genetic family studies in Alzheimer's disease and other dementias within the current status of dementias' and, particularly, AD's genetic architecture.
Collapse
Affiliation(s)
- Rita Guerreiro
- Department of Molecular Neuroscience, Institute of Neurology, UCL, 1 Wakefield Street, London, WC1N 1PJ, UK,
| | | |
Collapse
|
3149
|
Clinical workout for the early detection of cognitive decline and dementia. Eur J Clin Nutr 2014; 68:1186-91. [PMID: 25271010 DOI: 10.1038/ejcn.2014.189] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 07/18/2014] [Indexed: 02/02/2023]
Abstract
Aging is the major risk factor for the development of human neurodegenerative maladies such as Alzheimer's, Huntington's and Parkinson's diseases (PDs) and prion disorders, all of which stem from toxic protein aggregation. All of these diseases are correlated with cognitive decline. Cognitive Decline is a dynamic state from normal cognition of aging to dementia. According to the original criteria for Alzheimer's Disease (AD) (1984), a clinical diagnosis was possible only when someone was already demented. The prevalence rates of Cognitive Decline (mild cognitive impairment plus dementia) are very high now and will be higher in future because of the increasing survival time of people. Many neurological and psychiatric diseases are correlated with cognitive decline. Diagnosis of cognitive decline is mostly clinical (clinical criteria), but there are multiple biomarkers that could help us mostly in research programs such as short or long, paper and pencil or computerized neuropsychological batteries for cognition, activities of daily living and behavior, electroencephalograph, event-related potentials, and imaging-structural magnetic resonance imaging (MRI) and functional (fMRI, Pittsburgh bound positron emission tomography, FDG-PET, single photon emission computerized tomography and imaging of tau pathology)-cerebrospinal fluid proteins (Abeta, tau and phospho-tau in AD and α-synuclein (αSyn) for PD). Blood biomarkers need more studies to confirm their usefulness. Genetic markers are also studied but until now are not used in clinical praxis. Finally, in everyday clinical praxis and in research workout for early detection of cognitive decline, the combination of biomarkers is useful.
Collapse
|
3150
|
Morris GP, Clark IA, Vissel B. Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer's disease. Acta Neuropathol Commun 2014; 2:135. [PMID: 25231068 PMCID: PMC4207354 DOI: 10.1186/s40478-014-0135-5] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 08/30/2014] [Indexed: 12/16/2022] Open
Abstract
The amyloid hypothesis has driven drug development strategies for Alzheimer's disease for over 20 years. We review why accumulation of amyloid-beta (Aβ) oligomers is generally considered causal for synaptic loss and neurodegeneration in AD. We elaborate on and update arguments for and against the amyloid hypothesis with new data and interpretations, and consider why the amyloid hypothesis may be failing therapeutically. We note several unresolved issues in the field including the presence of Aβ deposition in cognitively normal individuals, the weak correlation between plaque load and cognition, questions regarding the biochemical nature, presence and role of Aβ oligomeric assemblies in vivo, the bias of pre-clinical AD models toward the amyloid hypothesis and the poorly explained pathological heterogeneity and comorbidities associated with AD. We also illustrate how extensive data cited in support of the amyloid hypothesis, including genetic links to disease, can be interpreted independently of a role for Aβ in AD. We conclude it is essential to expand our view of pathogenesis beyond Aβ and tau pathology and suggest several future directions for AD research, which we argue will be critical to understanding AD pathogenesis.
Collapse
Affiliation(s)
- Gary P Morris
- />Garvan Institute of Medical Research, Neuroscience Department, Neurodegenerative Disorders Laboratory, 384 Victoria Street, Darlinghurst, NSW 2010 Australia
- />Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Ian A Clark
- />Research School of Biology, Australian National University, Canberra, Australia
| | - Bryce Vissel
- />Garvan Institute of Medical Research, Neuroscience Department, Neurodegenerative Disorders Laboratory, 384 Victoria Street, Darlinghurst, NSW 2010 Australia
- />Faculty of Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|