1
|
Ofori E, Solis A, Punjani N. The Association among Hypothalamic Subnits, Gonadotropic and Sex Hormone Plasmas Levels in Alzheimer's Disease. Brain Sci 2024; 14:276. [PMID: 38539664 PMCID: PMC10968390 DOI: 10.3390/brainsci14030276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 04/28/2024] Open
Abstract
This study investigates the sex-specific role of the Hypothalamic-Pituitary-Gonadal axis in Alzheimer's disease progression, utilizing ADNI1 data for 493 individuals, analyzing plasma levels of gonadotropic and sex hormones, and examining neurodegeneration-related brain structures. We assessed plasma levels of follicle stimulating hormone (FSH), luteinizing hormone (LH), progesterone (P4), and testosterone (T), along with volumetric measures of the hippocampus, entorhinal cortex, and hypothalamic subunits, to explore their correlation with Alzheimer's disease markers across different cognitive statuses and sexes. Significant cognitive status effects were observed for all volumetric measures, with a distinct sex-by-cognitive status interaction for hypothalamic volume, indicating a decrease in males but not in females across cognitive impairment stages. Regression analyses showed specific hypothalamic subunit volume related to hormone levels, accounting for up to approximately 40% of the variance (p < 0.05). The findings highlight sex differences in neurodegeneration and hormonal regulation, suggesting potential for personalized treatments and advancing the understanding of Alzheimer's disease etiology.
Collapse
Affiliation(s)
- Edward Ofori
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Anamaria Solis
- Department of Social Work, University of Texas at El Paso, El Paso, TX 79968, USA;
| | - Nahid Punjani
- College of Medicine and Sciences, Mayo Clinic, Phoenix, AZ 85054, USA
| | | |
Collapse
|
2
|
Khan SH, Perkins AJ, Jawaid S, Wang S, Lindroth H, Schmitt RE, Doles J, True JD, Gao S, Caplan GA, Twigg HL, Kesler K, Khan BA. Serum proteomic analysis in esophagectomy patients with postoperative delirium: A case-control study. Heart Lung 2024; 63:35-41. [PMID: 37748302 PMCID: PMC10843392 DOI: 10.1016/j.hrtlng.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/24/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Postoperative delirium occurs in up to 80% of patients undergoing esophagectomy. We performed an exploratory proteomic analysis to identify protein pathways that may be associated with delirium post-esophagectomy. OBJECTIVES Identify proteins associated with delirium and delirium severity in a younger and higher-risk surgical population. METHODS We performed a case-control study using blood samples collected from patients enrolled in a negative, randomized, double-blind clinical trial. English speaking adults aged 18 years or older, undergoing esophagectomy, who had blood samples obtained were included. Cases were defined by a positive delirium screen after surgery while controls were patients with negative delirium assessments. Delirium was assessed using Richmond Agitation Sedation Scale and Confusion Assessment Method for the Intensive Care Unit, and delirium severity was assessed by Delirium Rating Scale-Revised-98. Blood samples were collected pre-operatively and on post-operative day 1, and discovery proteomic analysis was performed. Between-group differences in median abundance ratios were reported using Wilcoxon-Mann-Whitney Odds (WMWodds1) test. RESULTS 52 (26 cases, 26 controls) patients were included in the study with a mean age of 64 (SD 9.6) years, 1.9% were females and 25% were African American. The median duration of delirium was 1 day (IQR: 1-2), and the median delirium/coma duration was 2.5 days (IQR: 2-4). Two proteins with greater relative abundance ratio in patients with delirium were: Coagulation factor IX (WMWodds: 1.89 95%CI: 1.0-4.2) and mannosyl-oligosaccharide 1,2-alpha-mannosidase (WMWodds: 2.4 95%CI: 1.03-9.9). Protein abundance ratios associated with mean delirium severity at postoperative day 1 were Complement C2 (Spearman rs = -0.31, 95%CI [-0.55, -0.02]) and Mannosyl-oligosaccharide 1,2-alpha-mannosidase (rs = 0.61, 95%CI = [0.29, 0.81]). CONCLUSIONS We identified changes in proteins associated with coagulation, inflammation, and protein handling; larger, follow-up studies are needed to confirm our hypothesis-generating findings.
Collapse
Affiliation(s)
- Sikandar H Khan
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA; Indiana University Center for Aging Research, Regenstrief Institute, Indianapolis, Indiana, USA; Indiana University Center of Health Innovation and Implementation Science, Indianapolis, Indiana, USA.
| | - Anthony J Perkins
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Samreen Jawaid
- Indiana University Center for Aging Research, Regenstrief Institute, Indianapolis, Indiana, USA
| | - Sophia Wang
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Heidi Lindroth
- Department of Nursing, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Rebecca E Schmitt
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jason Doles
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jason D True
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | - Sujuan Gao
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Gideon A Caplan
- Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia; Department of Geriatric Medicine, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Homer L Twigg
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kenneth Kesler
- Department of Cardiothoracic Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Babar A Khan
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA; Indiana University Center for Aging Research, Regenstrief Institute, Indianapolis, Indiana, USA; Indiana University Center of Health Innovation and Implementation Science, Indianapolis, Indiana, USA
| |
Collapse
|
3
|
Horgusluoglu E, Neff R, Song W, Wang M, Wang Q, Arnold M, Krumsiek J, Galindo‐Prieto B, Ming C, Nho K, Kastenmüller G, Han X, Baillie R, Zeng Q, Andrews S, Cheng H, Hao K, Goate A, Bennett DA, Saykin AJ, Kaddurah‐Daouk R, Zhang B. Integrative metabolomics-genomics approach reveals key metabolic pathways and regulators of Alzheimer's disease. Alzheimers Dement 2022; 18:1260-1278. [PMID: 34757660 PMCID: PMC9085975 DOI: 10.1002/alz.12468] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 12/29/2022]
Abstract
Metabolites, the biochemical products of the cellular process, can be used to measure alterations in biochemical pathways related to the pathogenesis of Alzheimer's disease (AD). However, the relationships between systemic abnormalities in metabolism and the pathogenesis of AD are poorly understood. In this study, we aim to identify AD-specific metabolomic changes and their potential upstream genetic and transcriptional regulators through an integrative systems biology framework for analyzing genetic, transcriptomic, metabolomic, and proteomic data in AD. Metabolite co-expression network analysis of the blood metabolomic data in the Alzheimer's Disease Neuroimaging Initiative (ADNI) shows short-chain acylcarnitines/amino acids and medium/long-chain acylcarnitines are most associated with AD clinical outcomes, including episodic memory scores and disease severity. Integration of the gene expression data in both the blood from the ADNI and the brain from the Accelerating Medicines Partnership Alzheimer's Disease (AMP-AD) program reveals ABCA1 and CPT1A are involved in the regulation of acylcarnitines and amino acids in AD. Gene co-expression network analysis of the AMP-AD brain RNA-seq data suggests the CPT1A- and ABCA1-centered subnetworks are associated with neuronal system and immune response, respectively. Increased ABCA1 gene expression and adiponectin protein, a regulator of ABCA1, correspond to decreased short-chain acylcarnitines and amines in AD in the ADNI. In summary, our integrated analysis of large-scale multiomics data in AD systematically identifies novel metabolites and their potential regulators in AD and the findings pave a way for not only developing sensitive and specific diagnostic biomarkers for AD but also identifying novel molecular mechanisms of AD pathogenesis.
Collapse
Affiliation(s)
- Emrin Horgusluoglu
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Ryan Neff
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Won‐Min Song
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Minghui Wang
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Qian Wang
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Matthias Arnold
- Institute of Computational BiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- Department of Psychiatry and Behavioral SciencesDuke UniversityDurhamNorth CarolinaUSA
| | - Jan Krumsiek
- Department of Physiology and BiophysicsWeill Cornell MedicineInstitute for Computational BiomedicineEnglander Institute for Precision MedicineNew YorkNew YorkUSA
| | - Beatriz Galindo‐Prieto
- Department of Physiology and BiophysicsWeill Cornell MedicineInstitute for Computational BiomedicineEnglander Institute for Precision MedicineNew YorkNew YorkUSA
- Helen and Robert Appel Alzheimer's Disease Research InstituteBrain and Mind Research InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Chen Ming
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences; Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Gabi Kastenmüller
- Institute of Computational BiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
| | - Xianlin Han
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | | | - Qi Zeng
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Shea Andrews
- Department of NeuroscienceRonald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Haoxiang Cheng
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Ke Hao
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Alison Goate
- Department of NeuroscienceRonald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences; Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rima Kaddurah‐Daouk
- Department of Psychiatry and Behavioral SciencesDuke UniversityDurhamNorth CarolinaUSA
- Duke Institute of Brain SciencesDuke UniversityDurhamNorth CarolinaUSA
- Department of MedicineDuke UniversityDurhamNorth CarolinaUSA
| | - Bin Zhang
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | | | | |
Collapse
|
4
|
Genetics meets proteomics: perspectives for large population-based studies. Nat Rev Genet 2020; 22:19-37. [PMID: 32860016 DOI: 10.1038/s41576-020-0268-2] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2020] [Indexed: 12/22/2022]
Abstract
Proteomic analysis of cells, tissues and body fluids has generated valuable insights into the complex processes influencing human biology. Proteins represent intermediate phenotypes for disease and provide insight into how genetic and non-genetic risk factors are mechanistically linked to clinical outcomes. Associations between protein levels and DNA sequence variants that colocalize with risk alleles for common diseases can expose disease-associated pathways, revealing novel drug targets and translational biomarkers. However, genome-wide, population-scale analyses of proteomic data are only now emerging. Here, we review current findings from studies of the plasma proteome and discuss their potential for advancing biomedical translation through the interpretation of genome-wide association analyses. We highlight the challenges faced by currently available technologies and provide perspectives relevant to their future application in large-scale biobank studies.
Collapse
|
5
|
Detecting genetic associations with brain imaging phenotypes in Alzheimer's disease via a novel structured SCCA approach. Med Image Anal 2020; 61:101656. [PMID: 32062154 DOI: 10.1016/j.media.2020.101656] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 11/27/2019] [Accepted: 01/22/2020] [Indexed: 01/15/2023]
Abstract
Brain imaging genetics becomes an important research topic since it can reveal complex associations between genetic factors and the structures or functions of the human brain. Sparse canonical correlation analysis (SCCA) is a popular bi-multivariate association identification method. To mine the complex genetic basis of brain imaging phenotypes, there arise many SCCA methods with a variety of norms for incorporating different structures of interest. They often use the group lasso penalty, the fused lasso or the graph/network guided fused lasso ones. However, the group lasso methods have limited capability because of the incomplete or unavailable prior knowledge in real applications. The fused lasso and graph/network guided methods are sensitive to the sign of the sample correlation which may be incorrectly estimated. In this paper, we introduce two new penalties to improve the fused lasso and the graph/network guided lasso penalties in structured sparse learning. We impose both penalties to the SCCA model and propose an optimization algorithm to solve it. The proposed SCCA method has a strong upper bound of grouping effects for both positively and negatively highly correlated variables. We show that, on both synthetic and real neuroimaging genetics data, the proposed SCCA method performs better than or equally to the conventional methods using fused lasso or graph/network guided fused lasso. In particular, the proposed method identifies higher canonical correlation coefficients and captures clearer canonical weight patterns, demonstrating its promising capability in revealing biologically meaningful imaging genetic associations.
Collapse
|
6
|
Solomon T, Lapek JD, Jensen SB, Greenwald WW, Hindberg K, Matsui H, Latysheva N, Braekken SK, Gonzalez DJ, Frazer KA, Smith EN, Hansen JB. Identification of Common and Rare Genetic Variation Associated With Plasma Protein Levels Using Whole-Exome Sequencing and Mass Spectrometry. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002170. [PMID: 30562114 DOI: 10.1161/circgen.118.002170] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Identifying genetic variation associated with plasma protein levels, and the mechanisms by which they act, could provide insight into alterable processes involved in regulation of protein levels. Although protein levels can be affected by genetic variants, their estimation can also be biased by missense variants in coding exons causing technical artifacts. Integrating genome sequence genotype data with mass spectrometry-based protein level estimation could reduce bias, thereby improving detection of variation that affects RNA or protein metabolism. METHODS Here, we integrate the blood plasma protein levels of 664 proteins from 165 participants of the Tromsø Study, measured via tandem mass tag mass spectrometry, with whole-exome sequencing data to identify common and rare genetic variation associated with peptide and protein levels (protein quantitative trait loci [pQTLs]). We additionally use literature and database searches to prioritize putative functional variants for each pQTL. RESULTS We identify 109 independent associations (36 protein and 73 peptide) and use genotype data to exclude 49 (4 protein and 45 peptide) as technical artifacts. We describe 2 particular cases of rare variation: 1 associated with the complement pathway and 1 with platelet degranulation. We identify putative functional variants and show that pQTLs act through diverse molecular mechanisms that affect both RNA and protein metabolism. CONCLUSIONS We show that although the majority of pQTLs exert their effects by modulating RNA metabolism, many affect protein levels directly. Our work demonstrates the extent by which pQTL studies are affected by technical artifacts and highlights how prioritizing the functional variant in pQTL studies can lead to insights into the molecular steps by which a protein may be regulated.
Collapse
Affiliation(s)
- Terry Solomon
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.)
| | - John D Lapek
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla (J.D.L., D.J.G.)
| | - Søren Beck Jensen
- Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Center, UiT-The Arctic University of Norway (S.B.J., K.H., N.L., S.K.B., K.A.F., E.N.S., J.-B.H.)
| | - William W Greenwald
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla (W.W.G.)
| | - Kristian Hindberg
- Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Center, UiT-The Arctic University of Norway (S.B.J., K.H., N.L., S.K.B., K.A.F., E.N.S., J.-B.H.)
| | - Hiroko Matsui
- Institue of Genomic Medicine, University of California, San Diego, La Jolla (H.M., K.A.F.)
| | - Nadezhda Latysheva
- Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Center, UiT-The Arctic University of Norway (S.B.J., K.H., N.L., S.K.B., K.A.F., E.N.S., J.-B.H.)
| | - Sigrid K Braekken
- Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Center, UiT-The Arctic University of Norway (S.B.J., K.H., N.L., S.K.B., K.A.F., E.N.S., J.-B.H.).,Division of Internal Medicine, University Hospital of North Norway, Tromsû (S.K.B., J.-B.H.)
| | - David J Gonzalez
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla (J.D.L., D.J.G.)
| | - Kelly A Frazer
- Institue of Genomic Medicine, University of California, San Diego, La Jolla (H.M., K.A.F.).,Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla (K.A.F., E.N.S.).,Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Center, UiT-The Arctic University of Norway (S.B.J., K.H., N.L., S.K.B., K.A.F., E.N.S., J.-B.H.)
| | - Erin N Smith
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla (K.A.F., E.N.S.).,Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Center, UiT-The Arctic University of Norway (S.B.J., K.H., N.L., S.K.B., K.A.F., E.N.S., J.-B.H.)
| | - John-Bjarne Hansen
- Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Center, UiT-The Arctic University of Norway (S.B.J., K.H., N.L., S.K.B., K.A.F., E.N.S., J.-B.H.).,Division of Internal Medicine, University Hospital of North Norway, Tromsû (S.K.B., J.-B.H.)
| |
Collapse
|
7
|
Yao X, Cong S, Yan J, Risacher SL, Saykin AJ, Moore JH, Shen L. Mining Regional Imaging Genetic Associations via Voxel-wise Enrichment Analysis. ... IEEE-EMBS INTERNATIONAL CONFERENCE ON BIOMEDICAL AND HEALTH INFORMATICS. IEEE-EMBS INTERNATIONAL CONFERENCE ON BIOMEDICAL AND HEALTH INFORMATICS 2019; 2019. [PMID: 31742256 DOI: 10.1109/bhi.2019.8834450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Brain imaging genetics aims to reveal genetic effects on brain phenotypes, where most studies examine phenotypes defined on anatomical or functional regions of interest (ROIs) given their biologically meaningful annotation and modest dimensionality compared with voxel-wise approaches. Typical ROI-level measures used in these studies are summary statistics from voxel-wise measures in the region, without making full use of individual voxel signals. In this paper, we propose a flexible and powerful framework for mining regional imaging genetic associations via voxel-wise enrichment analysis, which embraces the collective effect of weak voxel-level signals within an ROI. We demonstrate our method on an imaging genetic analysis using data from the Alzheimers Disease Neuroimaging Initiative, where we assess the collective regional genetic effects of voxel-wise FDGPET measures between 116 ROIs and 19 AD candidate SNPs. Compared with traditional ROI-wise and voxel-wise approaches, our method identified 102 additional significant associations, some of which were further supported by evidences in brain tissue-specific expression analysis. This demonstrates the promise of the proposed method as a flexible and powerful framework for exploring imaging genetic effects on the brain.
Collapse
Affiliation(s)
- Xiaohui Yao
- Biostatistics, Epidemiology and Informatics University of Pennsylvania, Philadelphia, PA
| | - Shan Cong
- Electrical and Computer Engineering Purdue University, West Lafayette, IN
| | - Jingwen Yan
- Informatics and Computing Indiana University, Indianapolis, IN
| | - Shannon L Risacher
- Radiology and Imaging Sciences Indiana University School of Medicine, Indianapolis, IN
| | - Andrew J Saykin
- Radiology and Imaging Sciences Indiana University School of Medicine, Indianapolis, IN
| | - Jason H Moore
- Biostatistics, Epidemiology and Informatics University of Pennsylvania, Philadelphia, PA
| | - Li Shen
- Biostatistics, Epidemiology and Informatics University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
8
|
Yao X, Risacher SL, Nho K, Saykin AJ, Wang Z, Shen L. Targeted genetic analysis of cerebral blood flow imaging phenotypes implicates the INPP5D gene. Neurobiol Aging 2019; 81:213-221. [PMID: 31319229 PMCID: PMC6732252 DOI: 10.1016/j.neurobiolaging.2019.06.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 06/05/2019] [Accepted: 06/08/2019] [Indexed: 12/22/2022]
Abstract
The vascular hypothesis of Alzheimer's disease (AD) has proposed the involvement of brain hypoperfusion in AD pathogenesis, where cognitive decline and dysfunction result from dwindling cerebral blood flow (CBF). Based on the vascular hypothesis of Alzheimer's disease, we focused on exploring how genetic factors influence AD pathogenesis via the cerebrovascular system. To investigate the role of CBF endophenotypes in AD pathogenesis, we performed a targeted genetic analysis of 258 subjects from the Alzheimer's Disease Neuroimaging Initiative cohort to examine associations between 4033 single-nucleotide polymorphisms of 24 AD genes and CBF measures in 4 brain regions. A novel association with CBF measure in the left angular gyrus was identified in an INPP5D single-nucleotide polymorphism (i.e., rs61068452; p = 1.48E-7; corrected p = 2.39E-3). The gene-based analysis discovered both INPP5D and CD2AP associated with the left angular gyrus CBF. Further analyses on nonoverlapping samples revealed that rs61068452-G was associated with lower CSF t-tau/Aβ1-42 ratio. Our findings suggest a protective role of rs61068452-G in an AD-relevant cerebrovascular endophenotype, which has the potential to provide novel insights for better mechanistic understanding of AD.
Collapse
Affiliation(s)
- Xiaohui Yao
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ze Wang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Nho K, Kueider-Paisley A, Ahmad S, MahmoudianDehkordi S, Arnold M, Risacher SL, Louie G, Blach C, Baillie R, Han X, Kastenmüller G, Trojanowski JQ, Shaw LM, Weiner MW, Doraiswamy PM, van Duijn C, Saykin AJ, Kaddurah-Daouk R. Association of Altered Liver Enzymes With Alzheimer Disease Diagnosis, Cognition, Neuroimaging Measures, and Cerebrospinal Fluid Biomarkers. JAMA Netw Open 2019; 2:e197978. [PMID: 31365104 PMCID: PMC6669786 DOI: 10.1001/jamanetworkopen.2019.7978] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
IMPORTANCE Increasing evidence suggests an important role of liver function in the pathophysiology of Alzheimer disease (AD). The liver is a major metabolic hub; therefore, investigating the association of liver function with AD, cognition, neuroimaging, and CSF biomarkers would improve the understanding of the role of metabolic dysfunction in AD. OBJECTIVE To examine whether liver function markers are associated with cognitive dysfunction and the "A/T/N" (amyloid, tau, and neurodegeneration) biomarkers for AD. DESIGN, SETTING, AND PARTICIPANTS In this cohort study, serum-based liver function markers were measured from September 1, 2005, to August 31, 2013, in 1581 AD Neuroimaging Initiative participants along with cognitive measures, cerebrospinal fluid (CSF) biomarkers, brain atrophy, brain glucose metabolism, and amyloid-β accumulation. Associations of liver function markers with AD-associated clinical and A/T/N biomarkers were assessed using generalized linear models adjusted for confounding variables and multiple comparisons. Statistical analysis was performed from November 1, 2017, to February 28, 2019. EXPOSURES Five serum-based liver function markers (total bilirubin, albumin, alkaline phosphatase, alanine aminotransferase, and aspartate aminotransferase) from AD Neuroimaging Initiative participants were used as exposure variables. MAIN OUTCOMES AND MEASURES Primary outcomes included diagnosis of AD, composite scores for executive functioning and memory, CSF biomarkers, atrophy measured by magnetic resonance imaging, brain glucose metabolism measured by fludeoxyglucose F 18 (18F) positron emission tomography, and amyloid-β accumulation measured by [18F]florbetapir positron emission tomography. RESULTS Participants in the AD Neuroimaging Initiative (n = 1581; 697 women and 884 men; mean [SD] age, 73.4 [7.2] years) included 407 cognitively normal older adults, 20 with significant memory concern, 298 with early mild cognitive impairment, 544 with late mild cognitive impairment, and 312 with AD. An elevated aspartate aminotransferase (AST) to alanine aminotransferase (ALT) ratio and lower levels of ALT were associated with AD diagnosis (AST to ALT ratio: odds ratio, 7.932 [95% CI, 1.673-37.617]; P = .03; ALT: odds ratio, 0.133 [95% CI, 0.042-0.422]; P = .004) and poor cognitive performance (AST to ALT ratio: β [SE], -0.465 [0.180]; P = .02 for memory composite score; β [SE], -0.679 [0.215]; P = .006 for executive function composite score; ALT: β [SE], 0.397 [0.128]; P = .006 for memory composite score; β [SE], 0.637 [0.152]; P < .001 for executive function composite score). Increased AST to ALT ratio values were associated with lower CSF amyloid-β 1-42 levels (β [SE], -0.170 [0.061]; P = .04) and increased amyloid-β deposition (amyloid biomarkers), higher CSF phosphorylated tau181 (β [SE], 0.175 [0.055]; P = .02) (tau biomarkers) and higher CSF total tau levels (β [SE], 0.160 [0.049]; P = .02) and reduced brain glucose metabolism (β [SE], -0.123 [0.042]; P = .03) (neurodegeneration biomarkers). Lower levels of ALT were associated with increased amyloid-β deposition (amyloid biomarkers), and reduced brain glucose metabolism (β [SE], 0.096 [0.030]; P = .02) and greater atrophy (neurodegeneration biomarkers). CONCLUSIONS AND RELEVANCE Consistent associations of serum-based liver function markers with cognitive performance and A/T/N biomarkers for AD highlight the involvement of metabolic disturbances in the pathophysiology of AD. Further studies are needed to determine if these associations represent a causative or secondary role. Liver enzyme involvement in AD opens avenues for novel diagnostics and therapeutics.
Collapse
Affiliation(s)
- Kwangsik Nho
- Center for Computational Biology and Bioinformatics, Indiana Alzheimer Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis
| | | | - Shahzad Ahmad
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | | | - Matthias Arnold
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Shannon L. Risacher
- Center for Computational Biology and Bioinformatics, Indiana Alzheimer Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis
| | - Gregory Louie
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina
| | - Colette Blach
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | | | - Xianlin Han
- University of Texas Health Science Center at San Antonio, San Antonio
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia
| | - Michael W. Weiner
- Center for Imaging of Neurodegenerative Diseases, Department of Radiology, San Francisco Veterans Affairs Medical Center and University of California, San Francisco
| | - P. Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina
- Duke Institute of Brain Sciences, Duke University, Durham, North Carolina
- Department of Medicine, Duke University, Durham, North Carolina
| | - Cornelia van Duijn
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
- Nuffield Department of Population Health, Oxford University, Oxford, United Kingdom
| | - Andrew J. Saykin
- Center for Computational Biology and Bioinformatics, Indiana Alzheimer Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina
- Duke Institute of Brain Sciences, Duke University, Durham, North Carolina
- Department of Medicine, Duke University, Durham, North Carolina
| |
Collapse
|
10
|
The genesis and evolution of bead-based multiplexing. Methods 2019; 158:2-11. [DOI: 10.1016/j.ymeth.2019.01.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/10/2018] [Accepted: 01/14/2019] [Indexed: 12/22/2022] Open
|
11
|
Reischer T, Balendran-Braun S, Liebmann-Reindl S, Streubel B, Umek W, Koelbl H, Koch M. Genetic association in female stress urinary incontinence based on proteomic findings: a case-control study. Int Urogynecol J 2019; 31:117-122. [PMID: 30715578 PMCID: PMC6949200 DOI: 10.1007/s00192-019-03878-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/14/2019] [Indexed: 12/13/2022]
Abstract
Introduction and hypothesis Previous studies have indicated a hereditary component of stress urinary incontinence; however, evidence on candidate genes or single-nucleotide polymorphisms (SNPs) is scarce. We hypothesize a genetic association of female stress urinary incontinence based on significant differences of the urinary and serum proteomic pattern in the identical study population. Methods Case-control study of 19 patients and 19 controls. We searched for known SNPs of SUI candidate genes (COL1A1, MMP1, SERPINA5, UMOD) in the database of short genetic variations and PubMed. Genomic DNA was isolated using QIAamp DNA Blood Midi Kit (Qiagen). We performed Sanger sequencing of selected exons and introns. Results The rs885786 SNP of the SERPINA5 gene was identified in 15 cases and 10 controls (p = 0.09). The rs6113 SNP of the SERPINA5 gene was present in 4 controls compared to 0 cases (p = 0.105). The rs4293393, rs13333226 and rs13335818 SNPs of the UMOD gene were identified in five cases and two controls (p = 0.20), the rs1800012 SNP of the COL1A1 gene in five cases versus four controls (p = 0.24) and the homozygous rs1799750 SNP of the MMP1 gene in eight cases versus five controls (p = 0.18). The combination of the rs885786 SNP of the SERPINA5 gene and rs179970 SNP of the MMP1 gene was detected in ten cases versus five controls (p = 0.072). Conclusions We found nonsignificant trends toward associations of SNPs on the SERPINA5, UMOD and MMP1 gene and SUI. Electronic supplementary material The online version of this article (10.1007/s00192-019-03878-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Theresa Reischer
- Department of Obstetrics and Gynecology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | | | | | - Berthold Streubel
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Umek
- Department of Obstetrics and Gynecology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria.,Karl Landsteiner Institut fuer Spezielle Gynaekologie und Geburtshilfe, Vienna, Austria
| | - Heinz Koelbl
- Department of Obstetrics and Gynecology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Marianne Koch
- Department of Obstetrics and Gynecology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria. .,Karl Landsteiner Institut fuer Spezielle Gynaekologie und Geburtshilfe, Vienna, Austria.
| |
Collapse
|
12
|
Nho K, Kueider-Paisley A, MahmoudianDehkordi S, Arnold M, Risacher SL, Louie G, Blach C, Baillie R, Han X, Kastenmüller G, Jia W, Xie G, Ahmad S, Hankemeier T, van Duijn CM, Trojanowski JQ, Shaw LM, Weiner MW, Doraiswamy PM, Saykin AJ, Kaddurah-Daouk R. Altered bile acid profile in mild cognitive impairment and Alzheimer's disease: Relationship to neuroimaging and CSF biomarkers. Alzheimers Dement 2019; 15:232-244. [PMID: 30337152 PMCID: PMC6454538 DOI: 10.1016/j.jalz.2018.08.012] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/03/2018] [Accepted: 08/21/2018] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Bile acids (BAs) are the end products of cholesterol metabolism produced by human and gut microbiome co-metabolism. Recent evidence suggests gut microbiota influence pathological features of Alzheimer's disease (AD) including neuroinflammation and amyloid-β deposition. METHOD Serum levels of 20 primary and secondary BA metabolites from the AD Neuroimaging Initiative (n = 1562) were measured using targeted metabolomic profiling. We assessed the association of BAs with the "A/T/N" (amyloid, tau, and neurodegeneration) biomarkers for AD: cerebrospinal fluid (CSF) biomarkers, atrophy (magnetic resonance imaging), and brain glucose metabolism ([18F]FDG PET). RESULTS Of 23 BAs and relevant calculated ratios after quality control procedures, three BA signatures were associated with CSF Aβ1-42 ("A") and three with CSF p-tau181 ("T") (corrected P < .05). Furthermore, three, twelve, and fourteen BA signatures were associated with CSF t-tau, glucose metabolism, and atrophy ("N"), respectively (corrected P < .05). DISCUSSION This is the first study to show serum-based BA metabolites are associated with "A/T/N" AD biomarkers, providing further support for a role of BA pathways in AD pathophysiology. Prospective clinical observations and validation in model systems are needed to assess causality and specific mechanisms underlying this association.
Collapse
Affiliation(s)
- Kwangsik Nho
- Department of Radiology and Imaging Sciences, Center for Computational Biology and Bioinformatics, and the Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Matthias Arnold
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA; Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Center for Computational Biology and Bioinformatics, and the Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gregory Louie
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Colette Blach
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | | | - Xianlin Han
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Wei Jia
- University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Guoxiang Xie
- University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Shahzad Ahmad
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Thomas Hankemeier
- Division of Analytical Biosciences, Leiden Academic Centre for Drug Research, Leiden University, RA Leiden, the Netherlands
| | | | - John Q Trojanowski
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leslie M Shaw
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael W Weiner
- Center for Imaging of Neurodegenerative Diseases, Department of Radiology, San Francisco VA Medical Center/University of California San Francisco, San Francisco, CA, USA
| | - P Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA; Duke Institute of Brain Sciences, Duke University, Durham, NC, USA; Department of Medicine, Duke University, Durham, NC, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Center for Computational Biology and Bioinformatics, and the Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA; Duke Institute of Brain Sciences, Duke University, Durham, NC, USA; Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
13
|
Peña-Bautista C, Baquero M, Vento M, Cháfer-Pericás C. Omics-based Biomarkers for the Early Alzheimer Disease Diagnosis and Reliable Therapeutic Targets Development. Curr Neuropharmacol 2019; 17:630-647. [PMID: 30255758 PMCID: PMC6712290 DOI: 10.2174/1570159x16666180926123722] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 08/31/2018] [Accepted: 09/19/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD), the most common cause of dementia in adulthood, has great medical, social, and economic impact worldwide. Available treatments result in symptomatic relief, and most of them are indicated from the early stages of the disease. Therefore, there is an increasing body of research developing accurate and early diagnoses, as well as diseasemodifying therapies. OBJECTIVE Advancing the knowledge of AD physiopathological mechanisms, improving early diagnosis and developing effective treatments from omics-based biomarkers. METHODS Studies using omics technologies to detect early AD, were reviewed with a particular focus on the metabolites/lipids, micro-RNAs and proteins, which are identified as potential biomarkers in non-invasive samples. RESULTS This review summarizes recent research on metabolomics/lipidomics, epigenomics and proteomics, applied to early AD detection. Main research lines are the study of metabolites from pathways, such as lipid, amino acid and neurotransmitter metabolisms, cholesterol biosynthesis, and Krebs and urea cycles. In addition, some microRNAs and proteins (microglobulins, interleukins), related to a common network with amyloid precursor protein and tau, have been also identified as potential biomarkers. Nevertheless, the reproducibility of results among studies is not good enough and a standard methodological approach is needed in order to obtain accurate information. CONCLUSION The assessment of metabolomic/lipidomic, epigenomic and proteomic changes associated with AD to identify early biomarkers in non-invasive samples from well-defined participants groups will potentially allow the advancement in the early diagnosis and improvement of therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - Consuelo Cháfer-Pericás
- Address correspondence to this author at the Health Research Institute La Fe, Avda de Fernando Abril Martorell, 106; 46026 Valencia, Spain;Tel: +34 96 124 66 61; Fax: + 34 96 124 57 46; E-mail:
| |
Collapse
|
14
|
Lemche E. Early Life Stress and Epigenetics in Late-onset Alzheimer's Dementia: A Systematic Review. Curr Genomics 2018; 19:522-602. [PMID: 30386171 PMCID: PMC6194433 DOI: 10.2174/1389202919666171229145156] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 07/27/2017] [Accepted: 12/12/2017] [Indexed: 11/22/2022] Open
Abstract
Involvement of life stress in Late-Onset Alzheimer's Disease (LOAD) has been evinced in longitudinal cohort epidemiological studies, and endocrinologic evidence suggests involvements of catecholamine and corticosteroid systems in LOAD. Early Life Stress (ELS) rodent models have successfully demonstrated sequelae of maternal separation resulting in LOAD-analogous pathology, thereby supporting a role of insulin receptor signalling pertaining to GSK-3beta facilitated tau hyper-phosphorylation and amyloidogenic processing. Discussed are relevant ELS studies, and findings from three mitogen-activated protein kinase pathways (JNK/SAPK pathway, ERK pathway, p38/MAPK pathway) relevant for mediating environmental stresses. Further considered were the roles of autophagy impairment, neuroinflammation, and brain insulin resistance. For the meta-analytic evaluation, 224 candidate gene loci were extracted from reviews of animal studies of LOAD pathophysiological mechanisms, of which 60 had no positive results in human LOAD association studies. These loci were combined with 89 gene loci confirmed as LOAD risk genes in previous GWAS and WES. Of the 313 risk gene loci evaluated, there were 35 human reports on epigenomic modifications in terms of methylation or histone acetylation. 64 microRNA gene regulation mechanisms were published for the compiled loci. Genomic association studies support close relations of both noradrenergic and glucocorticoid systems with LOAD. For HPA involvement, a CRHR1 haplotype with MAPT was described, but further association of only HSD11B1 with LOAD found; however, association of FKBP1 and NC3R1 polymorphisms was documented in support of stress influence to LOAD. In the brain insulin system, IGF2R, INSR, INSRR, and plasticity regulator ARC, were associated with LOAD. Pertaining to compromised myelin stability in LOAD, relevant associations were found for BIN1, RELN, SORL1, SORCS1, CNP, MAG, and MOG. Regarding epigenetic modifications, both methylation variability and de-acetylation were reported for LOAD. The majority of up-to-date epigenomic findings include reported modifications in the well-known LOAD core pathology loci MAPT, BACE1, APP (with FOS, EGR1), PSEN1, PSEN2, and highlight a central role of BDNF. Pertaining to ELS, relevant loci are FKBP5, EGR1, GSK3B; critical roles of inflammation are indicated by CRP, TNFA, NFKB1 modifications; for cholesterol biosynthesis, DHCR24; for myelin stability BIN1, SORL1, CNP; pertaining to (epi)genetic mechanisms, hTERT, MBD2, DNMT1, MTHFR2. Findings on gene regulation were accumulated for BACE1, MAPK signalling, TLR4, BDNF, insulin signalling, with most reports for miR-132 and miR-27. Unclear in epigenomic studies remains the role of noradrenergic signalling, previously demonstrated by neuropathological findings of childhood nucleus caeruleus degeneration for LOAD tauopathy.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| |
Collapse
|
15
|
Emilsson V, Ilkov M, Lamb JR, Finkel N, Gudmundsson EF, Pitts R, Hoover H, Gudmundsdottir V, Horman SR, Aspelund T, Shu L, Trifonov V, Sigurdsson S, Manolescu A, Zhu J, Olafsson Ö, Jakobsdottir J, Lesley SA, To J, Zhang J, Harris TB, Launer LJ, Zhang B, Eiriksdottir G, Yang X, Orth AP, Jennings LL, Gudnason V. Co-regulatory networks of human serum proteins link genetics to disease. Science 2018; 361:769-773. [PMID: 30072576 PMCID: PMC6190714 DOI: 10.1126/science.aaq1327] [Citation(s) in RCA: 364] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/07/2018] [Accepted: 07/13/2018] [Indexed: 12/25/2022]
Abstract
Proteins circulating in the blood are critical for age-related disease processes; however, the serum proteome has remained largely unexplored. To this end, 4137 proteins covering most predicted extracellular proteins were measured in the serum of 5457 Icelanders over 65 years of age. Pairwise correlation between proteins as they varied across individuals revealed 27 different network modules of serum proteins, many of which were associated with cardiovascular and metabolic disease states, as well as overall survival. The protein modules were controlled by cis- and trans-acting genetic variants, which in many cases were also associated with complex disease. This revealed co-regulated groups of circulating proteins that incorporated regulatory control between tissues and demonstrated close relationships to past, current, and future disease states.
Collapse
Affiliation(s)
- Valur Emilsson
- Icelandic Heart Association, Holtasmari 1, IS-201 Kopavogur, Iceland.
- Faculty of Pharmacology, University of Iceland, 101 Reykjavik, Iceland
| | - Marjan Ilkov
- Icelandic Heart Association, Holtasmari 1, IS-201 Kopavogur, Iceland
| | - John R Lamb
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA.
| | - Nancy Finkel
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | | | - Rebecca Pitts
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Heather Hoover
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | | | - Shane R Horman
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Thor Aspelund
- Icelandic Heart Association, Holtasmari 1, IS-201 Kopavogur, Iceland
- Centre of Public Health Sciences, University of Iceland, 101 Reykjavik, Iceland
| | - Le Shu
- Department of Integrative Biology and Physiology, University of California, Los Angeles CA, USA
| | - Vladimir Trifonov
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | | | - Andrei Manolescu
- School of Science and Engineering, Mentavegur 1, IS-101, Reykjavik University, 101 Reykjavik, Iceland
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Örn Olafsson
- Icelandic Heart Association, Holtasmari 1, IS-201 Kopavogur, Iceland
| | | | - Scott A Lesley
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Jeremy To
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Jia Zhang
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, MD 20892-9205, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, MD 20892-9205, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles CA, USA
| | - Anthony P Orth
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Lori L Jennings
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Holtasmari 1, IS-201 Kopavogur, Iceland.
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| |
Collapse
|
16
|
Timasheva YR, Nasibullin TR, Tuktarova IA, Erdman VV, Mustafina OE. CXCL13 polymorphism is associated with essential hypertension in Tatars from Russia. Mol Biol Rep 2018; 45:1557-1564. [PMID: 30019153 DOI: 10.1007/s11033-018-4257-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/12/2018] [Indexed: 01/10/2023]
Abstract
Essential arterial hypertension is a disease with distinct yet unexplored inflammatory component. Our aim was to assess the role of chemokine genes and their interaction in its development. Genotyping of polymorphic markers in six chemokine genes (CXCL13, CCL8, CCL16, CCL17, CCL18, and CCL23) was performed in the group of 522 men of Tatar ethnic origin from the Republic of Bashkortostan, Russia (213 patients with essential hypertension and 309 healthy individuals without history of cardiovascular disease). We found a strong association of CXCL13 rs355689*C allele with essential hypertension under additive (OR 0.56, PFDR = 0.008) and dominant (OR 0.41, PFDR 4.38 × 10- 4) genetic model. The analysis of gene-gene interactions revealed 12 allele/genotype combinations that remained significantly associated with essential hypertension after correction for multiple testing was applied, and each of these combinations included CXCL13 rs355689 polymorphism. Our results indicate that CXCL13 rs355689 polymorphism is strongly associated with essential hypertension in the ethnic group of Tatars, alone and in combination with polymorphic markers in other chemokine genes.
Collapse
Affiliation(s)
- Yanina R Timasheva
- Laboratory of Physiological Genetics, Institute of Biochemistry and Genetics of Ufa Federal Research Centre of Russian Academy of Sciences, October Avenue 71, Ufa, Russian Federation, 450054. .,Department of Medical Genetics and Fundamental Medicine, Bashkir State Medical University, Ufa, Russian Federation.
| | - Timur R Nasibullin
- Laboratory of Physiological Genetics, Institute of Biochemistry and Genetics of Ufa Federal Research Centre of Russian Academy of Sciences, October Avenue 71, Ufa, Russian Federation, 450054
| | - Ilsiyar A Tuktarova
- Laboratory of Physiological Genetics, Institute of Biochemistry and Genetics of Ufa Federal Research Centre of Russian Academy of Sciences, October Avenue 71, Ufa, Russian Federation, 450054
| | - Vera V Erdman
- Laboratory of Physiological Genetics, Institute of Biochemistry and Genetics of Ufa Federal Research Centre of Russian Academy of Sciences, October Avenue 71, Ufa, Russian Federation, 450054
| | - Olga E Mustafina
- Laboratory of Physiological Genetics, Institute of Biochemistry and Genetics of Ufa Federal Research Centre of Russian Academy of Sciences, October Avenue 71, Ufa, Russian Federation, 450054.,Department of Genetics and Fundamental Medicine, Bashkir State University, Ufa, Russian Federation
| |
Collapse
|
17
|
Zigon B, Li H, Yao X, Fang S, Hasan MA, Yan J, Moore JH, Saykin AJ, Shen L. GPU Accelerated Browser for Neuroimaging Genomics. Neuroinformatics 2018; 16:393-402. [PMID: 29691798 DOI: 10.1007/s12021-018-9376-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Neuroimaging genomics is an emerging field that provides exciting opportunities to understand the genetic basis of brain structure and function. The unprecedented scale and complexity of the imaging and genomics data, however, have presented critical computational bottlenecks. In this work we present our initial efforts towards building an interactive visual exploratory system for mining big data in neuroimaging genomics. A GPU accelerated browsing tool for neuroimaging genomics is created that implements the ANOVA algorithm for single nucleotide polymorphism (SNP) based analysis and the VEGAS algorithm for gene-based analysis, and executes them at interactive rates. The ANOVA algorithm is 110 times faster than the 4-core OpenMP version, while the VEGAS algorithm is 375 times faster than its 4-core OpenMP counter part. This approach lays a solid foundation for researchers to address the challenges of mining large-scale imaging genomics datasets via interactive visual exploration.
Collapse
Affiliation(s)
- Bob Zigon
- Beckman Coulter, Indianapolis, IN, 46268, USA.
| | - Huang Li
- Department of Computer Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Xiaohui Yao
- Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Shiaofen Fang
- Department of Computer Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Mohammad Al Hasan
- Department of Computer Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Jingwen Yan
- Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Jason H Moore
- Department of Biostatistics, Epidemiology, Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, IU School of Medicine, Indianapolis, IN, 46202, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology, Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
18
|
Du L, Liu K, Zhang T, Yao X, Yan J, Risacher SL, Han J, Guo L, Saykin AJ, Shen L. A novel SCCA approach via truncated ℓ1-norm and truncated group lasso for brain imaging genetics. Bioinformatics 2018; 34:278-285. [PMID: 28968815 PMCID: PMC5860211 DOI: 10.1093/bioinformatics/btx594] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 08/21/2017] [Accepted: 09/15/2017] [Indexed: 02/06/2023] Open
Abstract
MOTIVATION Brain imaging genetics, which studies the linkage between genetic variations and structural or functional measures of the human brain, has become increasingly important in recent years. Discovering the bi-multivariate relationship between genetic markers such as single-nucleotide polymorphisms (SNPs) and neuroimaging quantitative traits (QTs) is one major task in imaging genetics. Sparse Canonical Correlation Analysis (SCCA) has been a popular technique in this area for its powerful capability in identifying bi-multivariate relationships coupled with feature selection. The existing SCCA methods impose either the ℓ1-norm or its variants to induce sparsity. The ℓ0-norm penalty is a perfect sparsity-inducing tool which, however, is an NP-hard problem. RESULTS In this paper, we propose the truncated ℓ1-norm penalized SCCA to improve the performance and effectiveness of the ℓ1-norm based SCCA methods. Besides, we propose an efficient optimization algorithms to solve this novel SCCA problem. The proposed method is an adaptive shrinkage method via tuning τ. It can avoid the time intensive parameter tuning if given a reasonable small τ. Furthermore, we extend it to the truncated group-lasso (TGL), and propose TGL-SCCA model to improve the group-lasso-based SCCA methods. The experimental results, compared with four benchmark methods, show that our SCCA methods identify better or similar correlation coefficients, and better canonical loading profiles than the competing methods. This demonstrates the effectiveness and efficiency of our methods in discovering interesting imaging genetic associations. AVAILABILITY AND IMPLEMENTATION The Matlab code and sample data are freely available at http://www.iu.edu/∼shenlab/tools/tlpscca/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Lei Du
- Department of Control and Information, School of Automation, Northwestern Polytechnical University, Xi’an, China
| | - Kefei Liu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Tuo Zhang
- Department of Control and Information, School of Automation, Northwestern Polytechnical University, Xi’an, China
| | - Xiaohui Yao
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Jingwen Yan
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Junwei Han
- Department of Control and Information, School of Automation, Northwestern Polytechnical University, Xi’an, China
| | - Lei Guo
- Department of Control and Information, School of Automation, Northwestern Polytechnical University, Xi’an, China
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Li Shen
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | | |
Collapse
|
19
|
Pattern Discovery in Brain Imaging Genetics via SCCA Modeling with a Generic Non-convex Penalty. Sci Rep 2017; 7:14052. [PMID: 29070790 PMCID: PMC5656688 DOI: 10.1038/s41598-017-13930-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 10/02/2017] [Indexed: 01/21/2023] Open
Abstract
Brain imaging genetics intends to uncover associations between genetic markers and neuroimaging quantitative traits. Sparse canonical correlation analysis (SCCA) can discover bi-multivariate associations and select relevant features, and is becoming popular in imaging genetic studies. The L1-norm function is not only convex, but also singular at the origin, which is a necessary condition for sparsity. Thus most SCCA methods impose [Formula: see text]-norm onto the individual feature or the structure level of features to pursuit corresponding sparsity. However, the [Formula: see text]-norm penalty over-penalizes large coefficients and may incurs estimation bias. A number of non-convex penalties are proposed to reduce the estimation bias in regression tasks. But using them in SCCA remains largely unexplored. In this paper, we design a unified non-convex SCCA model, based on seven non-convex functions, for unbiased estimation and stable feature selection simultaneously. We also propose an efficient optimization algorithm. The proposed method obtains both higher correlation coefficients and better canonical loading patterns. Specifically, these SCCA methods with non-convex penalties discover a strong association between the APOE e4 rs429358 SNP and the hippocampus region of the brain. They both are Alzheimer's disease related biomarkers, indicating the potential and power of the non-convex methods in brain imaging genetics.
Collapse
|
20
|
Yao X, Yan J, Liu K, Kim S, Nho K, Risacher SL, Greene CS, Moore JH, Saykin AJ, Shen L. Tissue-specific network-based genome wide study of amygdala imaging phenotypes to identify functional interaction modules. Bioinformatics 2017; 33:3250-3257. [PMID: 28575147 PMCID: PMC6410887 DOI: 10.1093/bioinformatics/btx344] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 04/16/2017] [Accepted: 05/26/2017] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Network-based genome-wide association studies (GWAS) aim to identify functional modules from biological networks that are enriched by top GWAS findings. Although gene functions are relevant to tissue context, most existing methods analyze tissue-free networks without reflecting phenotypic specificity. RESULTS We propose a novel module identification framework for imaging genetic studies using the tissue-specific functional interaction network. Our method includes three steps: (i) re-prioritize imaging GWAS findings by applying machine learning methods to incorporate network topological information and enhance the connectivity among top genes; (ii) detect densely connected modules based on interactions among top re-prioritized genes; and (iii) identify phenotype-relevant modules enriched by top GWAS findings. We demonstrate our method on the GWAS of [18F]FDG-PET measures in the amygdala region using the imaging genetic data from the Alzheimer's Disease Neuroimaging Initiative, and map the GWAS results onto the amygdala-specific functional interaction network. The proposed network-based GWAS method can effectively detect densely connected modules enriched by top GWAS findings. Tissue-specific functional network can provide precise context to help explore the collective effects of genes with biologically meaningful interactions specific to the studied phenotype. AVAILABILITY AND IMPLEMENTATION The R code and sample data are freely available at http://www.iu.edu/shenlab/tools/gwasmodule/. CONTACT shenli@iu.edu. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Xiaohui Yao
- Department of BioHealth Informatics, Indiana University School of Informatics & Computing, Indianapolis, IN 46202, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jingwen Yan
- Department of BioHealth Informatics, Indiana University School of Informatics & Computing, Indianapolis, IN 46202, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kefei Liu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sungeun Kim
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Electrical and Computer Engineering, SUNY Oswego, NY 13126, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Casey S Greene
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason H Moore
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Li Shen
- Department of BioHealth Informatics, Indiana University School of Informatics & Computing, Indianapolis, IN 46202, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
21
|
Toledo JB, Arnold M, Kastenmüller G, Chang R, Baillie RA, Han X, Thambisetty M, Tenenbaum JD, Suhre K, Thompson JW, John-Williams LS, MahmoudianDehkordi S, Rotroff DM, Jack JR, Motsinger-Reif A, Risacher SL, Blach C, Lucas JE, Massaro T, Louie G, Zhu H, Dallmann G, Klavins K, Koal T, Kim S, Nho K, Shen L, Casanova R, Varma S, Legido-Quigley C, Moseley MA, Zhu K, Henrion MYR, van der Lee SJ, Harms AC, Demirkan A, Hankemeier T, van Duijn CM, Trojanowski JQ, Shaw LM, Saykin AJ, Weiner MW, Doraiswamy PM, Kaddurah-Daouk R. Metabolic network failures in Alzheimer's disease: A biochemical road map. Alzheimers Dement 2017; 13:965-984. [PMID: 28341160 PMCID: PMC5866045 DOI: 10.1016/j.jalz.2017.01.020] [Citation(s) in RCA: 324] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/25/2017] [Accepted: 01/26/2017] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The Alzheimer's Disease Research Summits of 2012 and 2015 incorporated experts from academia, industry, and nonprofit organizations to develop new research directions to transform our understanding of Alzheimer's disease (AD) and propel the development of critically needed therapies. In response to their recommendations, big data at multiple levels are being generated and integrated to study network failures in disease. We used metabolomics as a global biochemical approach to identify peripheral metabolic changes in AD patients and correlate them to cerebrospinal fluid pathology markers, imaging features, and cognitive performance. METHODS Fasting serum samples from the Alzheimer's Disease Neuroimaging Initiative (199 control, 356 mild cognitive impairment, and 175 AD participants) were analyzed using the AbsoluteIDQ-p180 kit. Performance was validated in blinded replicates, and values were medication adjusted. RESULTS Multivariable-adjusted analyses showed that sphingomyelins and ether-containing phosphatidylcholines were altered in preclinical biomarker-defined AD stages, whereas acylcarnitines and several amines, including the branched-chain amino acid valine and α-aminoadipic acid, changed in symptomatic stages. Several of the analytes showed consistent associations in the Rotterdam, Erasmus Rucphen Family, and Indiana Memory and Aging Studies. Partial correlation networks constructed for Aβ1-42, tau, imaging, and cognitive changes provided initial biochemical insights for disease-related processes. Coexpression networks interconnected key metabolic effectors of disease. DISCUSSION Metabolomics identified key disease-related metabolic changes and disease-progression-related changes. Defining metabolic changes during AD disease trajectory and its relationship to clinical phenotypes provides a powerful roadmap for drug and biomarker discovery.
Collapse
Affiliation(s)
- Jon B Toledo
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Neurology, Houston Methodist Hospital, Houston, TX, USA.
| | - Matthias Arnold
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Rui Chang
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Xianlin Han
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Unit, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jessica D Tenenbaum
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Karsten Suhre
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Department of Physiology and Biophysics, Weill Cornell Medical College, Qatar, Doha, Qatar
| | - J Will Thompson
- Duke Proteomics and Metabolomics Shared Resource, Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Lisa St John-Williams
- Duke Proteomics and Metabolomics Shared Resource, Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Siamak MahmoudianDehkordi
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Daniel M Rotroff
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - John R Jack
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Alison Motsinger-Reif
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; The Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Colette Blach
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA; Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Joseph E Lucas
- Institute for Genome Sciences and Policy, Duke University, Durham, NC, USA
| | - Tyler Massaro
- Institute for Genome Sciences and Policy, Duke University, Durham, NC, USA
| | - Gregory Louie
- Department of Psychiatry, Duke University, Durham, NC, USA; Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Hongjie Zhu
- Department of Psychiatry, Duke University, Durham, NC, USA; Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | | | | | | | - Sungeun Kim
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; The Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; The Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Li Shen
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; The Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ramon Casanova
- Clinical and Translational Neuroscience Unit, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Sudhir Varma
- Clinical and Translational Neuroscience Unit, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | | | - M Arthur Moseley
- Duke Proteomics and Metabolomics Shared Resource, Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Kuixi Zhu
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marc Y R Henrion
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Amy C Harms
- Analytical Biosciences, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Ayse Demirkan
- Department of Epidemiology, ErasmusMC, Rotterdam, The Netherlands
| | - Thomas Hankemeier
- Department of Epidemiology, ErasmusMC, Rotterdam, The Netherlands; Analytical Biosciences, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, ErasmusMC, Rotterdam, The Netherlands; Analytical Biosciences, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - John Q Trojanowski
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leslie M Shaw
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; The Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael W Weiner
- Department of Radiology, Center for Imaging of Neurodegenerative Diseases, San Francisco VA Medical Center/University of California San Francisco, San Francisco, CA, USA
| | - P Murali Doraiswamy
- Department of Psychiatry, Duke University, Durham, NC, USA; Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry, Duke University, Durham, NC, USA; Duke Institute for Brain Sciences, Duke University, Durham, NC, USA; Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
22
|
Du L, Zhang T, Liu K, Yan J, Yao X, Risacher SL, Saykin AJ, Han J, Guo L, Shen L. Identifying Associations Between Brain Imaging Phenotypes and Genetic Factors via A Novel Structured SCCA Approach. INFORMATION PROCESSING IN MEDICAL IMAGING : PROCEEDINGS OF THE ... CONFERENCE 2017; 10265:543-555. [PMID: 28867917 PMCID: PMC5576511 DOI: 10.1007/978-3-319-59050-9_43] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Brain imaging genetics attracts more and more attention since it can reveal associations between genetic factors and the structures or functions of human brain. Sparse canonical correlation analysis (SCCA) is a powerful bi-multivariate association identification technique in imaging genetics. There have been many SCCA methods which could capture different types of structured imaging genetic relationships. These methods either use the group lasso to recover the group structure, or employ the graph/network guided fused lasso to find out the network structure. However, the group lasso methods have limitation in generalization because of the incomplete or unavailable prior knowledge in real world. The graph/network guided methods are sensitive to the sign of the sample correlation which may be incorrectly estimated. We introduce a new SCCA model using a novel graph guided pairwise group lasso penalty, and propose an efficient optimization algorithm. The proposed method has a strong upper bound for the grouping effect for both positively and negatively correlated variables. We show that our method performs better than or equally to two state-of-the-art SCCA methods on both synthetic and real neuroimaging genetics data. In particular, our method identifies stronger canonical correlations and captures better canonical loading profiles, showing its promise for revealing biologically meaningful imaging genetic associations.
Collapse
Affiliation(s)
- Lei Du
- School of Automation, Northwestern Polytechnical University, Xi'an China
| | - Tuo Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an China
| | - Kefei Liu
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Jingwen Yan
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Xiaohui Yao
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Shannon L Risacher
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Andrew J Saykin
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Junwei Han
- School of Automation, Northwestern Polytechnical University, Xi'an China
| | - Lei Guo
- School of Automation, Northwestern Polytechnical University, Xi'an China
| | - Li Shen
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| |
Collapse
|
23
|
Hao X, Li C, Du L, Yao X, Yan J, Risacher SL, Saykin AJ, Shen L, Zhang D. Mining Outcome-relevant Brain Imaging Genetic Associations via Three-way Sparse Canonical Correlation Analysis in Alzheimer's Disease. Sci Rep 2017; 7:44272. [PMID: 28291242 PMCID: PMC5349597 DOI: 10.1038/srep44272] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 02/07/2017] [Indexed: 11/24/2022] Open
Abstract
Neuroimaging genetics is an emerging field that aims to identify the associations between genetic variants (e.g., single nucleotide polymorphisms (SNPs)) and quantitative traits (QTs) such as brain imaging phenotypes. In recent studies, in order to detect complex multi-SNP-multi-QT associations, bi-multivariate techniques such as various structured sparse canonical correlation analysis (SCCA) algorithms have been proposed and used in imaging genetics studies. However, associations between genetic markers and imaging QTs identified by existing bi-multivariate methods may not be all disease specific. To bridge this gap, we propose an analytical framework, based on three-way sparse canonical correlation analysis (T-SCCA), to explore the intrinsic associations among genetic markers, imaging QTs, and clinical scores of interest. We perform an empirical study using the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort to discover the relationships among SNPs from AD risk gene APOE, imaging QTs extracted from structural magnetic resonance imaging scans, and cognitive and diagnostic outcomes. The proposed T-SCCA model not only outperforms the traditional SCCA method in terms of identifying strong associations, but also discovers robust outcome-relevant imaging genetic patterns, demonstrating its promise for improving disease-related mechanistic understanding.
Collapse
Affiliation(s)
- Xiaoke Hao
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Chanxiu Li
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Lei Du
- School of Automation, Northwestern Polytechnical University, Xi’an 710072, China
| | - Xiaohui Yao
- Department of Radiology and Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Jingwen Yan
- Department of Radiology and Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- School of Informatics and Computing, Indiana University, Indianapolis, IN 46202, USA
| | - Shannon L. Risacher
- Department of Radiology and Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Li Shen
- Department of Radiology and Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Daoqiang Zhang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| |
Collapse
|
24
|
Connecting genetic risk to disease end points through the human blood plasma proteome. Nat Commun 2017; 8:14357. [PMID: 28240269 PMCID: PMC5333359 DOI: 10.1038/ncomms14357] [Citation(s) in RCA: 424] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/16/2016] [Indexed: 12/29/2022] Open
Abstract
Genome-wide association studies (GWAS) with intermediate phenotypes, like changes in metabolite and protein levels, provide functional evidence to map disease associations and translate them into clinical applications. However, although hundreds of genetic variants have been associated with complex disorders, the underlying molecular pathways often remain elusive. Associations with intermediate traits are key in establishing functional links between GWAS-identified risk-variants and disease end points. Here we describe a GWAS using a highly multiplexed aptamer-based affinity proteomics platform. We quantify 539 associations between protein levels and gene variants (pQTLs) in a German cohort and replicate over half of them in an Arab and Asian cohort. Fifty-five of the replicated pQTLs are located in trans. Our associations overlap with 57 genetic risk loci for 42 unique disease end points. We integrate this information into a genome-proteome network and provide an interactive web-tool for interrogations. Our results provide a basis for novel approaches to pharmaceutical and diagnostic applications. Individual genetic variation can affect the levels of protein in blood, but detailed data sets linking these two types of data are rare. Here, the authors carry out a genome-wide association study of levels of over a thousand different proteins, and describe many new SNP-protein interactions.
Collapse
|
25
|
Du L, Zhang T, Liu K, Yao X, Yan J, Risacher SL, Guo L, Saykin AJ, Shen L. Sparse Canonical Correlation Analysis via Truncated ℓ1-norm with Application to Brain Imaging Genetics. PROCEEDINGS. IEEE INTERNATIONAL CONFERENCE ON BIOINFORMATICS AND BIOMEDICINE 2017; 2016:707-711. [PMID: 28989812 DOI: 10.1109/bibm.2016.7822605] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Discovering bi-multivariate associations between genetic markers and neuroimaging quantitative traits is a major task in brain imaging genetics. Sparse Canonical Correlation Analysis (SCCA) is a popular technique in this area for its powerful capability in identifying bi-multivariate relationships coupled with feature selection. The existing SCCA methods impose either the ℓ1-norm or its variants. The ℓ0-norm is more desirable, which however remains unexplored since the ℓ0-norm minimization is NP-hard. In this paper, we impose the truncated ℓ1-norm to improve the performance of the ℓ1-norm based SCCA methods. Besides, we propose two efficient optimization algorithms and prove their convergence. The experimental results, compared with two benchmark methods, show that our method identifies better and meaningful canonical loading patterns in both simulated and real imaging genetic analyse.
Collapse
Affiliation(s)
- Lei Du
- School of Automation, Northwestern Polytechnical University, Xi'an, China 710072
| | - Tuo Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an, China 710072
| | - Kefei Liu
- Indiana University School of Medicine, Indianapolis, USA 46202
| | - Xiaohui Yao
- Indiana University School of Medicine, Indianapolis, USA 46202
| | - Jingwen Yan
- Indiana University School of Medicine, Indianapolis, USA 46202
| | | | - Lei Guo
- School of Automation, Northwestern Polytechnical University, Xi'an, China 710072
| | - Andrew J Saykin
- Indiana University School of Medicine, Indianapolis, USA 46202
| | - Li Shen
- Indiana University School of Medicine, Indianapolis, USA 46202
| | | |
Collapse
|
26
|
Ahola-Olli AV, Würtz P, Havulinna AS, Aalto K, Pitkänen N, Lehtimäki T, Kähönen M, Lyytikäinen LP, Raitoharju E, Seppälä I, Sarin AP, Ripatti S, Palotie A, Perola M, Viikari JS, Jalkanen S, Maksimow M, Salomaa V, Salmi M, Kettunen J, Raitakari OT. Genome-wide Association Study Identifies 27 Loci Influencing Concentrations of Circulating Cytokines and Growth Factors. Am J Hum Genet 2017; 100:40-50. [PMID: 27989323 DOI: 10.1016/j.ajhg.2016.11.007] [Citation(s) in RCA: 408] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 11/11/2016] [Indexed: 12/14/2022] Open
Abstract
Circulating cytokines and growth factors are regulators of inflammation and have been implicated in autoimmune and metabolic diseases. In this genome-wide association study (GWAS) of up to 8,293 Finns we identified 27 genome-widely significant loci (p < 1.2 × 10-9) for one or more cytokines. Fifteen of the associated variants had expression quantitative trait loci in whole blood. We provide genetic instruments to clarify the causal roles of cytokine signaling and upstream inflammation in immune-related and other chronic diseases. We further link inflammatory markers with variants previously associated with autoimmune diseases such as Crohn disease, multiple sclerosis, and ulcerative colitis and hereby elucidate the molecular mechanisms underpinning these diseases and suggest potential drug targets.
Collapse
|
27
|
Robinson RAS, Amin B, Guest PC. Multiplexing Biomarker Methods, Proteomics and Considerations for Alzheimer’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 974:21-48. [DOI: 10.1007/978-3-319-52479-5_2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
28
|
Kim S, Nho K, Ramanan VK, Lai D, Foroud TM, Lane K, Murrell JR, Gao S, Hall KS, Unverzagt FW, Baiyewu O, Ogunniyi A, Gureje O, Kling MA, Doraiswamy PM, Kaddurah-Daouk R, Hendrie HC, Saykin AJ. Genetic Influences on Plasma Homocysteine Levels in African Americans and Yoruba Nigerians. J Alzheimers Dis 2016; 49:991-1003. [PMID: 26519441 DOI: 10.3233/jad-150651] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Plasma homocysteine, a metabolite involved in key cellular methylation processes seems to be implicated in cognitive functions and cardiovascular health with its high levels representing a potential modifiable risk factor for Alzheimer's disease (AD) and other dementias. A better understanding of the genetic factors regulating homocysteine levels, particularly in non-white populations, may help in risk stratification analyses of existing clinical trials and may point to novel targets for homocysteine-lowering therapy. To identify genetic influences on plasma homocysteine levels in individuals with African ancestry, we performed a targeted gene and pathway-based analysis using a priori biological information and then to identify new association performed a genome-wide association study. All analyses used combined data from the African American and Yoruba cohorts from the Indianapolis-Ibadan Dementia Project. Targeted analyses demonstrated significant associations of homocysteine and variants within the CBS (Cystathionine beta-Synthase) gene. We identified a novel genome-wide significant association of the AD risk gene CD2AP (CD2-associated protein) with plasma homocysteine levels in both cohorts. Minor allele (T) carriers of identified CD2AP variant (rs6940729) exhibited decreased homocysteine level. Pathway enrichment analysis identified several interesting pathways including the GABA receptor activation pathway. This is noteworthy given the known antagonistic effect of homocysteine on GABA receptors. These findings identify several new targets warranting further investigation in relation to the role of homocysteine in neurodegeneration.
Collapse
Affiliation(s)
- Sungeun Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana University Network Science Institute, Bloomington, IN, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana University Network Science Institute, Bloomington, IN, USA
| | - Vijay K Ramanan
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Internal Medicine, Preliminary Medicine Residency, St. Vincent Indianapolis, Indianapolis, IN, USA
| | - Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tatiana M Foroud
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana University Network Science Institute, Bloomington, IN, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Katie Lane
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jill R Murrell
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sujuan Gao
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kathleen S Hall
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Frederick W Unverzagt
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Olusegun Baiyewu
- Department of Psychiatry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adesola Ogunniyi
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oye Gureje
- Department of Psychiatry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Mitchel A Kling
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Behavioral Health Service, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - P Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA.,Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA.,Duke Institute for Brain Sciences, Duke University, Durham, NC, USA.,Pharmacometabolomics Center, Duke University, Durham, NC, USA
| | - Hugh C Hendrie
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana University Center for Aging Research, Indianapolis, IN, USA.,Regenstrief Institute Inc., Indianapolis, IN, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana University Network Science Institute, Bloomington, IN, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
29
|
Solomon T, Smith EN, Matsui H, Braekkan SK, Wilsgaard T, Njølstad I, Mathiesen EB, Hansen JB, Frazer KA. Associations Between Common and Rare Exonic Genetic Variants and Serum Levels of 20 Cardiovascular-Related Proteins: The Tromsø Study. ACTA ACUST UNITED AC 2016; 9:375-83. [PMID: 27329291 PMCID: PMC4982757 DOI: 10.1161/circgenetics.115.001327] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/16/2016] [Indexed: 01/09/2023]
Abstract
Supplemental Digital Content is available in the text. Background— Genetic variation can be used to study causal relationships between biomarkers and diseases. Here, we identify new common and rare genetic variants associated with cardiovascular-related protein levels (protein quantitative trait loci [pQTLs]). We functionally annotate these pQTLs, predict and experimentally confirm a novel molecular interaction, and determine which pQTLs are associated with diseases and physiological phenotypes. Methods and Results— As part of a larger case–control study of venous thromboembolism, serum levels of 51 proteins implicated in cardiovascular diseases were measured in 330 individuals from the Tromsø Study. Exonic genetic variation near each protein’s respective gene (cis) was identified using sequencing and arrays. Using single site and gene-based tests, we identified 27 genetic associations between pQTLs and the serum levels of 20 proteins: 14 associated with common variation in cis, of which 6 are novel (ie, not previously reported); 7 associations with rare variants in cis, of which 4 are novel; and 6 associations in trans. Of the 20 proteins, 15 were associated with single sites and 7 with rare variants. cis-pQTLs for kallikrein and F12 also show trans associations for proteins (uPAR, kininogen) known to be cleaved by kallikrein and with NTproBNP. We experimentally demonstrate that kallikrein can cleave proBNP (NTproBNP precursor) in vitro. Nine of the pQTLs have previously identified associations with 17 disease and physiological phenotypes. Conclusions— We have identified cis and trans genetic variation associated with the serum levels of 20 proteins and utilized these pQTLs to study molecular mechanisms underlying disease and physiological phenotypes.
Collapse
Affiliation(s)
- Terry Solomon
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Erin N Smith
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Hiroko Matsui
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Sigrid K Braekkan
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | | | - Tom Wilsgaard
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Inger Njølstad
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Ellisiv B Mathiesen
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - John-Bjarne Hansen
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.)
| | - Kelly A Frazer
- From the Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla (T.S.), Department of Pediatrics, Rady's Children's Hospital, San Diego, La Jolla, CA (E.N.S., H.M., K.A.F.); Institute for Genomic Medicine, University of California, San Diego, La Jolla (K.A.F.); Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Centre (TREC) (E.N.S., S.K.B., I.N., E.B.M., J.-B.H., K.A.F.), Department of Community Medicine (T.W., I.N.), and Brain and Circulation Research Group, Department of Clinical Medicine (E.B.M.), UiT The Arctic University of Norway; and Division of Internal Medicine, University Hospital of North Norway, Tromsø (S.K.B., J.-B.H.).
| |
Collapse
|
30
|
Two-dimensional enrichment analysis for mining high-level imaging genetic associations. Brain Inform 2016; 4:27-37. [PMID: 27747820 PMCID: PMC5118198 DOI: 10.1007/s40708-016-0052-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/29/2016] [Indexed: 11/05/2022] Open
Abstract
Enrichment analysis has been widely applied in the genome-wide association studies, where gene sets corresponding to biological pathways are examined for significant associations with a phenotype to help increase statistical power and improve biological interpretation. In this work, we expand the scope of enrichment analysis into brain imaging genetics, an emerging field that studies how genetic variation influences brain structure and function measured by neuroimaging quantitative traits (QT). Given the high dimensionality of both imaging and genetic data, we propose to study Imaging Genetic Enrichment Analysis (IGEA), a new enrichment analysis paradigm that jointly considers meaningful gene sets (GS) and brain circuits (BC) and examines whether any given GS–BC pair is enriched in a list of gene–QT findings. Using gene expression data from Allen Human Brain Atlas and imaging genetics data from Alzheimer’s Disease Neuroimaging Initiative as test beds, we present an IGEA framework and conduct a proof-of-concept study. This empirical study identifies 25 significant high-level two-dimensional imaging genetics modules. Many of these modules are relevant to a variety of neurobiological pathways or neurodegenerative diseases, showing the promise of the proposal framework for providing insight into the mechanism of complex diseases.
Collapse
|
31
|
The Alzheimer's Disease Neuroimaging Initiative 2 Biomarker Core: A review of progress and plans. Alzheimers Dement 2016; 11:772-91. [PMID: 26194312 DOI: 10.1016/j.jalz.2015.05.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 05/04/2015] [Accepted: 05/05/2015] [Indexed: 11/20/2022]
Abstract
INTRODUCTION We describe Alzheimer's Disease Neuroimaging Initiative (ADNI) Biomarker Core progress including: the Biobank; cerebrospinal fluid (CSF) amyloid beta (Aβ1-42), t-tau, and p-tau181 analytical performance, definition of Alzheimer's disease (AD) profile for plaque, and tangle burden detection and increased risk for progression to AD; AD disease heterogeneity; progress in standardization; and new studies using ADNI biofluids. METHODS Review publications authored or coauthored by ADNI Biomarker core faculty and selected non-ADNI studies to deepen the understanding and interpretation of CSF Aβ1-42, t-tau, and p-tau181 data. RESULTS CSF AD biomarker measurements with the qualified AlzBio3 immunoassay detects neuropathologic AD hallmarks in preclinical and prodromal disease stages, based on CSF studies in non-ADNI living subjects followed by the autopsy confirmation of AD. Collaboration across ADNI cores generated the temporal ordering model of AD biomarkers varying across individuals because of genetic/environmental factors that increase/decrease resilience to AD pathologies. DISCUSSION Further studies will refine this model and enable the use of biomarkers studied in ADNI clinically and in disease-modifying therapeutic trials.
Collapse
|
32
|
Song A, Yan J, Kim S, Risacher SL, Wong AK, Saykin AJ, Shen L, Greene CS. Network-based analysis of genetic variants associated with hippocampal volume in Alzheimer's disease: a study of ADNI cohorts. BioData Min 2016; 9:3. [PMID: 26788126 PMCID: PMC4717572 DOI: 10.1186/s13040-016-0082-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 01/14/2016] [Indexed: 12/25/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a neurodegenerative disease that causes dementia. While molecular basis of AD is not fully understood, genetic factors are expected to participate in the development and progression of the disease. Our goal was to uncover novel genetic underpinnings of Alzheimer’s disease with a bioinformatics approach that accounts for tissue specificity. Findings We performed genome-wide association studies (GWAS) for hippocampal volume in two Alzheimer’s Disease Neuroimaging Initiative (ADNI) cohorts. We used these GWAS in a subsequent tissue-specific network-wide association study (NetWAS), which applied nominally significant associations in the initial GWAS to identify disease relevant patterns in a functional network for the hippocampus. We compared prioritized gene lists from NetWAS and GWAS with literature curated AD-associated genes from the Online Mendelian Inheritance in Man (OMIM) database. In the ADNI-1 GWAS, where we also observed an enrichment of low p-values, NetWAS prioritized disease-gene associations in accordance with OMIM annotations. This was not observed in the ADNI-2 dataset. We provide source code to replicate these analyses as well as complete results under permissive licenses. Conclusions We performed the first analysis of hippocampal volume using NetWAS, which uses machine learning algorithms applied to tissue-specific functional interaction network to prioritize GWAS results. Our findings support the idea that tissue-specific networks may provide helpful context for understanding the etiology of common human diseases and reveal challenges that network-based approaches encounter in some datasets. Our source code and intermediate results files can facilitate the development of methods to address these challenges. Electronic supplementary material The online version of this article (doi:10.1186/s13040-016-0082-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ailin Song
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire USA ; Dartmouth-Hitchcock Norris Cotton Cancer Center, Lebanon, New Hampshire USA ; Institute for Quantitative Biomedical Sciences, Dartmouth College, Hanover, New Hampshire USA
| | - Jingwen Yan
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana USA ; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana USA ; School of Informatics and Computing, Indiana University Indianapolis, Indianapolis, Indiana USA
| | - Sungeun Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana USA ; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, Indiana USA
| | - Shannon Leigh Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, Indiana USA
| | - Aaron K Wong
- Simons Center for Data Analysis, Simons Foundation, New York, NY USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, Indiana USA
| | - Li Shen
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana USA ; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana USA ; School of Informatics and Computing, Indiana University Indianapolis, Indianapolis, Indiana USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, Indiana USA
| | - Casey S Greene
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire USA ; Dartmouth-Hitchcock Norris Cotton Cancer Center, Lebanon, New Hampshire USA ; Institute for Quantitative Biomedical Sciences, Dartmouth College, Hanover, New Hampshire USA ; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvnia USA
| | | |
Collapse
|
33
|
Deming Y, Xia J, Cai Y, Lord J, Del-Aguila JL, Fernandez MV, Carrell D, Black K, Budde J, Ma S, Saef B, Howells B, Bertelsen S, Bailey M, Ridge PG, Holtzman D, Morris JC, Bales K, Pickering EH, Lee JM, Heitsch L, Kauwe J, Goate A, Piccio L, Cruchaga C. Genetic studies of plasma analytes identify novel potential biomarkers for several complex traits. Sci Rep 2016; 6:18092. [PMID: 36647296 PMCID: PMC4698720 DOI: 10.1038/srep18092] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/11/2015] [Indexed: 01/23/2023] Open
Abstract
Genome-wide association studies of 146 plasma protein levels in 818 individuals revealed 56 genome-wide significant associations (28 novel) with 47 analytes. Loci associated with plasma levels of 39 proteins tested have been previously associated with various complex traits such as heart disease, inflammatory bowel disease, Type 2 diabetes and multiple sclerosis. These data suggest that these plasma protein levels may constitute informative endophenotypes for these complex traits. We found three potential pleiotropic genes: ABO for plasma SELE and ACE levels, FUT2 for CA19-9 and CEA plasma levels and APOE for ApoE and CRP levels. We also found multiple independent signals in loci associated with plasma levels of ApoH, CA19-9, FetuinA, IL6r and LPa. Our study highlights the power of biological traits for genetic studies to identify genetic variants influencing clinically relevant traits, potential pleiotropic effects and complex disease associations in the same locus.
Collapse
Affiliation(s)
- Yuetiva Deming
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - Jian Xia
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yefei Cai
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - Jenny Lord
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
- Human Genetics Programme, Wellcome Trust Sanger Institute, Cambridge, CB10 1SA, UK
| | - Jorge L. Del-Aguila
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - Maria Victoria Fernandez
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - David Carrell
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - Kathleen Black
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - John Budde
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - ShengMei Ma
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - Benjamin Saef
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - Bill Howells
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - Sarah Bertelsen
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - Matthew Bailey
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Perry G. Ridge
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - David Holtzman
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, 4488 Forest Park Ave., St Louis, MO 63108, USA
- Hope Center for Neurological Disorders. Washington University School of Medicine, 660 S. Euclid Ave. B8111, St. Louis, MO 63110, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, 4488 Forest Park Ave., St Louis, MO 63108, USA
- Hope Center for Neurological Disorders. Washington University School of Medicine, 660 S. Euclid Ave. B8111, St. Louis, MO 63110, 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
| | - Jin-Moo Lee
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Laura Heitsch
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - John Kauwe
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Alison Goate
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, 4488 Forest Park Ave., St Louis, MO 63108, USA
- Hope Center for Neurological Disorders. Washington University School of Medicine, 660 S. Euclid Ave. B8111, St. Louis, MO 63110, USA
| | - Laura Piccio
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders. Washington University School of Medicine, 660 S. Euclid Ave. B8111, St. Louis, MO 63110, USA
| |
Collapse
|
34
|
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. 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
|
35
|
Ramanan VK, Nho K, Shen L, Risacher SL, Kim S, McDonald BC, Farlow MR, Foroud TM, Gao S, Soininen H, Kłoszewska I, Mecocci P, Tsolaki M, Vellas B, Lovestone S, Aisen PS, Petersen RC, Jack CR, Shaw LM, Trojanowski JQ, Weiner MW, Green RC, Toga AW, De Jager PL, Yu L, Bennett DA, Saykin AJ. FASTKD2 is associated with memory and hippocampal structure in older adults. Mol Psychiatry 2015; 20:1197-204. [PMID: 25385369 PMCID: PMC4427556 DOI: 10.1038/mp.2014.142] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 09/05/2014] [Accepted: 09/10/2014] [Indexed: 12/15/2022]
Abstract
Memory impairment is the cardinal early feature of Alzheimer's disease, a highly prevalent disorder whose causes remain only partially understood. To identify novel genetic predictors, we used an integrative genomics approach to perform the largest study to date of human memory (n=14 781). Using a genome-wide screen, we discovered a novel association of a polymorphism in the pro-apoptotic gene FASTKD2 (fas-activated serine/threonine kinase domains 2; rs7594645-G) with better memory performance and replicated this finding in independent samples. Consistent with a neuroprotective effect, rs7594645-G carriers exhibited increased hippocampal volume and gray matter density and decreased cerebrospinal fluid levels of apoptotic mediators. The MTOR (mechanistic target of rapamycin) gene and pathways related to endocytosis, cholinergic neurotransmission, epidermal growth factor receptor signaling and immune regulation, among others, also displayed association with memory. These findings nominate FASTKD2 as a target for modulating neurodegeneration and suggest potential mechanisms for therapies to combat memory loss in normal cognitive aging and dementia.
Collapse
Affiliation(s)
- Vijay K Ramanan
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA,Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Li Shen
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shannon L. Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sungeun Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brenna C. McDonald
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA,Indiana Alzheimer's Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Martin R. Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA,Indiana Alzheimer's Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tatiana M. Foroud
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA,Indiana Alzheimer's Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sujuan Gao
- Indiana Alzheimer's Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hilkka Soininen
- On behalf of the AddNeuroMed Consortium,Department of Neurology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Iwona Kłoszewska
- On behalf of the AddNeuroMed Consortium,Medical University of Lodz, Lodz, Poland
| | - Patrizia Mecocci
- On behalf of the AddNeuroMed Consortium,Institute of Gerontology and Geriatrics, University of Perugia, Perugia, Italy
| | - Magda Tsolaki
- On behalf of the AddNeuroMed Consortium,3rd Department of Neurology, Aristotle University, Thessaloniki, Greece
| | - Bruno Vellas
- On behalf of the AddNeuroMed Consortium,INSERM U 558, University of Toulouse, Toulouse, France
| | - Simon Lovestone
- On behalf of the AddNeuroMed Consortium,University of Oxford, Department of Psychiatry, Oxford, UK
| | - Paul S. Aisen
- Department of Neuroscience, University of California-San Diego, San Diego, CA, USA
| | | | - Clifford R. Jack
- Department of Radiology, Mayo Clinic Minnesota, Rochester, MN, USA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA,Institute on Aging, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA,Institute on Aging, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Michael W. Weiner
- Departments of Radiology, Medicine, and Psychiatry, University of California-San Francisco, San Francisco, CA, USA,Department of Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Robert C. Green
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Philip L. De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, Boston, MA, USA,Harvard Medical School, Boston, MA, USA,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Andrew J. Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA,Indiana Alzheimer's Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA,Correspondence to: Dr. Andrew J. Saykin, IU Health Neuroscience Center, Suite 4100 Indiana University School of Medicine 355 West 16th Street, Indianapolis, IN 46202, USA , Phone (317)963-7501, Fax (317)963-7547
| | | |
Collapse
|
36
|
Yao X, Yan J, Kim S, Nho K, Risacher SL, Inlow M, Moore JH, Saykin AJ, Shen L. Two-dimensional Enrichment Analysis for Mining High-level Imaging Genetic Associations. BRAIN INFORMATICS AND HEALTH : 8TH INTERNATIONAL CONFERENCE, BIH 2015, LONDON, UK, AUGUST 30-SEPTEMBER 2, 2015 : PROCEEDINGS. BIH (CONFERENCE) (8TH : 2015 : LONDON, ENGLAND) 2015; 9250:115-124. [PMID: 26568986 PMCID: PMC4640356 DOI: 10.1007/978-3-319-23344-4_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Enrichment analysis has been widely applied in the genome-wide association studies (GWAS), where gene sets corresponding to biological pathways are examined for significant associations with a phenotype to help increase statistical power and improve biological interpretation. In this work, we expand the scope of enrichment analysis into brain imaging genetics, an emerging field that studies how genetic variation influences brain structure and function measured by neuroimaging quantitative traits (QT). Given the high dimensionality of both imaging and genetic data, we propose to study Imaging Genetic Enrichment Analysis (IGEA), a new enrichment analysis paradigm that jointly considers meaningful gene sets (GS) and brain circuits (BC) and examines whether any given GS-BC pair is enriched in a list of gene-QT findings. Using gene expression data from Allen Human Brain Atlas and imaging genetics data from Alzheimer's Disease Neuroimaging Initiative as test beds, we present an IGEA framework and conduct a proof-of-concept study. This empirical study identifies 12 significant high level two dimensional imaging genetics modules. Many of these modules are relevant to a variety of neurobiological pathways or neurodegenerative diseases, showing the promise of the proposal framework for providing insight into the mechanism of complex diseases.
Collapse
Affiliation(s)
- Xiaohui Yao
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
- School of Informatics and Computing, Indiana University Indianapolis, IN, USA
| | - Jingwen Yan
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
- School of Informatics and Computing, Indiana University Indianapolis, IN, USA
| | - Sungeun Kim
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Kwangsik Nho
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Shannon L Risacher
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Mark Inlow
- Mathematics, Rose-Hulman Institute of Technology, IN, USA
| | - Jason H. Moore
- Biomedical Informatics, School of Medicine, University of Pennsylvania, PA, USA
| | - Andrew J. Saykin
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
| | - Li Shen
- Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
- School of Informatics and Computing, Indiana University Indianapolis, IN, USA
| | | |
Collapse
|
37
|
Hendrix JA, Finger B, Weiner MW, Frisoni GB, Iwatsubo T, Rowe CC, Kim SY, Guinjoan SM, Sevlever G, Carrillo MC. The Worldwide Alzheimer's Disease Neuroimaging Initiative: An update. Alzheimers Dement 2015; 11:850-9. [DOI: 10.1016/j.jalz.2015.05.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 01/06/2023]
Affiliation(s)
- James A. Hendrix
- Medical & Scientific Relations; Alzheimer's Association; Chicago IL USA
| | | | - Michael W. Weiner
- Center for Imaging of Neurodegenerative Diseases (CIND), Northern, California Institute of Research; San Francisco VA Medical Center; San Francisco CA USA
- Department of Radiology; University of California; San Francisco CA USA
| | - Giovanni B. Frisoni
- Laboratory of Neuroimaging of Aging; University Hospitals and University of Geneva; Geneva Switzerland
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine; The University Hospital of Tokyo; Japan
| | | | - Seong Yoon Kim
- Department of Psychiatry; Asian Medical Center; Seoul Republic of Korea
| | - Salvador M. Guinjoan
- Aging and Memory Center; Instituto de Investigaciones Neurologicas Raul Carrea (FLENI); Buenos Aires Argentina
| | - Gustavo Sevlever
- Aging and Memory Center; Instituto de Investigaciones Neurologicas Raul Carrea (FLENI); Buenos Aires Argentina
| | - Maria C. Carrillo
- Medical & Scientific Relations; Alzheimer's Association; Chicago IL USA
| |
Collapse
|
38
|
Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Cedarbaum J, Donohue MC, Green RC, Harvey D, Jack CR, Jagust W, Morris JC, Petersen RC, Saykin AJ, Shaw L, Thompson PM, Toga AW, Trojanowski JQ. Impact of the Alzheimer's Disease Neuroimaging Initiative, 2004 to 2014. Alzheimers Dement 2015; 11:865-84. [PMID: 26194320 PMCID: PMC4659407 DOI: 10.1016/j.jalz.2015.04.005] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/04/2015] [Accepted: 04/23/2015] [Indexed: 01/18/2023]
Abstract
INTRODUCTION The Alzheimer's Disease Neuroimaging Initiative (ADNI) was established in 2004 to facilitate the development of effective treatments for Alzheimer's disease (AD) by validating biomarkers for AD clinical trials. METHODS We searched for ADNI publications using established methods. RESULTS ADNI has (1) developed standardized biomarkers for use in clinical trial subject selection and as surrogate outcome measures; (2) standardized protocols for use across multiple centers; (3) initiated worldwide ADNI; (4) inspired initiatives investigating traumatic brain injury and post-traumatic stress disorder in military populations, and depression, respectively, as an AD risk factor; (5) acted as a data-sharing model; (6) generated data used in over 600 publications, leading to the identification of novel AD risk alleles, and an understanding of the relationship between biomarkers and AD progression; and (7) inspired other public-private partnerships developing biomarkers for Parkinson's disease and multiple sclerosis. DISCUSSION ADNI has made myriad impacts in its first decade. A competitive renewal of the project in 2015 would see the use of newly developed tau imaging ligands, and the continued development of recruitment strategies and outcome measures for clinical trials.
Collapse
Affiliation(s)
- Michael W Weiner
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA; Department of Radiology, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Dallas P Veitch
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA
| | - Paul S Aisen
- Department of Neurosciences, University of California- San Diego, La Jolla, CA, USA
| | - Laurel A Beckett
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, Davis, CA, USA
| | - Nigel J Cairns
- Department of Neurology, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jesse Cedarbaum
- Neurology Early Clinical Development, Biogen Idec, Cambridge, MA, USA
| | - Michael C Donohue
- Division of Biostatistics and Bioinformatics, Department of Family Medicine and Public Health, University of California, San Diego, San Diego, CA, USA
| | - Robert C Green
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Danielle Harvey
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, Davis, CA, USA
| | | | - William Jagust
- Helen Wills Neuroscience Institute and the School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | | | - Andrew J Saykin
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leslie Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Institute for Neuroimaging and Informatics, University of Southern California, Marina Del Rey, CA, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, Institute of Neuroimaging and Informatics, Keck School of Medicine of University of Southern California Los Angeles, CA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Alzheimer's Disease Core Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Udall Parkinson's Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
39
|
Saykin AJ, Shen L, Yao X, Kim S, Nho K, Risacher SL, Ramanan VK, Foroud TM, Faber KM, Sarwar N, Munsie LM, Hu X, Soares HD, Potkin SG, Thompson PM, Kauwe JSK, Kaddurah-Daouk R, Green RC, Toga AW, Weiner MW. Genetic studies of quantitative MCI and AD phenotypes in ADNI: Progress, opportunities, and plans. Alzheimers Dement 2015; 11:792-814. [PMID: 26194313 PMCID: PMC4510473 DOI: 10.1016/j.jalz.2015.05.009] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 05/08/2015] [Accepted: 05/08/2015] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Genetic data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) have been crucial in advancing the understanding of Alzheimer's disease (AD) pathophysiology. Here, we provide an update on sample collection, scientific progress and opportunities, conceptual issues, and future plans. METHODS Lymphoblastoid cell lines and DNA and RNA samples from blood have been collected and banked, and data and biosamples have been widely disseminated. To date, APOE genotyping, genome-wide association study (GWAS), and whole exome and whole genome sequencing data have been obtained and disseminated. RESULTS ADNI genetic data have been downloaded thousands of times, and >300 publications have resulted, including reports of large-scale GWAS by consortia to which ADNI contributed. Many of the first applications of quantitative endophenotype association studies used ADNI data, including some of the earliest GWAS and pathway-based studies of biospecimen and imaging biomarkers, as well as memory and other clinical/cognitive variables. Other contributions include some of the first whole exome and whole genome sequencing data sets and reports in healthy controls, mild cognitive impairment, and AD. DISCUSSION Numerous genetic susceptibility and protective markers for AD and disease biomarkers have been identified and replicated using ADNI data and have heavily implicated immune, mitochondrial, cell cycle/fate, and other biological processes. Early sequencing studies suggest that rare and structural variants are likely to account for significant additional phenotypic variation. Longitudinal analyses of transcriptomic, proteomic, metabolomic, and epigenomic changes will also further elucidate dynamic processes underlying preclinical and prodromal stages of disease. Integration of this unique collection of multiomics data within a systems biology framework will help to separate truly informative markers of early disease mechanisms and potential novel therapeutic targets from the vast background of less relevant biological processes. Fortunately, a broad swath of the scientific community has accepted this grand challenge.
Collapse
Affiliation(s)
- Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Li Shen
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiaohui Yao
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; School of Informatics and Computing, Indiana University, Purdue University - Indianapolis, Indianapolis, IN, USA
| | - Sungeun Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Vijay K Ramanan
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tatiana M Foroud
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kelley M Faber
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Xiaolan Hu
- Bristol-Myers Squibb, Wallingford, CT, USA
| | | | - Steven G Potkin
- Department of Psychiatry and Human Behavior, University of California - Irvine, Irvine, CA, USA
| | - Paul M Thompson
- Department of Neurology, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA; Imaging Genetics Center, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, UT, USA; Department of Neuroscience, Brigham Young University, Provo, UT, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA; Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Robert C Green
- Partners Center for Personalized Genetic Medicine, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, Institute for Neuroimaging and Neuroinformatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Michael W Weiner
- Department of Radiology, University of California-San Francisco, San Francisco, CA, USA; Department of Medicine, University of California-San Francisco, San Francisco, CA, USA; Department of Psychiatry, University of California-San Francisco, San Francisco, CA, USA; Center for Imaging of Neurodegenerative Diseases, San Francisco VA Medical Center, San Francisco, CA, USA
| |
Collapse
|
40
|
Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Cedarbaum J, Green RC, Harvey D, Jack CR, Jagust W, Luthman J, Morris JC, Petersen RC, Saykin AJ, Shaw L, Shen L, Schwarz A, Toga AW, Trojanowski JQ. 2014 Update of the Alzheimer's Disease Neuroimaging Initiative: A review of papers published since its inception. Alzheimers Dement 2015; 11:e1-120. [PMID: 26073027 PMCID: PMC5469297 DOI: 10.1016/j.jalz.2014.11.001] [Citation(s) in RCA: 214] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/18/2013] [Indexed: 01/18/2023]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) is an ongoing, longitudinal, multicenter study designed to develop clinical, imaging, genetic, and biochemical biomarkers for the early detection and tracking of Alzheimer's disease (AD). The initial study, ADNI-1, enrolled 400 subjects with early mild cognitive impairment (MCI), 200 with early AD, and 200 cognitively normal elderly controls. ADNI-1 was extended by a 2-year Grand Opportunities grant in 2009 and by a competitive renewal, ADNI-2, which enrolled an additional 550 participants and will run until 2015. This article reviews all papers published since the inception of the initiative and summarizes the results to the end of 2013. The major accomplishments of ADNI have been as follows: (1) the development of standardized methods for clinical tests, magnetic resonance imaging (MRI), positron emission tomography (PET), and cerebrospinal fluid (CSF) biomarkers in a multicenter setting; (2) elucidation of the patterns and rates of change of imaging and CSF biomarker measurements in control subjects, MCI patients, and AD patients. CSF biomarkers are largely consistent with disease trajectories predicted by β-amyloid cascade (Hardy, J Alzheimer's Dis 2006;9(Suppl 3):151-3) and tau-mediated neurodegeneration hypotheses for AD, whereas brain atrophy and hypometabolism levels show predicted patterns but exhibit differing rates of change depending on region and disease severity; (3) the assessment of alternative methods of diagnostic categorization. Currently, the best classifiers select and combine optimum features from multiple modalities, including MRI, [(18)F]-fluorodeoxyglucose-PET, amyloid PET, CSF biomarkers, and clinical tests; (4) the development of blood biomarkers for AD as potentially noninvasive and low-cost alternatives to CSF biomarkers for AD diagnosis and the assessment of α-syn as an additional biomarker; (5) the development of methods for the early detection of AD. CSF biomarkers, β-amyloid 42 and tau, as well as amyloid PET may reflect the earliest steps in AD pathology in mildly symptomatic or even nonsymptomatic subjects and are leading candidates for the detection of AD in its preclinical stages; (6) the improvement of clinical trial efficiency through the identification of subjects most likely to undergo imminent future clinical decline and the use of more sensitive outcome measures to reduce sample sizes. Multimodal methods incorporating APOE status and longitudinal MRI proved most highly predictive of future decline. Refinements of clinical tests used as outcome measures such as clinical dementia rating-sum of boxes further reduced sample sizes; (7) the pioneering of genome-wide association studies that leverage quantitative imaging and biomarker phenotypes, including longitudinal data, to confirm recently identified loci, CR1, CLU, and PICALM and to identify novel AD risk loci; (8) worldwide impact through the establishment of ADNI-like programs in Japan, Australia, Argentina, Taiwan, China, Korea, Europe, and Italy; (9) understanding the biology and pathobiology of normal aging, MCI, and AD through integration of ADNI biomarker and clinical data to stimulate research that will resolve controversies about competing hypotheses on the etiopathogenesis of AD, thereby advancing efforts to find disease-modifying drugs for AD; and (10) the establishment of infrastructure to allow sharing of all raw and processed data without embargo to interested scientific investigators throughout the world.
Collapse
Affiliation(s)
- Michael W Weiner
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA; Department of Radiology, University of California, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, CA, USA.
| | - Dallas P Veitch
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA
| | - Paul S Aisen
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Laurel A Beckett
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Nigel J Cairns
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jesse Cedarbaum
- Neurology Early Clinical Development, Biogen Idec, Cambridge, MA, USA
| | - Robert C Green
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Danielle Harvey
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | | | - William Jagust
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Johan Luthman
- Neuroscience Clinical Development, Neuroscience & General Medicine Product Creation Unit, Eisai Inc., Philadelphia, PA, USA
| | - John C Morris
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | | | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leslie Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adam Schwarz
- Tailored Therapeutics, Eli Lilly and Company, Indianapolis, IN, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, Institute of Neuroimaging and Informatics, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - John Q Trojanowski
- Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Alzheimer's Disease Core Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Udall Parkinson's Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
41
|
Yan J, Kim S, Nho K, Chen R, Risacher SL, Moore JH, Saykin AJ, Shen L. Hippocampal transcriptome-guided genetic analysis of correlated episodic memory phenotypes in Alzheimer's disease. Front Genet 2015; 6:117. [PMID: 25859259 PMCID: PMC4374536 DOI: 10.3389/fgene.2015.00117] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 03/09/2015] [Indexed: 01/18/2023] Open
Abstract
As the most common type of dementia, Alzheimer's disease (AD) is a neurodegenerative disorder initially manifested by impaired memory performances. While the diagnosis information indicates a dichotomous status of a patient, memory scores have the potential to capture the continuous nature of the disease progression and may provide more insights into the underlying mechanism. In this work, we performed a targeted genetic study of memory scores on an AD cohort to identify the associations between a set of genes highly expressed in the hippocampal region and seven cognitive scores related to episodic memory. Both main effects and interaction effects of the targeted genetic markers on these correlated memory scores were examined. In addition to well-known AD genetic markers APOE and TOMM40, our analysis identified a new risk gene NAV2 through the gene-level main effect analysis. NAV2 was found to be significantly and consistently associated with all seven episodic memory scores. Genetic interaction analysis also yielded a few promising hits warranting further investigation, especially for the RAVLT list B Score.
Collapse
Affiliation(s)
- Jingwen Yan
- BioHealth, Indiana University School of Informatics and Computing Indianapolis, IN, USA ; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA
| | - Sungeun Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine Indianapolis, IN, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine Indianapolis, IN, USA
| | - Rui Chen
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Computer Science, Dartmouth College Hanover, NH, USA
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine Indianapolis, IN, USA
| | - Jason H Moore
- Genetics, Community and Family Medicine, Geisel School of Medicine at Dartmouth Lebanon, NH, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine Indianapolis, IN, USA ; Medical and Molecular Genetics, Indiana University School of Medicine Indianapolis, IN, USA
| | - Li Shen
- BioHealth, Indiana University School of Informatics and Computing Indianapolis, IN, USA ; Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine Indianapolis, IN, USA ; Indiana Alzheimer Disease Center, Indiana University School of Medicine Indianapolis, IN, USA ; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine Indianapolis, IN, USA
| | | |
Collapse
|
42
|
Kauwe JSK, Bailey MH, Ridge PG, Perry R, Wadsworth ME, Hoyt KL, Staley LA, Karch CM, Harari O, Cruchaga C, Ainscough BJ, Bales K, Pickering EH, Bertelsen S, Fagan AM, Holtzman DM, Morris JC, Goate AM. Genome-wide association study of CSF levels of 59 alzheimer's disease candidate proteins: significant associations with proteins involved in amyloid processing and inflammation. PLoS Genet 2014; 10:e1004758. [PMID: 25340798 PMCID: PMC4207667 DOI: 10.1371/journal.pgen.1004758] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 09/16/2014] [Indexed: 01/25/2023] Open
Abstract
Cerebrospinal fluid (CSF) 42 amino acid species of amyloid beta (Aβ42) and tau levels are strongly correlated with the presence of Alzheimer's disease (AD) neuropathology including amyloid plaques and neurodegeneration and have been successfully used as endophenotypes for genetic studies of AD. Additional CSF analytes may also serve as useful endophenotypes that capture other aspects of AD pathophysiology. Here we have conducted a genome-wide association study of CSF levels of 59 AD-related analytes. All analytes were measured using the Rules Based Medicine Human DiscoveryMAP Panel, which includes analytes relevant to several disease-related processes. Data from two independently collected and measured datasets, the Knight Alzheimer's Disease Research Center (ADRC) and Alzheimer's Disease Neuroimaging Initiative (ADNI), were analyzed separately, and combined results were obtained using meta-analysis. We identified genetic associations with CSF levels of 5 proteins (Angiotensin-converting enzyme (ACE), Chemokine (C-C motif) ligand 2 (CCL2), Chemokine (C-C motif) ligand 4 (CCL4), Interleukin 6 receptor (IL6R) and Matrix metalloproteinase-3 (MMP3)) with study-wide significant p-values (p<1.46×10−10) and significant, consistent evidence for association in both the Knight ADRC and the ADNI samples. These proteins are involved in amyloid processing and pro-inflammatory signaling. SNPs associated with ACE, IL6R and MMP3 protein levels are located within the coding regions of the corresponding structural gene. The SNPs associated with CSF levels of CCL4 and CCL2 are located in known chemokine binding proteins. The genetic associations reported here are novel and suggest mechanisms for genetic control of CSF and plasma levels of these disease-related proteins. Significant SNPs in ACE and MMP3 also showed association with AD risk. Our findings suggest that these proteins/pathways may be valuable therapeutic targets for AD. Robust associations in cognitively normal individuals suggest that these SNPs also influence regulation of these proteins more generally and may therefore be relevant to other diseases. The use of quantitative endophenotypes from cerebrospinal fluid has led to the identification of several genetic variants that alter risk or rate of progression of Alzheimer's disease. Here we have analyzed the levels of 58 disease-related proteins in the cerebrospinal fluid for association with millions of variants across the human genome. We have identified significant, replicable associations with 5 analytes, Angiotensin-converting enzyme, Chemokine (C-C motif) ligand 2, Chemokine (C-C motif) ligand 4, Interleukin 6 receptor and Matrix metalloproteinase-3. Our results suggest that these variants play a regulatory role in the respective protein levels and are relevant to the inflammatory and amyloid processing pathways. Variants in associated with ACE and those associated with MMP3 levels also show association with risk for Alzheimer's disease in the expected directions. These associations are consistent in cerebrospinal fluid and plasma and in samples with only cognitively normal individuals suggesting that they are relevant in the regulation of these protein levels beyond the context of Alzheimer's disease.
Collapse
Affiliation(s)
- John S. K. Kauwe
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Matthew H. Bailey
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Perry G. Ridge
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Rachel Perry
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Mark E. Wadsworth
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Kaitlyn L. Hoyt
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Lyndsay A. Staley
- Department of Biology, Brigham Young University, Provo, Utah, United States of America
| | - Celeste M. Karch
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Oscar Harari
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Benjamin J. Ainscough
- The Genome Institute, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Kelly Bales
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut, United States of America
| | - Eve H. Pickering
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut, United States of America
| | - Sarah Bertelsen
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, United States of America
| | | | - Anne M. Fagan
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - David M. Holtzman
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Developmental Biology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - John C. Morris
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Alison M. Goate
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, United States of America
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
43
|
Maternal immune activation and abnormal brain development across CNS disorders. Nat Rev Neurol 2014; 10:643-60. [PMID: 25311587 DOI: 10.1038/nrneurol.2014.187] [Citation(s) in RCA: 618] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Epidemiological studies have shown a clear association between maternal infection and schizophrenia or autism in the progeny. Animal models have revealed maternal immune activation (mIA) to be a profound risk factor for neurochemical and behavioural abnormalities in the offspring. Microglial priming has been proposed as a major consequence of mIA, and represents a critical link in a causal chain that leads to the wide spectrum of neuronal dysfunctions and behavioural phenotypes observed in the juvenile, adult or aged offspring. Such diversity of phenotypic outcomes in the mIA model are mirrored by recent clinical evidence suggesting that infectious exposure during pregnancy is also associated with epilepsy and, to a lesser extent, cerebral palsy in children. Preclinical research also suggests that mIA might precipitate the development of Alzheimer and Parkinson diseases. Here, we summarize and critically review the emerging evidence that mIA is a shared environmental risk factor across CNS disorders that varies as a function of interactions between genetic and additional environmental factors. We also review ongoing clinical trials targeting immune pathways affected by mIA that may play a part in disease manifestation. In addition, future directions and outstanding questions are discussed, including potential symptomatic, disease-modifying and preventive treatment strategies.
Collapse
|
44
|
Toledo JB, Da X, Weiner MW, Wolk DA, Xie SX, Arnold SE, Davatzikos C, Shaw LM, Trojanowski JQ. CSF Apo-E levels associate with cognitive decline and MRI changes. Acta Neuropathol 2014; 127:621-32. [PMID: 24385135 DOI: 10.1007/s00401-013-1236-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/16/2013] [Accepted: 12/17/2013] [Indexed: 12/31/2022]
Abstract
Apolipoprotein E (APOE) ε4 allele is the most important genetic risk factor for Alzheimer's disease (AD) and it is thought to do so by modulating levels of its product, apolipoprotein E (Apo-E), and regulating amyloid-β (Aβ) clearance. However, information on clinical and biomarker correlates of Apo-E proteins is scarce. We examined the relationship of cerebrospinal fluid (CSF) and plasma Apo-E protein levels, and APOE genotype to cognition and AD biomarker changes in 311 AD neuroimaging initiative subjects with CSF Apo-E measurements and 565 subjects with plasma Apo-E measurements. At baseline, higher CSF Apo-E levels were associated with higher total and phosphorylated CSF tau levels. CSF Apo-E levels were associated with longitudinal cognitive decline, MCI conversion to dementia, and gray matter atrophy rate in total tau/Aβ1-42 ratio and APOE genotype-adjusted analyses. In analyses stratified by APOE genotype, our results were only significant in the group without the ε4 allele. Baseline CSF Apo-E levels did not predict longitudinal CSF Aβ or tau changes. Plasma Apo-E levels show a mild correlation with CSF Apo-E levels, but were not associated with longitudinal cognitive and MRI changes. Based on our analyses, we speculate that increased CSF Apo-E2 or -E3 levels might represent a protective response to injury in AD and may have neuroprotective effects by decreasing neuronal damage independent of tau and amyloid deposition in addition to its effects on amyloid clearance.
Collapse
Affiliation(s)
- Jon B Toledo
- Department of Pathology and Laboratory Medicine, Institute on Aging, Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, HUP, Maloney 3rd, 36th and Spruce Streets, Philadelphia, PA, 19104-4283, USA
| | | | | | | | | | | | | | | | | |
Collapse
|