2901
|
Lista S, Khachaturian ZS, Rujescu D, Garaci F, Dubois B, Hampel H. Application of Systems Theory in Longitudinal Studies on the Origin and Progression of Alzheimer's Disease. Methods Mol Biol 2016; 1303:49-67. [PMID: 26235059 DOI: 10.1007/978-1-4939-2627-5_2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This chapter questions the prevailing "implicit" assumption that molecular mechanisms and the biological phenotype of dominantly inherited early-onset alzheimer's disease (EOAD) could serve as a linear model to study the pathogenesis of sporadic late-onset alzheimer's disease (LOAD). Now there is growing evidence to suggest that such reductionism may not be warranted; these suppositions are not adequate to explain the molecular complexities of LOAD. For example, the failure of some recent amyloid-centric clinical trials, which were largely based on the extrapolations from EOAD biological phenotypes to the molecular mechanisms in the pathogenesis of LOAD, might be due to such false assumptions. The distinct difference in the biology of LOAD and EOAD is underscored by the presence of EOAD cases without evidence of familial clustering or Mendelian transmission and, conversely, the discovery and frequent reports of such clustering and transmission patterns in LOAD cases. The primary thesis of this chapter is that a radically different way of thinking is required for comprehensive explanations regarding the distinct complexities in the molecular pathogenesis of inherited and sporadic forms of Alzheimer's disease (AD). We propose using longitudinal analytical methods and the paradigm of systems biology (using transcriptomics, proteomics, metabolomics, and lipidomics) to provide us a more comprehensive insight into the lifelong origin and progression of different molecular mechanisms and neurodegeneration. Such studies should aim to clarify the role of specific pathophysiological and signaling pathways such as neuroinflammation, altered lipid metabolism, apoptosis, oxidative stress, tau hyperphosphorylation, protein misfolding, tangle formation, and amyloidogenic cascade leading to overproduction and reduced clearance of aggregating amyloid-beta (Aβ) species. A more complete understanding of the distinct difference in molecular mechanisms, signaling pathways, as well as comparability of the various forms of AD is of paramount importance. The development of knowledge and technologies for early detection and characterization of the disease across all stages will improve the predictions regarding the course of the disease, prognosis, and response to treatment. No doubt such advances will have a significant impact on the clinical management of both EOAD and LOAD patients. The approach propped here, combining longitudinal studies with the systems biology paradigm, will create a more effective and comprehensive framework for development of prevention therapies in AD.
Collapse
Affiliation(s)
- Simone Lista
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Julius-Kühn-Straße 7, 06112, Halle (Saale), Germany,
| | | | | | | | | | | |
Collapse
|
2902
|
Masdeu JC, Pascual B. Genetic and degenerative disorders primarily causing dementia. HANDBOOK OF CLINICAL NEUROLOGY 2016; 135:525-564. [PMID: 27432682 DOI: 10.1016/b978-0-444-53485-9.00026-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neuroimaging comprises a powerful set of instruments to diagnose the different causes of dementia, clarify their neurobiology, and monitor their treatment. Magnetic resonance imaging (MRI) depicts volume changes with neurodegeneration and inflammation, as well as abnormalities in functional and structural connectivity. MRI arterial spin labeling allows for the quantification of regional cerebral blood flow, characteristically altered in Alzheimer's disease, diffuse Lewy-body disease, and the frontotemporal dementias. Positron emission tomography allows for the determination of regional metabolism, with similar abnormalities as flow, and for the measurement of β-amyloid and abnormal tau deposition in the brain, as well as regional inflammation. These instruments allow for the quantification in vivo of most of the pathologic features observed in disorders causing dementia. Importantly, they allow for the longitudinal study of these abnormalities, having revealed, for instance, that the deposition of β-amyloid in the brain can antecede by decades the onset of dementia. Thus, a therapeutic window has been opened and the efficacy of immunotherapies directed at removing β-amyloid from the brain of asymptomatic individuals is currently being tested. Tau and inflammation imaging, still in their infancy, combined with genomics, should provide powerful insights into these disorders and facilitate their treatment.
Collapse
Affiliation(s)
- Joseph C Masdeu
- Department of Neurology, Houston Methodist Hospital, Houston, TX, USA.
| | - Belen Pascual
- Department of Neurology, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
2903
|
Harrison TM, Bookheimer SY. Neuroimaging genetic risk for Alzheimer's disease in preclinical individuals: From candidate genes to polygenic approaches. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2016; 1:14-23. [PMID: 26858991 DOI: 10.1016/j.bpsc.2015.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Better characterization of the preclinical phase of Alzheimer's disease (AD) is needed in order to develop effective interventions. Neuropathological changes in AD, including neuronal loss and the formation of proteinaceous deposits, begin up to 20 years before the onset of clinical symptoms. As such, the emergence of cognitive impairment should not be the sole basis used to diagnose AD nor to evaluate individuals for enrollment in clinical trials for preventative AD treatments. Instead, early preclinical biomarkers of disease and genetic risk should be used to determine most likely prognosis and enroll individuals in appropriate clinical trials. Neuroimaging-based biomarkers and genetic analysis together present a powerful system for classifying preclinical pathology in patients. Disease modifying interventions are more likely to produce positive outcomes when administered early in the course of AD. In this review, we examine the utility of the neuroimaging genetics field as it applies to AD and early detection during the preclinical phase. Neuroimaging studies focused on single genetic risk factors are summarized. However, we particularly focus on the recent increased interest in polygenic methods and discuss the benefits and disadvantages of these approaches. We discuss challenges in the neuroimaging genetics field, including limitations of statistical power arising from small effect sizes and the over-use of cross-sectional designs. Despite the limitations, neuroimaging genetics has already begun to influence clinical trial design and will play a major role in the prevention of AD.
Collapse
Affiliation(s)
- Theresa M Harrison
- Neuroscience Interdepartmental Graduate Program, UCLA, Los Angeles, CA; Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA
| | - Susan Y Bookheimer
- Department of Psychiatry and Biobehavioral Sciences, UCLA, Los Angeles, CA; Center for Cognitive Neuroscience, UCLA, Los Angeles, CA
| |
Collapse
|
2904
|
Kunkle BW, Jaworski J, Barral S, Vardarajan B, Beecham GW, Martin ER, Cantwell LS, Partch A, Bird TD, Raskind WH, DeStefano AL, Carney RM, Cuccaro M, Vance JM, Farrer LA, Goate AM, Foroud T, Mayeux RP, Schellenberg GD, Haines JL, Pericak-Vance MA. Genome-wide linkage analyses of non-Hispanic white families identify novel loci for familial late-onset Alzheimer's disease. Alzheimers Dement 2016; 12:2-10. [PMID: 26365416 PMCID: PMC4717829 DOI: 10.1016/j.jalz.2015.05.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 05/14/2015] [Accepted: 05/29/2015] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Few high penetrance variants that explain risk in late-onset Alzheimer's disease (LOAD) families have been found. METHODS We performed genome-wide linkage and identity-by-descent (IBD) analyses on 41 non-Hispanic white families exhibiting likely dominant inheritance of LOAD, and having no mutations at known familial Alzheimer's disease (AD) loci, and a low burden of APOE ε4 alleles. RESULTS Two-point parametric linkage analysis identified 14 significantly linked regions, including three novel linkage regions for LOAD (5q32, 11q12.2-11q14.1, and 14q13.3), one of which replicates a genome-wide association LOAD locus, the MS4A6A-MS4A4E gene cluster at 11q12.2. Five of the 14 regions (3q25.31, 4q34.1, 8q22.3, 11q12.2-14.1, and 19q13.41) are supported by strong multipoint results (logarithm of odds [LOD*] ≥1.5). Nonparametric multipoint analyses produced an additional significant locus at 14q32.2 (LOD* = 4.18). The 1-LOD confidence interval for this region contains one gene, C14orf177, and the microRNA Mir_320, whereas IBD analyses implicates an additional gene BCL11B, a regulator of brain-derived neurotrophic signaling, a pathway associated with pathogenesis of several neurodegenerative diseases. DISCUSSION Examination of these regions after whole-genome sequencing may identify highly penetrant variants for familial LOAD.
Collapse
Affiliation(s)
- Brian W Kunkle
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - James Jaworski
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Sandra Barral
- The Taub Institute of Research on Alzheimer's Disease, College of Physicians and Surgeons, Columbia University, New York, NY, USA; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Badri Vardarajan
- The Taub Institute of Research on Alzheimer's Disease, College of Physicians and Surgeons, Columbia University, New York, NY, USA; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gary W Beecham
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Eden R Martin
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Laura S Cantwell
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amanda Partch
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas D Bird
- Department of Neurology, University of Washington, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - Wendy H Raskind
- Department of Medicine, University of Washington, Seattle, WA, USA; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Anita L DeStefano
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Regina M Carney
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Michael Cuccaro
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA; Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jeffrey M Vance
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA; Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Lindsay A Farrer
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA; Department of Medicine (Biomedical Genetics), Boston University School of Medicine and Public Health, MA, USA; Department of Neurology, Boston University School of Medicine and Public Health, MA, USA; Department of Ophthalmology, Boston University School of Medicine and Public Health, MA, USA; Department of Epidemiology, Boston University School of Public Health, MA, USA
| | - Alison M Goate
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Richard P Mayeux
- The Taub Institute of Research on Alzheimer's Disease, College of Physicians and Surgeons, Columbia University, New York, NY, USA; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA; The Department of Epidemiology, School of Public Health, Columbia University, New York, NY, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan L Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Margaret A Pericak-Vance
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA; Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
2905
|
Neural transcriptome of constitutional Pten dysfunction in mice and its relevance to human idiopathic autism spectrum disorder. Mol Psychiatry 2016; 21:118-25. [PMID: 25754085 PMCID: PMC4565786 DOI: 10.1038/mp.2015.17] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/12/2014] [Accepted: 01/08/2015] [Indexed: 01/09/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition with a clear, but heterogeneous, genetic component. Germline mutations in the tumor suppressor Pten are a well-established risk factor for ASD with macrocephaly, and conditional Pten mouse models have impaired social behavior and brain development. Some mutations observed in patients disrupt the normally balanced nuclear-cytoplasmic localization of the Pten protein, and we developed the Pten(m3m4) model to study the effects of a cytoplasm-predominant Pten. In this model, germline mislocalization of Pten causes inappropriate social behavior with intact learning and memory, a profile reminiscent of high-functioning ASD. These animals also exhibit histological evidence of neuroinflammation and expansion of glial populations by 6 weeks of age. We hypothesized that the neural transcriptome of this model would be altered in a manner that could inform human idiopathic ASD, a constitutional condition. Using total RNA sequencing, we found progressive disruption of neural gene expression in Pten(m3m4) mice from 2-6 weeks of age, involving both immune and synaptic pathways. These alterations include downregulation of many highly coexpressed human ASD-susceptibility genes. Comparison with a human cortical development coexpression network revealed that genes disrupted in Pten(m3m4) mice were enriched in the same areas as those of human ASD. Although Pten-related ASD is relatively uncommon, our observations suggest that the Pten(m3m4) model recapitulates multiple molecular features of human ASD, and that Pten operates far upstream of common pathways within ASD pathogenesis.
Collapse
|
2906
|
Liu C, Saffen D, Schulze TG, Burmeister M, Sham PC, Yao YG, Kuo PH, Chen C, An Y, Dai J, Yue W, Li MX, Xue H, Su B, Chen L, Shi Y, Qiao M, Liu T, Xia K, Chan RCK. Psychiatric genetics in China: achievements and challenges. Mol Psychiatry 2016; 21:4-9. [PMID: 26481319 PMCID: PMC4830695 DOI: 10.1038/mp.2015.95] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
To coordinate research efforts in psychiatric genetics in China, a group of Chinese and foreign investigators have established an annual “Summit on Chinese Psychiatric Genetics” to present their latest research and discuss the current state and future directions of this field. To date, two Summits have been held, the first in Changsha in April, 2014, and the second in Kunming in April, 2015. The consensus of roundtable discussions held at these meetings is that psychiatric genetics in China is in need of new policies to promote collaborations aimed at creating a framework for genetic research appropriate for the Chinese population: relying solely on Caucasian population-based studies may result in missed opportunities to diagnose and treat psychiatric disorders. In addition, participants agree on the importance of promoting collaborations and data sharing in areas where China has especially strong resources, such as advanced facilities for non-human primate studies and traditional Chinese medicine: areas that may also provide overseas investigators with unique research opportunities. In this paper, we present an overview of the current state of psychiatric genetics research in China, with emphasis on genome-level studies, and describe challenges and opportunities for future advances, particularly at the dawn of “precision medicine.” Together, we call on administrative bodies, funding agencies, the research community, and the public at large for increased support for research on the genetic basis of psychiatric disorders in the Chinese population. In our opinion, increased public awareness and effective collaborative research hold the keys to the future of psychiatric genetics in China.
Collapse
Affiliation(s)
- Chunyu Liu
- State Key Laboratory of Medical Genetics of China, Central South University, Changsha, China
- Department of Psychiatry, University of Illinois at Chicago, Chicago, United States of America
| | - David Saffen
- Depatement of Cellular and Genetic Medicine, Fudan University, Shanghai, China
| | - Thomas G Schulze
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-Universität, Göttingen, Germany
- Institute of Psychiatric Phenomics and Genomics, Ludwig Maximilians-University, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health Mannheim, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Margit Burmeister
- Molecular and Behavioral Neuroscience Institute, Departments of Psychiatry, Human Genetics and Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan, United States of America
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Pak Chung Sham
- Department of Psychiatry, University of Hong Kong, Pokfulam, Hong Kong
| | - 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, China
| | - Po-Hsiu Kuo
- Department of Public Health and Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chao Chen
- State Key Laboratory of Medical Genetics of China, Central South University, Changsha, China
| | - Yu An
- Institute of Biomedical Sciences and MOE Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai, China
| | - Jiapei Dai
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, China
| | - Weihua Yue
- Key Laboratory of Mental Health, Ministry of Health, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
| | - Miao Xin Li
- Department of Psychiatry, University of Hong Kong, Pokfulam, Hong Kong
| | - Hong Xue
- Division of Life Science and Applied Genomics Center, The Hong Kong University of Science & Technology, Hong Kong, China
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology and Kunming Primate Research Centre, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Li Chen
- Depatement of Cellular and Genetic Medicine, Fudan University, Shanghai, China
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Mingqi Qiao
- Institute of Traditional Chinese Medicine theory, School of Basic Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Tiebang Liu
- Shenzhen Kang Ning Hospital, No.1080, Cuizhu Street, Luohu District, Shenzhen, Guangdong, 518020, China
| | - Kun Xia
- State Key Laboratory of Medical Genetics of China, Central South University, Changsha, China
- School of Life Sciences, Central South University, Changsha, China
| | - Raymond C K Chan
- Neuropsychology and Applied Cognitive Neuroscience Laboratory, Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences
| |
Collapse
|
2907
|
Guillemin N, Horvatić A, Kuleš J, Galan A, Mrljak V, Bhide M. Omics approaches to probe markers of disease resistance in animal sciences. MOLECULAR BIOSYSTEMS 2016; 12:2036-46. [DOI: 10.1039/c6mb00220j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Disease pathways can be explained into a list of biomarkers at different scales to develop applications.
Collapse
Affiliation(s)
- N. Guillemin
- ERA Chair FP7
- Internal Diseases
- Faculty of Veterinary Medicine
- 10000 Zagreb
- Croatia
| | - A. Horvatić
- ERA Chair FP7
- Internal Diseases
- Faculty of Veterinary Medicine
- 10000 Zagreb
- Croatia
| | - J. Kuleš
- ERA Chair FP7
- Internal Diseases
- Faculty of Veterinary Medicine
- 10000 Zagreb
- Croatia
| | - A. Galan
- ERA Chair FP7
- Internal Diseases
- Faculty of Veterinary Medicine
- 10000 Zagreb
- Croatia
| | - Vladimir Mrljak
- ERA Chair FP7
- Internal Diseases
- Faculty of Veterinary Medicine
- 10000 Zagreb
- Croatia
| | - M. Bhide
- ERA Chair FP7
- Internal Diseases
- Faculty of Veterinary Medicine
- 10000 Zagreb
- Croatia
| |
Collapse
|
2908
|
Zhang B, Tran L, Emilsson V, Zhu J. Characterization of Genetic Networks Associated with Alzheimer's Disease. Methods Mol Biol 2016; 1303:459-77. [PMID: 26235085 DOI: 10.1007/978-1-4939-2627-5_28] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
At the molecular level, the genetics of complex disease such as Alzheimer's disease (AD) manifests itself as series of alterations in the molecular interactions in pathways and networks that define biological processes underlying the pathophysiological states of disease. While large-scale genome-wide association (GWA) studies of late-onset alzheimer's disease (LOAD) have uncovered prominent genomic regions linked to the disease, the cause for the vast majority of LOAD cases still remains unknown. Increasingly available large-scale genomic and genetic data related to LOAD has made it possible to comprehensively uncover the mechanisms causally lined to LOAD in a completely data-driven manner. Here we review the various aspects of systems/network biology approaches and methodology in constructing genetic networks associated with AD from large sampling of postmortem brain tissues. We describe in detail a multiscale network modeling approach (MNMA) that integrates interaction and causal gene networks to analyze large-scale DNA, gene expression and pathophysiological data from multiple post-mortem brain regions of LOAD patients as well non-demented normal controls. MNMA first employs weighted gene co-expression network analysis (WGCNA) to construct multi-tissue networks that simultaneously capture intra-tissue and inter-tissue gene-gene interactions and then quantifies the change in connectivity among highly co-expressed genes in LOAD with respect to the normal state. Co-expressed gene modules are then rank ordered by relevance to pathophysiological traits and enrichment of genes differentially expressed in LOAD. Causal regulatory relationships among the genes in each module are then determined by a Bayesian network inference framework that is used to formally integrate genetic and gene expression information. MNMA has uncovered a massive remodeling of network structures in LOAD and identified novel subnetworks and key regulators that are causally linked to LOAD. In the end, we will outline the challenges in systems/network approaches to LOAD.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY, 10029, USA,
| | | | | | | |
Collapse
|
2909
|
Deming Y, Xia J, Cai Y, Lord J, Holmans P, Bertelsen S, Holtzman D, Morris JC, Bales K, Pickering EH, Kauwe J, Goate A, Cruchaga C. A potential endophenotype for Alzheimer's disease: cerebrospinal fluid clusterin. Neurobiol Aging 2016; 37:208.e1-208.e9. [PMID: 26545630 PMCID: PMC5118651 DOI: 10.1016/j.neurobiolaging.2015.09.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/12/2015] [Indexed: 12/30/2022]
Abstract
Genome-wide association studies have associated clusterin (CLU) variants with Alzheimer's disease (AD). However, the role of CLU on AD pathogenesis is not totally understood. We used cerebrospinal fluid (CSF) and plasma CLU levels as endophenotypes for genetic studies to understand the role of CLU in AD. CSF, but not plasma, CLU levels were significantly associated with AD status and CSF tau/amyloid-beta ratio, and highly correlated with CSF apolipoprotein E (APOE) levels. Several loci showed almost genome-wide significant associations including LINC00917 (p = 3.98 × 10(-7)) and interleukin 6 (IL6, p = 9.94 × 10(-6), in the entire data set and in the APOE ε4- individuals p = 7.40 × 10(-8)). Gene ontology analyses suggest that CSF CLU levels may be associated with wound healing and immune response which supports previous functional studies that demonstrated an association between CLU and IL6. CLU may play a role in AD by influencing immune system changes that have been observed in AD or by disrupting healing after neurodegeneration.
Collapse
Affiliation(s)
- Yuetiva Deming
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Jian Xia
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Yefei Cai
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Jenny Lord
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter Holmans
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Center for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cardiff, UK
| | - Sarah Bertelsen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - David Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Kelly Bales
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer, Inc., Groton, CT, USA
| | - Eve H Pickering
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer, Inc., Groton, CT, USA
| | - John Kauwe
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Alison Goate
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
2910
|
Hong S, Dissing-Olesen L, Stevens B. New insights on the role of microglia in synaptic pruning in health and disease. Curr Opin Neurobiol 2015; 36:128-34. [PMID: 26745839 DOI: 10.1016/j.conb.2015.12.004] [Citation(s) in RCA: 411] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/09/2015] [Accepted: 12/10/2015] [Indexed: 01/17/2023]
Abstract
Recent genome-wide association studies implicate microglia in Alzheimer's disease (AD) pathogenesis; however, their biological significance remains poorly understood. Synapse loss is a significant correlate of cognitive decline that serves as a critical hallmark of AD and other neurodegenerative diseases; however, mechanisms underlying synaptic vulnerability remain elusive. Emerging research on microglia function in the healthy brain is providing new insight into fundamental roles of microglia and immune molecules in brain wiring. Among their many roles, microglia prune developing synapses and regulate synaptic plasticity and function. Here, we review and discuss how this emerging work may provide new insight into how disruptions in microglia-synapse interactions could contribute to synapse loss and dysfunction, and consequently cognitive impairment, in AD.
Collapse
Affiliation(s)
- Soyon Hong
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lasse Dissing-Olesen
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Beth Stevens
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
2911
|
Cholerton B, Larson EB, Quinn JF, Zabetian CP, Mata IF, Keene CD, Flanagan M, Crane PK, Grabowski TJ, Montine KS, Montine TJ. Precision Medicine: Clarity for the Complexity of Dementia. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:500-6. [PMID: 26724389 DOI: 10.1016/j.ajpath.2015.12.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 11/08/2015] [Accepted: 12/03/2015] [Indexed: 01/11/2023]
Abstract
Three key elements to precision medicine are stratification by risk, detection of pathophysiological processes as early as possible (even before clinical presentation), and alignment of mechanism of action of intervention(s) with an individual's molecular driver(s) of disease. Used for decades in the management of some rare diseases and now gaining broad currency in cancer care, a precision medicine approach is beginning to be adapted to cognitive impairment and dementia. This review focuses on the application of precision medicine to address the clinical and biological complexity of two common neurodegenerative causes of dementia: Alzheimer disease and Parkinson disease.
Collapse
Affiliation(s)
- Brenna Cholerton
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington
| | - Eric B Larson
- Group Health Research Institute, Seattle, Washington
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, Oregon; Portland Veterans Affairs Medical Center, Portland, Oregon
| | - Cyrus P Zabetian
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington; Parkinson's Disease Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington; Department of Neurology, University of Washington, Seattle, Washington
| | - Ignacio F Mata
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington; Department of Neurology, University of Washington, Seattle, Washington
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington
| | - Margaret Flanagan
- Department of Pathology, University of Washington, Seattle, Washington
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, Washington
| | - Thomas J Grabowski
- Department of Neurology, University of Washington, Seattle, Washington; Department of Radiology, University of Washington, Seattle, Washington
| | | | - Thomas J Montine
- Department of Pathology, University of Washington, Seattle, Washington.
| |
Collapse
|
2912
|
Rollo JL, Banihashemi N, Vafaee F, Crawford JW, Kuncic Z, Holsinger RMD. Unraveling the mechanistic complexity of Alzheimer's disease through systems biology. Alzheimers Dement 2015; 12:708-18. [PMID: 26703952 DOI: 10.1016/j.jalz.2015.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 08/18/2015] [Accepted: 10/21/2015] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is a complex, multifactorial disease that has reached global epidemic proportions. The challenge remains to fully identify its underlying molecular mechanisms that will enable development of accurate diagnostic tools and therapeutics. Conventional experimental approaches that target individual or small sets of genes or proteins may overlook important parts of the regulatory network, which limits the opportunity of identifying multitarget interventions. Our perspective is that a more complete insight into potential treatment options for AD will only be made possible through studying the disease as a system. We propose an integrative systems biology approach that we argue has been largely untapped in AD research. We present key publications to demonstrate the value of this approach and discuss the potential to intensify research efforts in AD through transdisciplinary collaboration. We highlight challenges and opportunities for significant breakthroughs that could be made if a systems biology approach is fully exploited.
Collapse
Affiliation(s)
- Jennifer L Rollo
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Laboratory of Molecular Neuroscience, Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia; Department of Molecular Neuroscience, Institute of Neurology, University College of London, London, UK.
| | - Nahid Banihashemi
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Fatemeh Vafaee
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| | | | - Zdenka Kuncic
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; School of Physics, The University of Sydney, Sydney, NSW, Australia
| | - R M Damian Holsinger
- Laboratory of Molecular Neuroscience, Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia; Discipline of Biomedical Science, School of Medical Sciences, Sydney Medical School, The University of Sydney, Lidcombe, NSW, Australia
| |
Collapse
|
2913
|
Jiao B, Liu X, Zhou L, Wang MH, Zhou Y, Xiao T, Zhang W, Sun R, Waye MMY, Tang B, Shen L. Polygenic Analysis of Late-Onset Alzheimer's Disease from Mainland China. PLoS One 2015; 10:e0144898. [PMID: 26680604 PMCID: PMC4683047 DOI: 10.1371/journal.pone.0144898] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 11/24/2015] [Indexed: 01/14/2023] Open
Abstract
Recently, a number of single nucleotide polymorphisms (SNPs) were identified to be associated with late-onset Alzheimer disease (LOAD) through genome-wide association study data. Identification of SNP-SNP interaction played an important role in better understanding genetic basis of LOAD. In this study, fifty-eight SNPs were screened in a cohort of 229 LOAD cases and 318 controls from mainland China, and their interaction was evaluated by a series of analysis methods. Seven risk SNPs and six protective SNPs were identified to be associated with LOAD. Risk SNPs included rs9331888 (CLU), rs6691117 (CR1), rs4938933 (MS4A), rs9349407 (CD2AP), rs1160985 (TOMM40), rs4945261 (GAB2) and rs5984894 (PCDH11X); Protective SNPs consisted of rs744373 (BIN1), rs1562990 (MS4A), rs597668 (EXOC3L2), rs9271192 (HLA-DRB5/DRB1), rs157581 and rs11556505 (TOMM40). Among positive SNPs presented above, we found the interaction between rs4938933 (risk) and rs1562990 (protective) in MS4A weakened their each effect for LOAD; for three significant SNPs in TOMM40, their cumulative interaction induced the two protective SNPs effects lost and made the risk SNP effect aggravate for LOAD. Finally, we found rs6656401-rs3865444 (CR1-CD33) pairs were significantly associated with decreasing LOAD risk, while rs28834970-rs6656401 (PTK2B-CR1), and rs28834970-rs6656401 (PTK2B-CD33) were associated with increasing LOAD risk. In a word, our study indicates that SNP-SNP interaction existed in the same gene or cross different genes, which could weaken or aggravate their initial single effects for LOAD.
Collapse
Affiliation(s)
- Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyan Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Maggie Haitian Wang
- Division of Biostatistics, School of Public Health and Primary Care, the Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | - Yafang Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Tingting Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Weiwei Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Rui Sun
- Division of Biostatistics, School of Public Health and Primary Care, the Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | - Mary Miu Yee Waye
- School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- State Key Laboratory of Medical Genetics, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- State Key Laboratory of Medical Genetics, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| |
Collapse
|
2914
|
Haas LT, Salazar SV, Kostylev MA, Um JW, Kaufman AC, Strittmatter SM. Metabotropic glutamate receptor 5 couples cellular prion protein to intracellular signalling in Alzheimer's disease. Brain 2015; 139:526-46. [PMID: 26667279 DOI: 10.1093/brain/awv356] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 10/17/2015] [Indexed: 01/27/2023] Open
Abstract
Alzheimer's disease-related phenotypes in mice can be rescued by blockade of either cellular prion protein or metabotropic glutamate receptor 5. We sought genetic and biochemical evidence that these proteins function cooperatively as an obligate complex in the brain. We show that cellular prion protein associates via transmembrane metabotropic glutamate receptor 5 with the intracellular protein mediators Homer1b/c, calcium/calmodulin-dependent protein kinase II, and the Alzheimer's disease risk gene product protein tyrosine kinase 2 beta. Coupling of cellular prion protein to these intracellular proteins is modified by soluble amyloid-β oligomers, by mouse brain Alzheimer's disease transgenes or by human Alzheimer's disease pathology. Amyloid-β oligomer-triggered phosphorylation of intracellular protein mediators and impairment of synaptic plasticity in vitro requires Prnp-Grm5 genetic interaction, being absent in transheterozygous loss-of-function, but present in either single heterozygote. Importantly, genetic coupling between Prnp and Grm5 is also responsible for signalling, for survival and for synapse loss in Alzheimer's disease transgenic model mice. Thus, the interaction between metabotropic glutamate receptor 5 and cellular prion protein has a central role in Alzheimer's disease pathogenesis, and the complex is a potential target for disease-modifying intervention.
Collapse
Affiliation(s)
- Laura T Haas
- 1 Cellular Neuroscience, Neurodegeneration and Repair Program, Department of Neurology, Yale University School of Medicine, New Haven, CT 06536, USA 2 Graduate School of Cellular and Molecular Neuroscience, University of Tuebingen, D-72074 Tuebingen, Germany
| | - Santiago V Salazar
- 1 Cellular Neuroscience, Neurodegeneration and Repair Program, Department of Neurology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Mikhail A Kostylev
- 1 Cellular Neuroscience, Neurodegeneration and Repair Program, Department of Neurology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Ji Won Um
- 1 Cellular Neuroscience, Neurodegeneration and Repair Program, Department of Neurology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Adam C Kaufman
- 1 Cellular Neuroscience, Neurodegeneration and Repair Program, Department of Neurology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Stephen M Strittmatter
- 1 Cellular Neuroscience, Neurodegeneration and Repair Program, Department of Neurology, Yale University School of Medicine, New Haven, CT 06536, USA
| |
Collapse
|
2915
|
Montañola A, de Retana SF, López-Rueda A, Merino-Zamorano C, Penalba A, Fernández-Álvarez P, Rodríguez-Luna D, Malagelada A, Pujadas F, Montaner J, Hernández-Guillamon M. ApoA1, ApoJ and ApoE Plasma Levels and Genotype Frequencies in Cerebral Amyloid Angiopathy. Neuromolecular Med 2015; 18:99-108. [PMID: 26661731 DOI: 10.1007/s12017-015-8381-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/17/2015] [Indexed: 01/26/2023]
Abstract
The involvement of apolipoproteins, such as the ApoE4 isoform, in Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA) highlights the fact that certain lipid carriers may participate in soluble β-amyloid (Aβ) transport. Our general aim was to characterize the soluble levels of the apolipoproteins apoE, apoA1 and apoJ/clusterin and their genotype status in patients with CAA. We analyzed the genotypes frequency of APOA1 (rs5069, rs670), CLU (rs11136000, rs1532278, rs7012010, rs9331888) and APOE (rs429358, rs7412) in a cohort of patients with CAA-associated intracerebral hemorrhage (ICH) (n = 59) and compared the results with those from hypertension-associated ICH (n = 42), AD patients (n = 73) and controls (n = 88). In a subgroup of patients, we also determined the plasma concentrations of apoE, apoA1 and apoJ/clusterin. We found increased plasma apoJ/clusterin levels in CAA patients compared to AD patients or controls after adjusting for sex and age (CAA vs. controls, p = 0.033; CAA vs. AD, p = 0.013). ApoA1 levels were not altered between groups, although a strong correlation was observed between plasma Aβ(1-40) and apoA1 among CAA patients (r = 0.583, p = 0.007). Regarding plasma apoE concentration, a robust association between circulating levels and genotype status was confirmed (p < 0.001). Whereas the APOE4 frequency was higher in AD (p < 0.001) and CAA (p = 0.013), the APOA1 and CLU genotypes were not different among groups. In the CAA cohort, the risk-linked CLU variant (C) rs11136000 was associated with white matter hyperintensities (p = 0.045) and the presence of lobar microbleeds (p = 0.023) on MRI. In summary, our findings suggest that apoA1 may act as a physiological transporter of Aβ(1-40) and that apoJ/clusterin appears to be a chaperone related to distinctive lesions in CAA brains.
Collapse
Affiliation(s)
- Alex Montañola
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Sofía Fernández de Retana
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Antonio López-Rueda
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Cristina Merino-Zamorano
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Anna Penalba
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | | | - David Rodríguez-Luna
- Neurovascular Unit, Neurology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Ana Malagelada
- Dementia Unit, Neurology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Francesc Pujadas
- Dementia Unit, Neurology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Neurovascular Unit, Neurology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| |
Collapse
|
2916
|
Cao S, Yu L, Mao J, Wang Q, Ruan J. Uncovering the Molecular Mechanism of Actions between Pharmaceuticals and Proteins on the AD Network. PLoS One 2015; 10:e0144387. [PMID: 26650760 PMCID: PMC4674063 DOI: 10.1371/journal.pone.0144387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 11/17/2015] [Indexed: 11/18/2022] Open
Abstract
This study begins with constructing the mini metabolic networks (MMNs) of beta amyloid (Aβ) and acetylcholine (ACh) which stimulate the Alzheimer's Disease (AD). Then we generate the AD network by incorporating MMNs of Aβ and ACh, and other MMNs of stimuli of AD. The panel of proteins contains 49 enzymes/receptors on the AD network which have the 3D-structure in PDB. The panel of drugs is formed by 5 AD drugs and 5 AD nutraceutical drugs, and 20 non-AD drugs. All of these complexes formed by these 30 drugs and 49 proteins are transformed into dyadic arrays. Utilizing the prior knowledge learned from the drug panel, we propose a statistical classification (dry-lab). According to the wet-lab for the complex of amiloride and insulin degrading enzyme, and the complex of amiloride and neutral endopeptidase, we are confident that this dry-lab is reliable. As the consequences of the dry-lab, we discover many interesting implications. Especially, we show that possible causes of Tacrine, donepezil, galantamine and huperzine A cannot improve the level of ACh which is against to their original design purpose but they still prevent AD to be worse as Aβ deposition appeared. On the other hand, we recommend Miglitol and Atenolol as the safe and potent drugs to improve the level of ACh before Aβ deposition appearing. Moreover, some nutrients such as NADH and Vitamin E should be controlled because they may harm health if being used in wrong way and wrong time. Anyway, the insights shown in this study are valuable to be developed further.
Collapse
Affiliation(s)
- Shujuan Cao
- College of Mathematical Sciences and LPMC, Nankai University, Tianjin, PRC
| | - Liang Yu
- College of Mathematical Sciences and LPMC, Nankai University, Tianjin, PRC
| | - Jingyuan Mao
- Internal Medicine for the Heart, Tianjin University of Traditional Chinese Medicine, Tianjin, PRC
| | - Quan Wang
- National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Science, Beijing, PRC
- High Throughput Molecular Drug Discovery Center, Tianjin International Joint Academy of Biomedicine, TEDA, Tianjin, PRC
| | - Jishou Ruan
- College of Mathematical Sciences and LPMC, Nankai University, Tianjin, PRC
- State Key Laboratory for Medical Chemical and Biology, Nankai University, Tianjin, PRC
- * E-mail:
| |
Collapse
|
2917
|
Del-Aguila JL, Fernández MV, Jimenez J, Black K, Ma S, Deming Y, Carrell D, Saef B, Howells B, Budde J, Cruchaga C. Role of ABCA7 loss-of-function variant in Alzheimer's disease: a replication study in European-Americans. Alzheimers Res Ther 2015; 7:73. [PMID: 26654793 PMCID: PMC4675010 DOI: 10.1186/s13195-015-0154-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/12/2015] [Indexed: 11/10/2022]
Abstract
INTRODUCTION A recent study found a significant increase of ABCA7 loss-of-function variants in Alzheimer's disease (AD) cases compared to controls. Some variants were located on noncoding regions, but it was demonstrated that they affect splicing. Here, we try to replicate the association between AD risk and ABCA7 loss-of-function variants at both the single-variant and gene level in a large and well-characterized European American dataset. METHODS We genotyped the GWAS common variant and four rare variants previously reported for ABCA7 in 3476 European-Americans. RESULTS We were not able to replicate the association at the single-variant level, likely due to a lower effect size on the European American population which led to limited statistical power. However, we did replicate the association at the gene level; we found a significant enrichment of ABCA7 loss-of-function variants in AD cases compared to controls (P = 0.0388; odds ratio =1.54). We also confirmed that the association of the loss-of-function variants is independent of the previously reported genome-wide association study signal. CONCLUSIONS Although the effect size for the association of ABCA7 loss-of-function variants with AD risk is lower in our study (odds ratio = 1.54) compared to the original report (odds ratio = 2.2), the replication of the findings of the original report provides a stronger foundation for future functional applications. The data indicate that different independent signals that modify risk for complex traits may exist on the same locus. Additionally, our results suggest that replication of rare-variant studies should be performed at the gene level rather than focusing on a single variant.
Collapse
Affiliation(s)
- Jorge L Del-Aguila
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO, 63110, USA
- The Hope Center for Neurological Disorders, BJC Institute of Health, 425 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Maria Victoria Fernández
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO, 63110, USA
- The Hope Center for Neurological Disorders, BJC Institute of Health, 425 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Jessica Jimenez
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO, 63110, USA
- The Hope Center for Neurological Disorders, BJC Institute of Health, 425 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Kathleen Black
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO, 63110, USA
- The Hope Center for Neurological Disorders, BJC Institute of Health, 425 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Shengmei Ma
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO, 63110, USA
- The Hope Center for Neurological Disorders, BJC Institute of Health, 425 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Yuetiva Deming
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO, 63110, USA
- The Hope Center for Neurological Disorders, BJC Institute of Health, 425 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - David Carrell
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO, 63110, USA
| | - Ben Saef
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO, 63110, USA
- The Hope Center for Neurological Disorders, BJC Institute of Health, 425 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Bill Howells
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO, 63110, USA
- The Hope Center for Neurological Disorders, BJC Institute of Health, 425 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - John Budde
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO, 63110, USA
- The Hope Center for Neurological Disorders, BJC Institute of Health, 425 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue B8134, St. Louis, MO, 63110, USA.
- The Hope Center for Neurological Disorders, BJC Institute of Health, 425 S. Euclid Ave, St. Louis, MO, 63110, USA.
| |
Collapse
|
2918
|
Genomic Discoveries and Personalized Medicine in Neurological Diseases. Pharmaceutics 2015; 7:542-53. [PMID: 26690205 PMCID: PMC4695833 DOI: 10.3390/pharmaceutics7040542] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/30/2015] [Accepted: 12/02/2015] [Indexed: 12/22/2022] Open
Abstract
In the past decades, we have witnessed dramatic changes in clinical diagnoses and treatments due to the revolutions of genomics and personalized medicine. Undoubtedly we also met many challenges when we use those advanced technologies in drug discovery and development. In this review, we describe when genomic information is applied in personal healthcare in general. We illustrate some case examples of genomic discoveries and promising personalized medicine applications in the area of neurological disease particular. Available data suggest that individual genomics can be applied to better treat patients in the near future.
Collapse
|
2919
|
Hofmann-Apitius M, Ball G, Gebel S, Bagewadi S, de Bono B, Schneider R, Page M, Kodamullil AT, Younesi E, Ebeling C, Tegnér J, Canard L. Bioinformatics Mining and Modeling Methods for the Identification of Disease Mechanisms in Neurodegenerative Disorders. Int J Mol Sci 2015; 16:29179-206. [PMID: 26690135 PMCID: PMC4691095 DOI: 10.3390/ijms161226148] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/10/2015] [Accepted: 11/12/2015] [Indexed: 12/22/2022] Open
Abstract
Since the decoding of the Human Genome, techniques from bioinformatics, statistics, and machine learning have been instrumental in uncovering patterns in increasing amounts and types of different data produced by technical profiling technologies applied to clinical samples, animal models, and cellular systems. Yet, progress on unravelling biological mechanisms, causally driving diseases, has been limited, in part due to the inherent complexity of biological systems. Whereas we have witnessed progress in the areas of cancer, cardiovascular and metabolic diseases, the area of neurodegenerative diseases has proved to be very challenging. This is in part because the aetiology of neurodegenerative diseases such as Alzheimer´s disease or Parkinson´s disease is unknown, rendering it very difficult to discern early causal events. Here we describe a panel of bioinformatics and modeling approaches that have recently been developed to identify candidate mechanisms of neurodegenerative diseases based on publicly available data and knowledge. We identify two complementary strategies-data mining techniques using genetic data as a starting point to be further enriched using other data-types, or alternatively to encode prior knowledge about disease mechanisms in a model based framework supporting reasoning and enrichment analysis. Our review illustrates the challenges entailed in integrating heterogeneous, multiscale and multimodal information in the area of neurology in general and neurodegeneration in particular. We conclude, that progress would be accelerated by increasing efforts on performing systematic collection of multiple data-types over time from each individual suffering from neurodegenerative disease. The work presented here has been driven by project AETIONOMY; a project funded in the course of the Innovative Medicines Initiative (IMI); which is a public-private partnership of the European Federation of Pharmaceutical Industry Associations (EFPIA) and the European Commission (EC).
Collapse
Affiliation(s)
- Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Institutszentrum Birlinghoven, Sankt Augustin D-53754, Germany.
- Rheinische Friedrich-Wilhelms-Universitaet Bonn, University of Bonn, Bonn 53113, Germany.
| | - Gordon Ball
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine, and Unit of Clinical Epidemiology, Karolinska University Hospital, Stockholm SE-171 77, Sweden.
- Science for Life Laboratories, Karolinska Institutet, Stockholm SE-171 77, Sweden.
| | - Stephan Gebel
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7, avenue des Hauts-Fourneaux, Esch-sur-Alzette L-4362, Luxembourg.
| | - Shweta Bagewadi
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Institutszentrum Birlinghoven, Sankt Augustin D-53754, Germany.
| | - Bernard de Bono
- Institute of Health Informatics, University College London, London NW1 2DA, UK.
- Auckland Bioengineering Institute, University of Auckland, Symmonds Street, Auckland 1142, New Zealand.
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7, avenue des Hauts-Fourneaux, Esch-sur-Alzette L-4362, Luxembourg.
| | - Matt Page
- Translational Bioinformatics, UCB Pharma, 216 Bath Rd, Slough SL1 3WE, UK.
| | - Alpha Tom Kodamullil
- Rheinische Friedrich-Wilhelms-Universitaet Bonn, University of Bonn, Bonn 53113, Germany.
| | - Erfan Younesi
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Institutszentrum Birlinghoven, Sankt Augustin D-53754, Germany.
| | - Christian Ebeling
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Institutszentrum Birlinghoven, Sankt Augustin D-53754, Germany.
| | - Jesper Tegnér
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine, and Unit of Clinical Epidemiology, Karolinska University Hospital, Stockholm SE-171 77, Sweden.
- Science for Life Laboratories, Karolinska Institutet, Stockholm SE-171 77, Sweden.
| | - Luc Canard
- Translational Science Unit, SANOFI Recherche & Développement, 1 Avenue Pierre Brossolette, Chilly-Mazarin Cedex 91385, France.
| |
Collapse
|
2920
|
Abstract
Late-onset Alzheimer's disease (AD), a highly prevalent neurodegenerative disorder characterized by progressive deterioration in cognition, function and behavior terminating in incapacity and death, is a clinically and pathologically heterogeneous disease with a substantial heritable component. During the past 5 years, the technological developments in next-generation high-throughput genome technologies have led to the identification of more than 20 novel susceptibility loci for AD, and have implicated specific pathways in the disease, in particular intracellular trafficking/endocytosis, inflammation and immune response and lipid metabolism. These observations have significantly advanced our understanding of underlying pathogenic mechanisms and potential therapeutic targets. This review article summarizes these recent advances in AD genomics and discusses the value of identified susceptibility loci for diagnosis and prognosis of AD.
Collapse
|
2921
|
Benedet AL, Labbe A, Lemay P, Zimmer ER, Pascoal TA, Leuzy A, Mathotaarachchi S, Mohades S, Shin M, Dionne-Laporte A, Beaudry T, Picard C, Gauthier S, Poirier J, Rouleau G, Rosa-Neto P. Epistasis analysis links immune cascades and cerebral amyloidosis. J Neuroinflammation 2015; 12:227. [PMID: 26626881 PMCID: PMC4666175 DOI: 10.1186/s12974-015-0436-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/16/2015] [Indexed: 12/21/2022] Open
Abstract
Background Several lines of evidence suggest the involvement of neuroinflammatory changes in Alzheimer’s disease (AD) pathophysiology such as amyloidosis and neurodegeneration. In fact, genome-wide association studies (GWAS) have shown a link between genes involved in neuroinflammation and AD. In order to further investigate whether interactions between candidate genetic variances coding for neuroinflammatory molecules are associated with brain amyloid β (Aβ) fibrillary accumulation, we conducted an epistasis analysis on a pool of genes associated with molecular mediators of inflammation. Methods [18F]Florbetapir positron emission tomography (PET) imaging was employed to assess brain Aβ levels in 417 participants from ADNI-GO/2 and posteriorly 174 from ADNI-1. IL-1β, IL4, IL6, IL6r, IL10, IL12, IL18, C5, and C9 genes were chosen based on previous studies conducted in AD patients. Using the [18F]florbetapir standardized uptake value ratio (SUVR) as a quantitative measure of fibrillary Aβ, epistasis analyses were performed between two sets of markers of immune-related genes using gender, diagnosis, and apolipoprotein E (APOE) as covariates. Voxel-based analyses were also conducted. The results were corrected for multiple comparison tests. Cerebrospinal fluid (CSF) Aβ1-42/phosphorylated tau (p-tau) ratio concentrations were used to confirm such associations. Results Epistasis analysis unveiled two significant single nucleotide polymorphism (SNP)-SNP interactions (false discovery rate (FDR) threshold 0.1), both interactions between C9 gene (rs261752) and IL6r gene (rs4240872, rs7514452). In a combined sample, the interactions were confirmed (p ≤ 10–5) and associated with amyloid accumulation within cognitively normal and AD spectrum groups. Voxel-based analysis corroborated initial findings. CSF biomarker (Aβ1-42/p-tau) confirmed the genetic interaction. Additionally, rs4240872 and rs7514452 SNPs were shown to be associated with CSF and plasma concentrations of IL6r protein. Conclusions Certain allele combinations involving IL6r and C9 genes are associated with Aβ burden in the brain. Hypothesis-driven search for epistasis is a valuable strategy for investigating imaging endophenotypes in complex neurodegenerative diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0436-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andréa L Benedet
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, 6825 LaSalle Blvd, H4H 1R3, Montreal, QC, Canada. .,CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil.
| | - Aurélie Labbe
- Douglas Hospital Research Centre, McGill University, Montreal, Canada. .,Department of Epidemiology, Biostatistics & Occupational Health, McGill University, Montreal, Canada. .,Department of Psychiatry, McGill University, Montreal, Canada.
| | - Philippe Lemay
- Department of Biochemistry, Université de Montréal, Montréal, Canada.
| | - Eduardo R Zimmer
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, 6825 LaSalle Blvd, H4H 1R3, Montreal, QC, Canada. .,Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil. .,Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, 6825 LaSalle Blvd, H4H 1R3, Montreal, QC, Canada.
| | - Antoine Leuzy
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, 6825 LaSalle Blvd, H4H 1R3, Montreal, QC, Canada. .,Department of NVS, Center for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden. .,Alzheimer's Disease Research Unit, McGill University Research Centre for Studies in Aging, McGill University, Montreal, Canada.
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, 6825 LaSalle Blvd, H4H 1R3, Montreal, QC, Canada.
| | - Sara Mohades
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, 6825 LaSalle Blvd, H4H 1R3, Montreal, QC, Canada.
| | - Monica Shin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, 6825 LaSalle Blvd, H4H 1R3, Montreal, QC, Canada.
| | - Alexandre Dionne-Laporte
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada. .,Montreal Neurological Institute, Montreal, Canada.
| | - Thomas Beaudry
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, 6825 LaSalle Blvd, H4H 1R3, Montreal, QC, Canada.
| | - Cynthia Picard
- Douglas Hospital Research Centre, McGill University, Montreal, Canada.
| | - Serge Gauthier
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.
| | - Judes Poirier
- Douglas Hospital Research Centre, McGill University, Montreal, Canada. .,Alzheimer's Disease Research Unit, McGill University Research Centre for Studies in Aging, McGill University, Montreal, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.
| | - Guy Rouleau
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada. .,Montreal Neurological Institute, Montreal, Canada.
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, 6825 LaSalle Blvd, H4H 1R3, Montreal, QC, Canada. .,Alzheimer's Disease Research Unit, McGill University Research Centre for Studies in Aging, McGill University, Montreal, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada. .,Montreal Neurological Institute, Montreal, Canada.
| | | |
Collapse
|
2922
|
Barber RC, Phillips NR, Tilson JL, Huebinger RM, Shewale SJ, Koenig JL, Mitchel JS, O’Bryant SE, Waring SC, Diaz-Arrastia R, Chasse S, Wilhelmsen KC, for the Alzheimer’s Disease Neuroimaging Initiative and the Texas Alzheimer’s Research and Care Consortium. Can Genetic Analysis of Putative Blood Alzheimer's Disease Biomarkers Lead to Identification of Susceptibility Loci? PLoS One 2015; 10:e0142360. [PMID: 26625115 PMCID: PMC4666664 DOI: 10.1371/journal.pone.0142360] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/21/2015] [Indexed: 01/22/2023] Open
Abstract
Although 24 Alzheimer’s disease (AD) risk loci have been reliably identified, a large portion of the predicted heritability for AD remains unexplained. It is expected that additional loci of small effect will be identified with an increased sample size. However, the cost of a significant increase in Case-Control sample size is prohibitive. The current study tests whether exploring the genetic basis of endophenotypes, in this case based on putative blood biomarkers for AD, can accelerate the identification of susceptibility loci using modest sample sizes. Each endophenotype was used as the outcome variable in an independent GWAS. Endophenotypes were based on circulating concentrations of proteins that contributed significantly to a published blood-based predictive algorithm for AD. Endophenotypes included Monocyte Chemoattractant Protein 1 (MCP1), Vascular Cell Adhesion Molecule 1 (VCAM1), Pancreatic Polypeptide (PP), Beta2 Microglobulin (B2M), Factor VII (F7), Adiponectin (ADN) and Tenascin C (TN-C). Across the seven endophenotypes, 47 SNPs were associated with outcome with a p-value ≤1x10-7. Each signal was further characterized with respect to known genetic loci associated with AD. Signals for several endophenotypes were observed in the vicinity of CR1, MS4A6A/MS4A4E, PICALM, CLU, and PTK2B. The strongest signal was observed in association with Factor VII levels and was located within the F7 gene. Additional signals were observed in MAP3K13, ZNF320, ATP9B and TREM1. Conditional regression analyses suggested that the SNPs contributed to variation in protein concentration independent of AD status. The identification of two putatively novel AD loci (in the Factor VII and ATP9B genes), which have not been located in previous studies despite massive sample sizes, highlights the benefits of an endophenotypic approach for resolving the genetic basis for complex diseases. The coincidence of several of the endophenotypic signals with known AD loci may point to novel genetic interactions and should be further investigated.
Collapse
Affiliation(s)
- Robert C. Barber
- Department of Molecular & Medical Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Institute for Aging and Alzheimer’s Disease Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- * E-mail:
| | - Nicole R. Phillips
- Department of Biology, University of Dallas, Dallas, Texas, United States of America
| | - Jeffrey L. Tilson
- Renaissance Computing Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Ryan M. Huebinger
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Shantanu J. Shewale
- Department of Molecular & Medical Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Jessica L. Koenig
- Department of Molecular & Medical Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Jeffrey S. Mitchel
- Department of Molecular & Medical Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Sid E. O’Bryant
- Institute for Aging and Alzheimer’s Disease Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Department of Internal Medicine, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Stephen C. Waring
- Essentia Institute of Rural Health, Duluth, Minnesota, United States of America
| | - Ramon Diaz-Arrastia
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Rockville, Maryland, United States of America
| | - Scott Chasse
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Kirk C. Wilhelmsen
- Renaissance Computing Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Genetic Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | | |
Collapse
|
2923
|
Abstract
A treatment trial of the monoclonal anti-amyloid antibody solanezumab showed slight benefits in people with dementia due to mild Alzheimer disease. Drug effects on several neuropsychological testing outcomes were statistically significant, but the effect sizes were unlikely to manifest as meaningful functional benefits. Here, we discuss the implications and possible molecular underpinnings.
Collapse
Affiliation(s)
- Sam Gandy
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Mary Sano
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
2924
|
Gregersen PK, Klein G, Keogh M, Kern M, DeFranco M, Simpfendorfer KR, Kim SJ, Diamond B. The Genotype and Phenotype (GaP) registry: a living biobank for the analysis of quantitative traits. Immunol Res 2015; 63:107-12. [PMID: 26467974 DOI: 10.1007/s12026-015-8711-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We describe the development of the Genotype and Phenotype (GaP) Registry, a living biobank of normal volunteers who are genotyped for genetic markers related to human disease. Participants in the GaP can be recalled for hypothesis driven study of disease associated genetic variants. The GaP has facilitated functional studies of several autoimmune disease associated loci including Csk, Blk, PDRM1 (Blimp-1) and PTPN22. It is likely that expansion of such living biobank registries will play an important role in studying and understanding the function of disease associated alleles in complex disease.
Collapse
Affiliation(s)
- Peter K Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| | - Gila Klein
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Mary Keogh
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Marlena Kern
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Margaret DeFranco
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Kim R Simpfendorfer
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Sun Jung Kim
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| |
Collapse
|
2925
|
Barral S, Cheng R, Reitz C, Vardarajan B, Lee J, Kunkle B, Beecham G, Cantwell LS, Pericak-Vance MA, Farrer LA, Haines JL, Goate AM, Foroud T, Boerwinkle E, Schellenberg GD, Mayeux R. Linkage analyses in Caribbean Hispanic families identify novel loci associated with familial late-onset Alzheimer's disease. Alzheimers Dement 2015; 11:1397-1406. [PMID: 26433351 PMCID: PMC4690771 DOI: 10.1016/j.jalz.2015.07.487] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 06/08/2015] [Accepted: 07/13/2015] [Indexed: 10/23/2022]
Abstract
INTRODUCTION We performed linkage analyses in Caribbean Hispanic families with multiple late-onset Alzheimer's disease (LOAD) cases to identify regions that may contain disease causative variants. METHODS We selected 67 LOAD families to perform genome-wide linkage scan. Analysis of the linked regions was repeated using the entire sample of 282 families. Validated chromosomal regions were analyzed using joint linkage and association. RESULTS We identified 26 regions linked to LOAD (HLOD ≥3.6). We validated 13 of the regions (HLOD ≥2.5) using the entire family sample. The strongest signal was at 11q12.3 (rs2232932: HLODmax = 4.7, Pjoint = 6.6 × 10(-6)), a locus located ∼2 Mb upstream of the membrane-spanning 4A gene cluster. We additionally identified a locus at 7p14.3 (rs10255835: HLODmax = 4.9, Pjoint = 1.2 × 10(-5)), a region harboring genes associated with the nervous system (GARS, GHRHR, and NEUROD6). DISCUSSION Future sequencing efforts should focus on these regions because they may harbor familial LOAD causative mutations.
Collapse
Affiliation(s)
- Sandra Barral
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY, USA; Department of Neurology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA
| | - Rong Cheng
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY, USA
| | - Christiane Reitz
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY, USA; Department of Neurology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Department of Epidemiology, School of Public Health, Columbia University, New York, NY, USA
| | - Badri Vardarajan
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY, USA; Department of Neurology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA
| | - Joseph Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY, USA; Department of Epidemiology, School of Public Health, Columbia University, New York, NY, USA
| | - Brian Kunkle
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Gary Beecham
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Laura S Cantwell
- Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA
| | - Margaret A Pericak-Vance
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA; Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Lindsay A Farrer
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA; Department of Medicine (Biomedical Genetics), Boston University School of Medicine and Public Health, Boston, MA, USA; Department of Neurology, Boston University School of Medicine and Public Health, Boston, MA, USA; Department of Ophthalmology, Boston University School of Medicine and Public Health, Boston, MA, USA; Department of Epidemiology, Boston University School of Medicine and Public Health, Boston, MA, USA
| | - Jonathan L Haines
- Department of Epidemiology & Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Alison M Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences (EHGES), University of Texas School of Public Health at Houston, Houston, TX, USA; Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY, USA; Department of Neurology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA; Department of Epidemiology, School of Public Health, Columbia University, New York, NY, USA.
| |
Collapse
|
2926
|
Desikan RS, Schork AJ, Wang Y, Witoelar A, Sharma M, McEvoy LK, Holland D, Brewer JB, Chen CH, Thompson WK, Harold D, Williams J, Owen MJ, O’Donovan MC, Pericak-Vance MA, Mayeux R, Haines JL, Farrer LA, Schellenberg GD, Heutink P, Singleton AB, Brice A, Wood NW, Hardy J, Martinez M, Choi SH, DeStefano A, Ikram MA, Bis JC, Smith A, Fitzpatrick AL, Launer L, van Duijn C, Seshadri S, Ulstein ID, Aarsland D, Fladby T, Djurovic S, Hyman BT, Snaedal J, Stefansson H, Stefansson K, Gasser T, Andreassen OA, Dale AM, for the ADNI *, ADGC, GERAD, CHARGE and IPDGC Investigators **. Genetic overlap between Alzheimer's disease and Parkinson's disease at the MAPT locus. Mol Psychiatry 2015; 20:1588-95. [PMID: 25687773 PMCID: PMC4539304 DOI: 10.1038/mp.2015.6] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/29/2014] [Accepted: 01/08/2015] [Indexed: 12/18/2022]
Abstract
We investigated the genetic overlap between Alzheimer's disease (AD) and Parkinson's disease (PD). Using summary statistics (P-values) from large recent genome-wide association studies (GWAS) (total n=89 904 individuals), we sought to identify single nucleotide polymorphisms (SNPs) associating with both AD and PD. We found and replicated association of both AD and PD with the A allele of rs393152 within the extended MAPT region on chromosome 17 (meta analysis P-value across five independent AD cohorts=1.65 × 10(-7)). In independent datasets, we found a dose-dependent effect of the A allele of rs393152 on intra-cerebral MAPT transcript levels and volume loss within the entorhinal cortex and hippocampus. Our findings identify the tau-associated MAPT locus as a site of genetic overlap between AD and PD, and extending prior work, we show that the MAPT region increases risk of Alzheimer's neurodegeneration.
Collapse
Affiliation(s)
- Rahul S. Desikan
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA,Correspondence should be addressed to: Drs. Rahul S. Desikan and Anders M. Dale, Department of Radiology, University of California, San Diego, 8950 Villa La Jolla Drive, Suite C101, La Jolla, CA, USA 92037-0841, , , Phone: (858)-822-6671, Fax: (858)-534-1078, Dr. Ole A. Andreassen: KG Jebsen Centre for Psychosis Research, Building 49, Oslo University Hospital, Ullevål, Kirkeveien 166, PO Box 4956 Nydalen, 0424 Oslo, Norway, , Ph: +47 23 02 73 50 (22 11 78 43 dir), Fax: +47 23 02 73 33
| | - Andrew J. Schork
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA
| | - Yunpeng Wang
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA,NORMENT; Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Aree Witoelar
- NORMENT; Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Manu Sharma
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research University of Tubingen, Germany,Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Germany
| | - Linda K. McEvoy
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | - Dominic Holland
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - James B. Brewer
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA,Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Chi-Hua Chen
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Wesley K. Thompson
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Denise Harold
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, Wales
| | - Julie Williams
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, Wales
| | - Michael J. Owen
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, Wales
| | - Michael C. O’Donovan
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, Wales
| | | | - Richard Mayeux
- Department of Neurology, Taub Institute on Alzheimer's Disease and the Aging Brain, and Gertrude H. Sergievsky Center, Columbia University, New York, New York, USA
| | - Jonathan L. Haines
- Department of Molecular Physiology and Biophysics, Vanderbilt Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA
| | - Lindsay A. Farrer
- Departments of Medicine (Biomedical Genetics), Neurology, Ophthalmology, Biostatistics, and Epidemiology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | | | - Alexis Brice
- Sorbonne Université, UPMC Univ Paris 06, UM 75, ICM; Inserm, U 1127, ICM; Cnrs, UMR 7225, ICM; ICM, Paris, F-75013 Paris, France
| | - Nicolas W. Wood
- UCL Genetics Institute; and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | | | - Seung Hoi Choi
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Anita DeStefano
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA,The National Heart Lung and Blood Institute’s Framingham Heart Study, Framingham, MA
| | - M. Arfan Ikram
- Deparment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Joshua C. Bis
- Deparment of Internal Medicine, University of Washington, Seattle, WA, USA
| | | | | | - Lenore Launer
- Laboratory of Epidemiology, Demography and Biometry, Intramural Research Program, National Institute on Aging, Washington, DC, USA
| | - Cornelia van Duijn
- Deparment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA,The National Heart Lung and Blood Institute’s Framingham Heart Study, Framingham, MA
| | - Ingun Dina Ulstein
- Norwegian Centre for Dementia Research, Department of Old Age Psychiatry, Oslo University Hospital, Oslo, Norway
| | - Dag Aarsland
- Alzheimer’s Disease Research Centre, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden; Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway; Department of Geriatric Psychiatry, Akershus University Hospital, Oslo, Norway
| | - Tormod Fladby
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Akershus University Hospital, Norway
| | - Srdjan Djurovic
- NORMENT; Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Bradley T. Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Jon Snaedal
- Department of Geriatric Medicine, University Hospital Reykjavik, Iceland
| | | | - Kari Stefansson
- deCODE Genetics/Amgen, Reykjavik, Iceland,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Thomas Gasser
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research University of Tubingen, Germany
| | - Ole A. Andreassen
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,NORMENT; Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway,Correspondence should be addressed to: Drs. Rahul S. Desikan and Anders M. Dale, Department of Radiology, University of California, San Diego, 8950 Villa La Jolla Drive, Suite C101, La Jolla, CA, USA 92037-0841, , , Phone: (858)-822-6671, Fax: (858)-534-1078, Dr. Ole A. Andreassen: KG Jebsen Centre for Psychosis Research, Building 49, Oslo University Hospital, Ullevål, Kirkeveien 166, PO Box 4956 Nydalen, 0424 Oslo, Norway, , Ph: +47 23 02 73 50 (22 11 78 43 dir), Fax: +47 23 02 73 33
| | - Anders M. Dale
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA,Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA,Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,Correspondence should be addressed to: Drs. Rahul S. Desikan and Anders M. Dale, Department of Radiology, University of California, San Diego, 8950 Villa La Jolla Drive, Suite C101, La Jolla, CA, USA 92037-0841, , , Phone: (858)-822-6671, Fax: (858)-534-1078, Dr. Ole A. Andreassen: KG Jebsen Centre for Psychosis Research, Building 49, Oslo University Hospital, Ullevål, Kirkeveien 166, PO Box 4956 Nydalen, 0424 Oslo, Norway, , Ph: +47 23 02 73 50 (22 11 78 43 dir), Fax: +47 23 02 73 33
| | | |
Collapse
|
2927
|
Epigenetic Research of Neurodegenerative Disorders Using Patient iPSC-Based Models. Stem Cells Int 2015; 2016:9464591. [PMID: 26697081 PMCID: PMC4677257 DOI: 10.1155/2016/9464591] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/18/2015] [Indexed: 01/15/2023] Open
Abstract
Epigenetic mechanisms play a role in human disease but their involvement in pathologies from the central nervous system has been hampered by the complexity of the brain together with its unique cellular architecture and diversity. Until recently, disease targeted neural types were only available as postmortem materials after many years of disease evolution. Current in vitro systems of induced pluripotent stem cells (iPSCs) generated by cell reprogramming of somatic cells from patients have provided valuable disease models recapitulating key pathological molecular events. Yet whether cell reprogramming on itself implies a truly epigenetic reprogramming, the epigenetic mechanisms governing this process are only partially understood. Moreover, elucidating epigenetic regulation using patient-specific iPSC-derived neural models is expected to have a great impact to unravel the pathophysiology of neurodegenerative diseases and to hopefully expand future therapeutic possibilities. Here we will critically review current knowledge of epigenetic involvement in neurodegenerative disorders focusing on the potential of iPSCs as a promising tool for epigenetic research of these diseases.
Collapse
|
2928
|
Jaffe AE, Gao Y, Deep-Soboslay A, Tao R, Hyde TM, Weinberger DR, Kleinman JE. Mapping DNA methylation across development, genotype and schizophrenia in the human frontal cortex. Nat Neurosci 2015; 19:40-7. [PMID: 26619358 PMCID: PMC4783176 DOI: 10.1038/nn.4181] [Citation(s) in RCA: 351] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023]
Abstract
DNA methylation (DNAm) is important in brain development and is potentially important in schizophrenia. We characterized DNAm in prefrontal cortex from 335 non-psychiatric controls across the lifespan and 191 patients with schizophrenia and identified widespread changes in the transition from prenatal to postnatal life. These DNAm changes manifest in the transcriptome, correlate strongly with a shifting cellular landscape and overlap regions of genetic risk for schizophrenia. A quarter of published genome-wide association studies (GWAS)-suggestive loci (4,208 of 15,930, P < 10(-100)) manifest as significant methylation quantitative trait loci (meQTLs), including 59.6% of GWAS-positive schizophrenia loci. We identified 2,104 CpGs that differ between schizophrenia patients and controls that were enriched for genes related to development and neurodifferentiation. The schizophrenia-associated CpGs strongly correlate with changes related to the prenatal-postnatal transition and show slight enrichment for GWAS risk loci while not corresponding to CpGs differentiating adolescence from later adult life. These data implicate an epigenetic component to the developmental origins of this disorder.
Collapse
Affiliation(s)
- Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland, USA.,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Yuan Gao
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland, USA
| | - Amy Deep-Soboslay
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland, USA
| | - Ran Tao
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Department of Neuroscience and the Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland, USA
| |
Collapse
|
2929
|
Holm IE, Alstrup AKO, Luo Y. Genetically modified pig models for neurodegenerative disorders. J Pathol 2015; 238:267-87. [DOI: 10.1002/path.4654] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 09/22/2015] [Accepted: 10/05/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Ida E Holm
- Department of Pathology; Randers Hospital; 8930 Randers Denmark
- Department of Clinical Medicine; Aarhus University; 8000 Aarhus C Denmark
| | | | - Yonglun Luo
- Department of Biomedicine; Aarhus University; 8000 Aarhus C Denmark
| |
Collapse
|
2930
|
Li MJ, Liu Z, Wang P, Wong MP, Nelson MR, Kocher JPA, Yeager M, Sham PC, Chanock SJ, Xia Z, Wang J. GWASdb v2: an update database for human genetic variants identified by genome-wide association studies. Nucleic Acids Res 2015; 44:D869-76. [PMID: 26615194 PMCID: PMC4702921 DOI: 10.1093/nar/gkv1317] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/10/2015] [Indexed: 12/19/2022] Open
Abstract
Genome-wide association studies (GWASs), now as a routine approach to study single-nucleotide polymorphism (SNP)-trait association, have uncovered over ten thousand significant trait/disease associated SNPs (TASs). Here, we updated GWASdb (GWASdb v2, http://jjwanglab.org/gwasdb) which provides comprehensive data curation and knowledge integration for GWAS TASs. These updates include: (i) Up to August 2015, we collected 2479 unique publications from PubMed and other resources; (ii) We further curated moderate SNP-trait associations (P-value < 1.0×10−3) from each original publication, and generated a total of 252 530 unique TASs in all GWASdb v2 collected studies; (iii) We manually mapped 1610 GWAS traits to 501 Human Phenotype Ontology (HPO) terms, 435 Disease Ontology (DO) terms and 228 Disease Ontology Lite (DOLite) terms. For each ontology term, we also predicted the putative causal genes; (iv) We curated the detailed sub-populations and related sample size for each study; (v) Importantly, we performed extensive function annotation for each TAS by incorporating gene-based information, ENCODE ChIP-seq assays, eQTL, population haplotype, functional prediction across multiple biological domains, evolutionary signals and disease-related annotation; (vi) Additionally, we compiled a SNP-drug response association dataset for 650 pharmacogenetic studies involving 257 drugs in this update; (vii) Last, we improved the user interface of website.
Collapse
Affiliation(s)
- Mulin Jun Li
- Centre for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zipeng Liu
- Centre for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Panwen Wang
- Centre for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Maria P Wong
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Matthew R Nelson
- Quantitative Sciences, GlaxoSmithKline, Research Triangle Park, NC, USA
| | - Jean-Pierre A Kocher
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Pak Chung Sham
- Centre for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China State Key Laboratory of Brain and Cognitive Sciences and Department of Psychiatry, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Zhengyuan Xia
- Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Junwen Wang
- Centre for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
2931
|
Galvan V, Hart MJ. Vascular mTOR-dependent mechanisms linking the control of aging to Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2015; 1862:992-1007. [PMID: 26639036 DOI: 10.1016/j.bbadis.2015.11.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 02/07/2023]
Abstract
Aging is the strongest known risk factor for Alzheimer's disease (AD). With the discovery of the mechanistic target of rapamycin (mTOR) as a critical pathway controlling the rate of aging in mice, molecules at the interface between the regulation of aging and the mechanisms of specific age-associated diseases can be identified. We will review emerging evidence that mTOR-dependent brain vascular dysfunction, a universal feature of aging, may be one of the mechanisms linking the regulation of the rate of aging to the pathogenesis of Alzheimer's disease. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Veronica Galvan
- Department of Physiology and the Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio.
| | - Matthew J Hart
- Department of Biochemistry, University of Texas Health Science Center at San Antonio
| |
Collapse
|
2932
|
Kozlenkov A, Wang M, Roussos P, Rudchenko S, Barbu M, Bibikova M, Klotzle B, Dwork AJ, Zhang B, Hurd YL, Koonin EV, Wegner M, Dracheva S. Substantial DNA methylation differences between two major neuronal subtypes in human brain. Nucleic Acids Res 2015; 44:2593-612. [PMID: 26612861 PMCID: PMC4824074 DOI: 10.1093/nar/gkv1304] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/09/2015] [Indexed: 12/29/2022] Open
Abstract
The brain is built from a large number of cell types which have been historically classified using location, morphology and molecular markers. Recent research suggests an important role of epigenetics in shaping and maintaining cell identity in the brain. To elucidate the role of DNA methylation in neuronal differentiation, we developed a new protocol for separation of nuclei from the two major populations of human prefrontal cortex neurons—GABAergic interneurons and glutamatergic (GLU) projection neurons. Major differences between the neuronal subtypes were revealed in CpG, non-CpG and hydroxymethylation (hCpG). A dramatically greater number of undermethylated CpG sites in GLU versus GABA neurons were identified. These differences did not directly translate into differences in gene expression and did not stem from the differences in hCpG methylation, as more hCpG methylation was detected in GLU versus GABA neurons. Notably, a comparable number of undermethylated non-CpG sites were identified in GLU and GABA neurons, and non-CpG methylation was a better predictor of subtype-specific gene expression compared to CpG methylation. Regions that are differentially methylated in GABA and GLU neurons were significantly enriched for schizophrenia risk loci. Collectively, our findings suggest that functional differences between neuronal subtypes are linked to their epigenetic specification.
Collapse
Affiliation(s)
- Alexey Kozlenkov
- James J. Peters VA Medical Center, Bronx, NY 10468, USA The Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Panos Roussos
- James J. Peters VA Medical Center, Bronx, NY 10468, USA The Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Mihaela Barbu
- Hospital for Special Surgery, New York, NY 10021, USA
| | | | | | - Andrew J Dwork
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yasmin L Hurd
- The Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eugene V Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Stella Dracheva
- James J. Peters VA Medical Center, Bronx, NY 10468, USA The Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
2933
|
Chi S, Song JH, Tan MS, Zhang W, Wang ZX, Jiang T, Tan L, Yu JT. Association of Single-Nucleotide Polymorphism in ANK1 with Late-Onset Alzheimer's Disease in Han Chinese. Mol Neurobiol 2015; 53:6476-6481. [PMID: 26611832 DOI: 10.1007/s12035-015-9547-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 11/17/2015] [Indexed: 02/01/2023]
Abstract
Recently, two CpG sites in ankyrin 1 (ANK1) gene were identified to be hypermethylated and associated with Alzheimer's disease (AD)-related neuropathology in two large independent studies. Genetic variations are indicated to be involved in DNA methylation, especially when the associated single-nucleotide polymorphisms (SNPs) are located adjacent to the CpG site. Accordingly, ANK1 polymorphisms might contribute to late-onset AD (LOAD) risk. One polymorphism rs515071 was identified to be a potential risk factor for type 2 diabetes (T2D). As shared genetic background was found underlying T2D and AD, we postulate that rs515071 polymorphism may be associated with late-onset AD (LOAD) risk and assessed the association in 982 LOAD patients and 1346 sex- and age-matched healthy controls. Our results showed that minor allele A of rs515071 significantly increased LOAD risk in the APOE ε4 (+) subgroup (genotype P = 0.015, allele P = 0.020). After adjusting for age and gender, the association remained significant under the dominant model (OR = 1.809, 95 % confidence interval (CI) = 1.186-2.757, P = 0.006). In conclusion, our findings demonstrate that rs515071 in ANK1 is a novel genetic risk for LOAD susceptibility in Han Chinese.
Collapse
Affiliation(s)
- Song Chi
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 5 Donghai Middle Road, Qingdao, Shandong Province, 266071, China
| | - Jing-Hui Song
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Meng-Shan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 5 Donghai Middle Road, Qingdao, Shandong Province, 266071, China
| | - Wei Zhang
- Department of Emergency, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, People's Republic of China
| | - Zi-Xuan Wang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 5 Donghai Middle Road, Qingdao, Shandong Province, 266071, China
| | - Teng Jiang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 5 Donghai Middle Road, Qingdao, Shandong Province, 266071, China.
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 5 Donghai Middle Road, Qingdao, Shandong Province, 266071, China. .,Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, Box 1207, San Francisco, CA, 94158, USA.
| |
Collapse
|
2934
|
Grassmann F, Ach T, Brandl C, Heid IM, Weber BH. What Does Genetics Tell Us About Age-Related Macular Degeneration? Annu Rev Vis Sci 2015; 1:73-96. [DOI: 10.1146/annurev-vision-082114-035609] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | - Thomas Ach
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, Alabama 35294
- Department of Ophthalmology, University Hospital Würzburg, Würzburg, D-97080, Germany
| | - Caroline Brandl
- Institute of Human Genetics and
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, D-93053, Germany;
- Department of Ophthalmology, University Hospital Regensburg, Regensburg, D-93042, Germany
| | - Iris M. Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, D-93053, Germany;
| | | |
Collapse
|
2935
|
Minter MR, Taylor JM, Crack PJ. The contribution of neuroinflammation to amyloid toxicity in Alzheimer's disease. J Neurochem 2015; 136:457-74. [PMID: 26509334 DOI: 10.1111/jnc.13411] [Citation(s) in RCA: 323] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/11/2015] [Accepted: 10/22/2015] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and the most common cause of dementia. Deposition of amyloid-β (Aβ) remains a hallmark feature of the disease, yet the precise mechanism(s) by which this peptide induces neurotoxicity remain unknown. Neuroinflammation has long been implicated in AD pathology, yet its contribution to disease progression is still not understood. Recent evidence suggests that various Aβ complexes interact with microglial and astrocytic expressed pattern recognition receptors that initiate innate immunity. This process involves secretion of pro-inflammatory cytokines, chemokines and generation of reactive oxygen species that, in excess, drive a dysregulated immune response that contributes to neurodegeneration. The mechanisms by which a neuroinflammatory response can influence Aβ production, aggregation and eventual clearance are now becoming key areas where future therapeutic intervention may slow progression of AD. This review will focus on evidence supporting the combined neuroinflammatory-amyloid hypothesis for pathogenesis of AD, describing the key cell types, pathways and mediators involved. Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the leading cause of dementia worldwide. Deposition of intracellular plaques containing amyloid-beta (Aβ) is a hallmark proteinopathy of the disease yet the precise mechanisms by which this peptide induces neurotoxicity remains unknown. A neuroinflammatory response involving polarized microglial activity, enhanced astrocyte reactivity and elevated pro-inflammatory cytokine and chemokine load has long been implicated in AD and proposed to facilitate neurodegeneration. In this issue we discuss key receptor systems of innate immunity that detect Aβ, drive pro-inflammatory cytokine and chemokine production and influence Aβ aggregation and clearance. Evidence summarized in this review supports the combined neuroinflammatory-amyloid hypothesis for pathogenesis of AD and highlights the potential of immunomodulatory agents as potential future therapies for AD patients.
Collapse
Affiliation(s)
- Myles R Minter
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Juliet M Taylor
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
2936
|
Inactivation of C4orf26 in toothless placental mammals. Mol Phylogenet Evol 2015; 95:34-45. [PMID: 26596502 DOI: 10.1016/j.ympev.2015.11.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/22/2015] [Accepted: 11/03/2015] [Indexed: 01/11/2023]
Abstract
Previous studies have reported inactivated copies of six enamel-related genes (AMBN, AMEL, AMTN, ENAM, KLK4, MMP20) and one dentin-related gene (DSPP) in one or more toothless vertebrates and/or vertebrates with enamelless teeth, thereby providing evidence that these genes are enamel or tooth-specific with respect to their critical functions that are maintained by natural selection. Here, we employ available genome sequences for edentulous and enamelless mammals to evaluate the enamel specificity of four genes (WDR72, SLC24A4, FAM83H, C4orf26) that have been implicated in amelogenesis imperfecta, a condition in which proper enamel formation is abrogated during tooth development. Coding sequences for WDR72, SCL24A4, and FAM83H are intact in four edentulous taxa (Chinese pangolin, three baleen whales) and three taxa (aardvark, nine-banded armadillo, Hoffmann's two-toed sloth) with enamelless teeth, suggesting that these genes have critical functions beyond their involvement in tooth development. By contrast, genomic data for C4orf26 reveal inactivating mutations in pangolin and bowhead whale as well as evidence for deletion of this gene in two minke whale species. Hybridization capture of exonic regions and PCR screens provide evidence for inactivation of C4orf26 in eight additional baleen whale species. However, C4orf26 is intact in all three species with enamelless teeth that were surveyed, as well as in 95 additional mammalian species with enamel-capped teeth. Estimates of selection intensity suggest that dN/dS ratios on branches leading to taxa with enamelless teeth are similar to the dN/dS ratio on branches leading to taxa with enamel-capped teeth. Based on these results, we conclude that C4orf26 is tooth-specific, but not enamel-specific, with respect to its essential functions that are maintained by natural selection. A caveat is that an alternative splice site variant, which translates exon 3 in a different reading frame, is putatively functional in Catarrhini and may have evolved an additional role in this primate clade.
Collapse
|
2937
|
Bergen AA, Kaing S, ten Brink JB, Gorgels TG, Janssen SF. Gene expression and functional annotation of human choroid plexus epithelium failure in Alzheimer's disease. BMC Genomics 2015; 16:956. [PMID: 26573292 PMCID: PMC4647590 DOI: 10.1186/s12864-015-2159-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/27/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common form of dementia. AD has a multifactorial disease etiology and is currently untreatable. Multiple genes and molecular mechanisms have been implicated in AD, including ß-amyloid deposition in the brain, neurofibrillary tangle accumulation of hyper-phosphorylated Tau, synaptic failure, oxidative stress and inflammation. Relatively little is known about the role of the blood-brain barriers, especially the blood-cerebrospinal fluid barrier (BCSFB), in AD. The BCSFB is involved in cerebrospinal fluid (CSF) production, maintenance of brain homeostasis and neurodegenerative disorders. RESULTS Using an Agilent platform with common reference design, we performed a large scale gene expression analysis and functional annotation of the Choroid Plexus Epithelium (CPE), which forms the BCSFB. We obtained 2 groups of freshly frozen Choroid Plexus (CP) of 7 human donor brains each, with and without AD: Braak stages (0-1) and (5-6). We cut CP cryo-sections and isolated RNA from cresyl-violet stained, laser dissected CPE cells. Gene expression results were analysed with T-tests (R) and the knowledge-database Ingenuity. We found statistically significantly altered gene expression data sets, biological functions, canonical pathways, molecular networks and functionalities in AD-affected CPE. We observed specific cellular changes due to increased oxidative stress, such as the unfolded protein response, E1F2 and NRF2 signalling and the protein ubiquitin pathway. Most likely, the AD-affected BCSFB barrier becomes more permeable due to downregulation of CLDN5. Finally, our data also predicted down regulation of the glutathione mediated detoxification pathway and the urea cycle in the AD CPE, which suggest that the CPE sink action may be impaired. Remarkably, the expression of a number of genes known to be involved in AD, such as APP, PSEN1, PSEN2, TTR and CLU is moderate to high and remains stable in both healthy and affected CPE. Literature labelling of our new functional molecular networks confirmed multiple previous (molecular) observations in the AD literature and revealed many new ones. CONCLUSIONS We conclude that CPE failure in AD exists. Combining our data with those of the literature, we propose the following chronological and overlapping chain of events: increased Aß burden on CPE; increased oxidative stress in CPE; despite continuous high expression of TTR: decreased capability of CPE to process amyloid; (pro-) inflammatory and growth factor signalling by CPE; intracellular ubiquitin involvement, remodelling of CPE tight junctions and, finally, cellular atrophy. Our data corroborates the hypothesis that increased BCSFB permeability, especially loss of selective CLDN5-mediated paracellular transport, altered CSF production and CPE sink action, as well as loss of CPE mediated macrophage recruitment contribute to the pathogenesis of AD.
Collapse
Affiliation(s)
- Arthur A Bergen
- Department of Clinical Genetics, Academic Medical Centre, Amsterdam, AMC, Meibergdreef 9, 1105 AZ AMC, Amsterdam, The Netherlands. .,The Netherlands Institute for Neurosciences (NIN-KNAW), Amsterdam, The Netherlands.
| | - Sovann Kaing
- The Netherlands Institute for Neurosciences (NIN-KNAW), Amsterdam, The Netherlands
| | - Jacoline B ten Brink
- Department of Clinical Genetics, Academic Medical Centre, Amsterdam, AMC, Meibergdreef 9, 1105 AZ AMC, Amsterdam, The Netherlands
| | | | - Theo G Gorgels
- The Netherlands Institute for Neurosciences (NIN-KNAW), Amsterdam, The Netherlands.,University Eye Clinic Maastricht, MUMC, Maastricht, The Netherlands
| | - Sarah F Janssen
- The Netherlands Institute for Neurosciences (NIN-KNAW), Amsterdam, The Netherlands.,Department of Ophthalmology, VUMC, Amsterdam, The Netherlands
| |
Collapse
|
2938
|
Sims R, Williams J. Defining the Genetic Architecture of Alzheimer's Disease: Where Next? NEURODEGENER DIS 2015; 16:6-11. [DOI: 10.1159/000440841] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/03/2015] [Indexed: 11/19/2022] Open
|
2939
|
Xiao Q, Liu ZJ, Tao S, Sun YM, Jiang D, Li HL, Chen H, Liu X, Lapin B, Wang CH, Zheng SL, Xu J, Wu ZY. Risk prediction for sporadic Alzheimer's disease using genetic risk score in the Han Chinese population. Oncotarget 2015; 6:36955-36964. [PMID: 26543236 PMCID: PMC4741908 DOI: 10.18632/oncotarget.6271] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 09/22/2015] [Indexed: 01/10/2023] Open
Abstract
More than 30 independent single-nucleotide polymorphisms (SNPs) have been associated with Alzheimer's disease (AD) risk by genome-wide association studies (GWAS) in European. We aimed to confirm these SNPs in Chinese Han and investigate the utility of these genetic markers. We randomly divided 459 sporadic AD (SAD) patients and 751 cognitively normal controls into two sets (discovery and testing). Thirty-three SAD risk-associated SNPs were firstly tested in the discovery set. Significant SNPs were used to calculate genetic risk score (GRS) in the testing set. Predictive performance of GRS was evaluated using the area under the receiver operating characteristic curve (AUC). In the discovery set, 6 SNPs were confirmed (P = 7.87 x 10(-11)~0.048), including rs9349407 in CD2AP, rs11218343 in SORL1, rs17125944 in FERMT2, rs6859 in PVRL2, rs157580 and rs2075650 in TOMM40. The first three SNPs were associated with SAD risk independent of APOE genotypes. GRS based on these three SNPs were significantly associated with SAD risk in the independent testing set (P = 0.002). The AUC for discriminating cases from controls was 0.58 for GRS, 0.60 for APOE, and 0.64 for GRS and APOE. Our data demonstrated that GRS based on AD risk-associated SNPs may supplement APOE for better assessing individual risk for AD in Chinese.
Collapse
Affiliation(s)
- Qianyi Xiao
- Center for Genomic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
| | - Zhi-Jun Liu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, China
- Department of Neurology and Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Sha Tao
- Center for Genomic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
| | - Yi-Min Sun
- Department of Neurology and Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Deke Jiang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Hong-Lei Li
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, China
| | - Haitao Chen
- Center for Genomic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
| | - Xu Liu
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Brittany Lapin
- Program for Personalized Cancer Care, NorthShore University Health System, Evanston, IL, USA
| | - Chi-Hsiung Wang
- Program for Personalized Cancer Care, NorthShore University Health System, Evanston, IL, USA
| | - S. Lilly Zheng
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Program for Personalized Cancer Care, NorthShore University Health System, Evanston, IL, USA
| | - Jianfeng Xu
- Center for Genomic Translational Medicine and Prevention, School of Public Health, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Program for Personalized Cancer Care, NorthShore University Health System, Evanston, IL, USA
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
2940
|
Amemori T, Jendelova P, Ruzicka J, Urdzikova LM, Sykova E. Alzheimer's Disease: Mechanism and Approach to Cell Therapy. Int J Mol Sci 2015; 16:26417-51. [PMID: 26556341 PMCID: PMC4661820 DOI: 10.3390/ijms161125961] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/26/2015] [Accepted: 10/26/2015] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. The risk of AD increases with age. Although two of the main pathological features of AD, amyloid plaques and neurofibrillary tangles, were already recognized by Alois Alzheimer at the beginning of the 20th century, the pathogenesis of the disease remains unsettled. Therapeutic approaches targeting plaques or tangles have not yet resulted in satisfactory improvements in AD treatment. This may, in part, be due to early-onset and late-onset AD pathogenesis being underpinned by different mechanisms. Most animal models of AD are generated from gene mutations involved in early onset familial AD, accounting for only 1% of all cases, which may consequently complicate our understanding of AD mechanisms. In this article, the authors discuss the pathogenesis of AD according to the two main neuropathologies, including senescence-related mechanisms and possible treatments using stem cells, namely mesenchymal and neural stem cells.
Collapse
Affiliation(s)
- Takashi Amemori
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Pavla Jendelova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague 5, Czech Republic.
| | - Jiri Ruzicka
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Lucia Machova Urdzikova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Eva Sykova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague 5, Czech Republic.
| |
Collapse
|
2941
|
Vincent B, Buntzman A, Hopson B, McEwen C, Cowell L, Akoglu A, Zhang H, Frelinger J. iWAS--A novel approach to analyzing Next Generation Sequence data for immunology. Cell Immunol 2015; 299:6-13. [PMID: 26547365 DOI: 10.1016/j.cellimm.2015.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 10/28/2015] [Accepted: 10/28/2015] [Indexed: 01/12/2023]
Abstract
In this communication we describe a novel way to use Next Generation Sequence from the receptors expressed on T and B cells. This informatics methodology is named iWAS, for immunonome Wide Association Study, where we use the immune receptor sequences derived from T and B cells and the features of those receptors (sequences themselves, V/J gene usage, length and character each of the CDR3 sub-regions) to define biomarkers of health and disease, as well as responses to therapies. Unlike GWAS, which do not provide immediate access to mechanism, the associations with immune receptors immediately suggest possible and plausible entrée's into disease pathogenesis and treatment.
Collapse
Affiliation(s)
- Benjamin Vincent
- Department of Medicine, University of North Carolina, Chapel Hill, NC 2714, United States.
| | - Adam Buntzman
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724, United States.
| | - Benjamin Hopson
- School of Engineering, The University of Edinburgh, Edinburgh EH9 3JL, UK; Cambridge Consultants, Science Park, Milton Rd, Cambridge CB4 0DW, UK.
| | - Chris McEwen
- Cambridge Consultants, Science Park, Milton Rd, Cambridge CB4 0DW, UK.
| | - Lindsay Cowell
- Division of Biomedical Informatics, Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| | - Ali Akoglu
- Department of Electrical and Computer Engineering, University of Arizona, Tucson, AZ 85721, United States.
| | - Helen Zhang
- Department of Mathematics, University of Arizona, Tucson, AZ 85721, United States.
| | - Jeffrey Frelinger
- Department of Immunobiology, University of Arizona, Tucson, AZ 85724, United States.
| |
Collapse
|
2942
|
Increased serum levels of sortilin are associated with depression and correlated with BDNF and VEGF. Transl Psychiatry 2015; 5:e677. [PMID: 26556286 PMCID: PMC5068760 DOI: 10.1038/tp.2015.167] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/17/2015] [Accepted: 09/19/2015] [Indexed: 12/25/2022] Open
Abstract
Neurotrophic factors have been investigated in relation to depression. The aim of the present study was to widen this focus to sortilin, a receptor involved in neurotrophic signalling. The serum sortilin level was investigated in 152 individuals with depression and 216 control individuals, and eight genetic markers located within the SORT1 gene were successfully analysed for association with depression. Genotyping was performed using the Sequenom MassARRAY platform. All the individuals returned a questionnaire and participated in a semi-structured diagnostic interview. Sortilin levels were measured by immunoassay, and potential determinants of the serum sortilin level were assessed by generalized linear models. Serum levels of brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) were measured in previous studies. We identified a significant increase of serum sortilin levels in depressed individuals compared with controls (P=0.0002) and significant positive correlation between serum sortilin levels and the corresponding levels of BDNF and VEGF. None of the genotyped SNPs were associated with depression. Additional analyses showed that the serum sortilin level was influenced by several other factors. Alcohol intake and body mass index, as well as depression, serum BDNF and serum VEGF were identified as predictors of serum sortilin levels in our final multivariate model. In conclusion, the results suggest a role of circulating sortilin in depression which may relate to altered activity of neurotrophic factors.
Collapse
|
2943
|
Chung WS, Welsh CA, Barres BA, Stevens B. Do glia drive synaptic and cognitive impairment in disease? Nat Neurosci 2015; 18:1539-1545. [PMID: 26505565 PMCID: PMC4739631 DOI: 10.1038/nn.4142] [Citation(s) in RCA: 287] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/05/2015] [Indexed: 02/06/2023]
Abstract
Synaptic dysfunction is a hallmark of many neurodegenerative and psychiatric brain disorders, yet we know little about the mechanisms that underlie synaptic vulnerability. Although neuroinflammation and reactive gliosis are prominent in virtually every CNS disease, glia are largely viewed as passive responders to neuronal damage rather than drivers of synaptic dysfunction. This perspective is changing with the growing realization that glia actively signal with neurons and influence synaptic development, transmission and plasticity through an array of secreted and contact-dependent signals. We propose that disruptions in neuron-glia signaling contribute to synaptic and cognitive impairment in disease. Illuminating the mechanisms by which glia influence synapse function may lead to the development of new therapies and biomarkers for synaptic dysfunction.
Collapse
Affiliation(s)
- Won-Suk Chung
- Department of Neurobiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Christina A Welsh
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ben A Barres
- Department of Neurobiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Beth Stevens
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
2944
|
Saad M, Brkanac Z, Wijsman EM. Family-based genome scan for age at onset of late-onset Alzheimer's disease in whole exome sequencing data. GENES, BRAIN, AND BEHAVIOR 2015; 14:607-17. [PMID: 26394601 PMCID: PMC4715764 DOI: 10.1111/gbb.12250] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/08/2015] [Accepted: 08/24/2015] [Indexed: 01/31/2023]
Abstract
Alzheimer's disease (AD) is a common and complex neurodegenerative disease. Age at onset (AAO) of AD is an important component phenotype with a genetic basis, and identification of genes in which variation affects AAO would contribute to identification of factors that affect timing of onset. Increase in AAO through prevention or therapeutic measures would have enormous benefits by delaying AD and its associated morbidities. In this paper, we performed a family-based genome-wide association study for AAO of late-onset AD in whole exome sequence data generated in multigenerational families with multiple AD cases. We conducted single marker and gene-based burden tests for common and rare variants, respectively. We combined association analyses with variance component linkage analysis, and with reference to prior studies, in order to enhance evidence of the identified genes. For variants and genes implicated by the association study, we performed a gene-set enrichment analysis to identify potential novel pathways associated with AAO of AD. We found statistically significant association with AAO for three genes (WRN, NTN4 and LAMC3) with common associated variants, and for four genes (SLC8A3, SLC19A3, MADD and LRRK2) with multiple rare-associated variants that have a plausible biological function related to AD. The genes we have identified are in pathways that are strong candidates for involvement in the development of AD pathology and may lead to a better understanding of AD pathogenesis.
Collapse
Affiliation(s)
- Mohamad Saad
- Department of Biostatistics, University of Washington, Seattle, USA
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, USA
| | - Zoran Brkanac
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, USA
| | - Ellen M. Wijsman
- Department of Biostatistics, University of Washington, Seattle, USA
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, USA
| |
Collapse
|
2945
|
Goldman JG, Aggarwal NT, Schroeder CD. Mild cognitive impairment: an update in Parkinson's disease and lessons learned from Alzheimer's disease. Neurodegener Dis Manag 2015; 5:425-43. [PMID: 26517759 DOI: 10.2217/nmt.15.34] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Cognitive dysfunction is an important focus of research in Parkinson's disease (PD) and Alzheimer's disease (AD). While the concept of amnestic mild cognitive impairment (MCI) as a prodrome to AD has been recognized for many years, the construct of MCI in PD is a relative newcomer with recent development of diagnostic criteria, biomarker research programs and treatment trials. Controversies and challenges, however, regarding PD-MCI's definition, application, heterogeneity and different trajectories have arisen. This review will highlight current research advances and challenges in PD-MCI. Furthermore, lessons from the AD field, which has witnessed an evolution in MCI/AD definitions, relevant advances in biomarker research and development of disease-modifying and targeted therapeutic trials will be discussed.
Collapse
Affiliation(s)
- Jennifer G Goldman
- Rush University Medical Center, Department of Neurological Sciences, Section of Parkinson Disease & Movement Disorders, 1725 W. Harrison Street, Suite 755, Chicago, IL 60612, USA
| | - Neelum T Aggarwal
- Rush University Medical Center, Department of Neurological Sciences & Rush Alzheimer's Disease Center, 600 South Paulina, Suite 1038, Chicago, IL 60612, USA
| | - Cynthia D Schroeder
- Rush University Medical Center, Department of Neurological Sciences, 1735 W. Harrison Street, Suite 306, Chicago, IL 60612, USA
| |
Collapse
|
2946
|
Villegas-Llerena C, Phillips A, Garcia-Reitboeck P, Hardy J, Pocock JM. Microglial genes regulating neuroinflammation in the progression of Alzheimer's disease. Curr Opin Neurobiol 2015; 36:74-81. [PMID: 26517285 DOI: 10.1016/j.conb.2015.10.004] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/24/2015] [Accepted: 10/07/2015] [Indexed: 01/09/2023]
Abstract
Neuroinflammation is a pathological hallmark of Alzheimer's disease (AD), and microglia, the brain's resident phagocyte, are pivotal for the immune response observed in AD. Microglia act as sentinel and protective cells, but may become inappropriately reactive in AD to drive neuropathology. Recent Genome Wide Association Studies (GWAS) have identified more than 20 gene variants associated with an increased risk of late-onset AD (LOAD), the most prevalent form of AD [1]. The findings strongly implicate genes related to the immune response (CR1, CD33, MS4A, CLU, ABCA7, EPHA1 and HLA-DRB5-HLA-DRB1), endocytosis (BIN1, PICALM, CD2AP, EPHA1 and SORL1) and lipid biology (CLU, ABCA7 and SORL1) [2-8], and many encode proteins which are highly expressed in microglia [1]. Furthermore, recent identification of a low frequency mutation in the gene encoding the triggering receptor expressed in myeloid cells 2 protein (TREM2) confers increased risk of AD in LOAD cohorts with an effect size similar to that for APOE, until recently the only identified genetic risk factor associated with LOAD [9,10(••)] (Figure 1). The present review summarises our current understanding of the probable roles of microglial genes in the regulation of neuroinflammatory processes in AD and their relation to other processes affecting the disease's progression.
Collapse
Affiliation(s)
- Claudio Villegas-Llerena
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK; Department of Molecular Neuroscience, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - Alexandra Phillips
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - Pablo Garcia-Reitboeck
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK; Department of Molecular Neuroscience, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - John Hardy
- Department of Molecular Neuroscience, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - Jennifer M Pocock
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK.
| |
Collapse
|
2947
|
Escott-Price V, Sims R, Bannister C, Harold D, Vronskaya M, Majounie E, Badarinarayan N, Morgan K, Passmore P, Holmes C, Powell J, Brayne C, Gill M, Mead S, Goate A, Cruchaga C, Lambert JC, van Duijn C, Maier W, Ramirez A, Holmans P, Jones L, Hardy J, Seshadri S, Schellenberg GD, Amouyel P, Williams J. Common polygenic variation enhances risk prediction for Alzheimer's disease. Brain 2015; 138:3673-84. [PMID: 26490334 DOI: 10.1093/brain/awv268] [Citation(s) in RCA: 303] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/07/2015] [Indexed: 01/08/2023] Open
Abstract
The identification of subjects at high risk for Alzheimer's disease is important for prognosis and early intervention. We investigated the polygenic architecture of Alzheimer's disease and the accuracy of Alzheimer's disease prediction models, including and excluding the polygenic component in the model. This study used genotype data from the powerful dataset comprising 17 008 cases and 37 154 controls obtained from the International Genomics of Alzheimer's Project (IGAP). Polygenic score analysis tested whether the alleles identified to associate with disease in one sample set were significantly enriched in the cases relative to the controls in an independent sample. The disease prediction accuracy was investigated in a subset of the IGAP data, a sample of 3049 cases and 1554 controls (for whom APOE genotype data were available) by means of sensitivity, specificity, area under the receiver operating characteristic curve (AUC) and positive and negative predictive values. We observed significant evidence for a polygenic component enriched in Alzheimer's disease (P = 4.9 × 10(-26)). This enrichment remained significant after APOE and other genome-wide associated regions were excluded (P = 3.4 × 10(-19)). The best prediction accuracy AUC = 78.2% (95% confidence interval 77-80%) was achieved by a logistic regression model with APOE, the polygenic score, sex and age as predictors. In conclusion, Alzheimer's disease has a significant polygenic component, which has predictive utility for Alzheimer's disease risk and could be a valuable research tool complementing experimental designs, including preventative clinical trials, stem cell selection and high/low risk clinical studies. In modelling a range of sample disease prevalences, we found that polygenic scores almost doubles case prediction from chance with increased prediction at polygenic extremes.
Collapse
Affiliation(s)
- Valentina Escott-Price
- 1 Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Rebecca Sims
- 1 Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Christian Bannister
- 1 Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Denise Harold
- 2 School of Medicine, Trinity College Dublin, College Green, Dublin 2, Ireland
| | - Maria Vronskaya
- 1 Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Elisa Majounie
- 1 Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Nandini Badarinarayan
- 1 Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | | | | | - Kevin Morgan
- 3 Institute of Genetics, Queens Medical Centre, University of Nottingham, UK
| | - Peter Passmore
- 4 Ageing Group, Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, UK
| | - Clive Holmes
- 5 Division of Clinical Neurosciences, School of Medicine, University of Southampton, Southampton, UK
| | - John Powell
- 6 Kings College London, Institute of Psychiatry, Department of Neuroscience, De Crespigny Park, Denmark Hill, London
| | - Carol Brayne
- 7 Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Michael Gill
- 8 Mercers Institute for Research on Aging, St. James Hospital and Trinity College, Dublin, Ireland
| | - Simon Mead
- 9 MRC Prion Unit, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Alison Goate
- 10 Neuroscience Department, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Carlos Cruchaga
- 11 Departments of Psychiatry, Neurology and Genetics, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jean-Charles Lambert
- 12 Inserm U744, Lille, 59000, France 13 Université Lille 2, Lille, 59000, France 14 Institut Pasteur de Lille, Lille, 59000, France
| | - Cornelia van Duijn
- 15 Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Wolfgang Maier
- 16 Department of Psychiatry and Psychotherapy, University of Bonn, 53127 Bonn, Germany 17 German Centre for Neurodegenerative Diseases (DZNE), Bonn, 53175, Germany
| | - Alfredo Ramirez
- 16 Department of Psychiatry and Psychotherapy, University of Bonn, 53127 Bonn, Germany 18 Institute of Human Genetics, University of Bonn, 53127, Bonn, Germany
| | - Peter Holmans
- 1 Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Lesley Jones
- 1 Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - John Hardy
- 19 Department of Molecular Neuroscience and Reta Lilla Weston Laboratories, Institute of Neurology, London, UK
| | - Sudha Seshadri
- 20 Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Gerard D Schellenberg
- 21 Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Philippe Amouyel
- 12 Inserm U744, Lille, 59000, France 13 Université Lille 2, Lille, 59000, France 14 Institut Pasteur de Lille, Lille, 59000, France 22 Centre Hospitalier Régional Universitaire de Lille, Lille, 59000, France
| | - Julie Williams
- 1 Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| |
Collapse
|
2948
|
Liao XJ, Li L, Zhang ZY, Long Y, Yang B, Ruan GR, Su Y, Ai HS, Zhang WC, Deng WY, Xiao SJ, Ren J, Ding NS, Huang LS. Susceptibility loci for umbilical hernia in swine detected by genome-wide association. RUSS J GENET+ 2015. [DOI: 10.1134/s1022795415100105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
2949
|
Varvel NH, Grathwohl SA, Degenhardt K, Resch C, Bosch A, Jucker M, Neher JJ. Replacement of brain-resident myeloid cells does not alter cerebral amyloid-β deposition in mouse models of Alzheimer's disease. ACTA ACUST UNITED AC 2015; 212:1803-9. [PMID: 26458770 PMCID: PMC4612086 DOI: 10.1084/jem.20150478] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 09/11/2015] [Indexed: 01/08/2023]
Abstract
Immune cells of myeloid lineage cluster around amyloid-β plaques in the Alzheimer’s disease brain. However, assigning functional roles to myeloid subtypes, namely brain-resident microglia versus peripherally derived monocytes, has been problematic. Now, Varvel et al. use a model of central nervous system myeloid cell depletion to demonstrate that repopulation by peripheral monocytes is insufficient to eliminate plaques. The findings indicate that myeloid replacement therapy by itself may not be an effective therapeutic strategy in Alzheimer’s disease. Immune cells of myeloid lineage are encountered in the Alzheimer’s disease (AD) brain, where they cluster around amyloid-β plaques. However, assigning functional roles to myeloid cell subtypes has been problematic, and the potential for peripheral myeloid cells to alleviate AD pathology remains unclear. Therefore, we asked whether replacement of brain-resident myeloid cells with peripheral monocytes alters amyloid deposition in two mouse models of cerebral β-amyloidosis (APP23 and APPPS1). Interestingly, early after repopulation, infiltrating monocytes neither clustered around plaques nor showed Trem2 expression. However, with increasing time in the brain, infiltrating monocytes became plaque associated and also Trem2 positive. Strikingly, however, monocyte repopulation for up to 6 mo did not modify amyloid load in either model, independent of the stage of pathology at the time of repopulation. Our results argue against a long-term role of peripheral monocytes that is sufficiently distinct from microglial function to modify cerebral β-amyloidosis. Therefore, myeloid replacement by itself is not likely to be effective as a therapeutic approach for AD.
Collapse
Affiliation(s)
- Nicholas H Varvel
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Stefan A Grathwohl
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Karoline Degenhardt
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Claudia Resch
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Andrea Bosch
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Jonas J Neher
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| |
Collapse
|
2950
|
Abstract
ATP-binding cassette transporter A7 (ABCA7) is highly expressed in the brain. Recent genome-wide association studies (GWAS) identify ABCA7 single nt polymorphisms (SNPs) that increase Alzheimer's disease (AD) risk. It is now important to understand the true function of ABCA7 in the AD context. We have begun to address this using in vitro and in vivo AD models. Our initial studies showed that transient overexpression of ABCA7 in Chinese hamster ovary cells stably expressing human amyloid precursor protein (APP) resulted in an approximate 50% inhibition in the production of the AD-related amyloid-β (Aβ) peptide as compared with mock-transfected cells. This increased ABCA7 expression was also associated with alterations in other markers of APP processing and an accumulation of cellular APP. To probe for a function of ABCA7 in vivo, we crossed Abca7−/− mice with J20 mice, an amyloidogenic transgenic AD mouse model [B6.Cg-Tg(PDGFB-APPSwInd)20Lms/J] expressing a mutant form of human APP bearing both the Swedish (K670N/M671L) and Indiana (V717F) familial AD mutations. We found that ABCA7 loss doubled insoluble Aβ levels and amyloid plaques in the brain. This did not appear to be related to changes in APP processing (C-terminal fragment analysis), which led us to assess other mechanism by which ABCA7 may modulate Aβ homoeostasis. As we have shown that microglia express high levels of ABCA7, we examined a role for ABCA7 in the phagocytic clearance of Aβ. Our data indicated that the capacity for bone marrow-derived macrophages derived from Abca7−/− mice to phagocytose Aβ was reduced by 51% compared with wild-type (WT) mice. This suggests ABCA7 plays a role in the regulation of Aβ homoeostasis in the brain and that this may be related to Aβ clearance by microglia.
Collapse
|