151
|
Salvatore JE, Han S, Farris SP, Mignogna KM, Miles MF, Agrawal A. Beyond genome-wide significance: integrative approaches to the interpretation and extension of GWAS findings for alcohol use disorder. Addict Biol 2019; 24:275-289. [PMID: 29316088 PMCID: PMC6037617 DOI: 10.1111/adb.12591] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 11/20/2017] [Accepted: 11/26/2017] [Indexed: 12/16/2022]
Abstract
Alcohol use disorder (AUD) is a heritable complex behavior. Due to the highly polygenic nature of AUD, identifying genetic variants that comprise this heritable variation has proved to be challenging. With the exception of functional variants in alcohol metabolizing genes (e.g. ADH1B and ALDH2), few other candidate loci have been confidently linked to AUD. Genome-wide association studies (GWAS) of AUD and other alcohol-related phenotypes have either produced few hits with genome-wide significance or have failed to replicate on further study. These issues reinforce the complex nature of the genetic underpinnings for AUD and suggest that both GWAS studies with larger samples and additional analysis approaches that better harness the nominally significant loci in existing GWAS are needed. Here, we review approaches of interest in the post-GWAS era, including in silico functional analyses; functional partitioning of single nucleotide polymorphism heritability; aggregation of signal into genes and gene networks; and validation of identified loci, genes and gene networks in postmortem brain tissue and across species. These integrative approaches hold promise to illuminate our understanding of the biological basis of AUD; however, we recognize that the main challenge continues to be the extremely polygenic nature of AUD, which necessitates large samples to identify multiple loci associated with AUD liability.
Collapse
Affiliation(s)
- Jessica E. Salvatore
- Department of Psychology; Virginia Commonwealth University; Richmond VA USA
- Virginia Institute for Psychiatric and Behavioral Genetics; Virginia Commonwealth University; Richmond VA USA
| | - Shizhong Han
- Department of Psychiatry; University of Iowa; Iowa City IA USA
- Department of Psychiatry and Behavioral Sciences; Johns Hopkins School of Medicine; Baltimore MD USA
| | - Sean P. Farris
- Waggoner Center for Alcohol and Addiction Research; The University of Texas at Austin; Austin TX USA
| | - Kristin M. Mignogna
- Virginia Institute for Psychiatric and Behavioral Genetics; Virginia Commonwealth University; Richmond VA USA
| | - Michael F. Miles
- Department of Pharmacology and Toxicology; Virginia Commonwealth University; Richmond VA USA
| | - Arpana Agrawal
- Department of Psychiatry; Washington University School of Medicine; Saint Louis MO USA
| |
Collapse
|
152
|
Candidate gene analyses for acute pain and morphine analgesia after pediatric day surgery: African American versus European Caucasian ancestry and dose prediction limits. THE PHARMACOGENOMICS JOURNAL 2019; 19:570-581. [PMID: 30760877 PMCID: PMC6693985 DOI: 10.1038/s41397-019-0074-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 10/30/2018] [Accepted: 12/21/2018] [Indexed: 12/17/2022]
Abstract
Acute pain and opioid analgesia demonstrate inter-individual variability and polygenic influence. In 241 children of African American and 277 of European Caucasian ancestry, we sought to replicate select candidate gene associations with morphine dose and postoperative pain and then to estimate dose prediction limits. Twenty-seven single-nucleotide polymorphisms (SNPs) from nine genes (ABCB1, ARRB2, COMT, DRD2, KCNJ6, MC1R, OPRD1, OPRM1, and UGT2B7) met selection criteria and were analyzed along with TAOK3. Few associations replicated: morphine dose (mcg/kg) in African American children and ABCB1 rs1045642 (A allele, β = -9.30, 95% CI: -17.25 to -1.35, p = 0.02) and OPRM1 rs1799971 (G allele, β = 23.19, 95% CI: 3.27-43.11, p = 0.02); KCNJ6 rs2211843 and high pain in African American subjects (T allele, OR 2.08, 95% CI: 1.17-3.71, p = 0.01) and in congruent European Caucasian pain phenotypes; and COMT rs740603 for high pain in European Caucasian subjects (A allele, OR: 0.69, 95% CI: 0.48-0.99, p = 0.046). With age, body mass index, and physical status as covariates, simple top SNP candidate gene models could explain theoretical maximums of 24.2% (European Caucasian) and 14.6% (African American) of morphine dose variances.
Collapse
|
153
|
Ferreira MAR, Vonk JM, Baurecht H, Marenholz I, Tian C, Hoffman JD, Helmer Q, Tillander A, Ullemar V, Lu Y, Rüschendorf F, Hinds DA, Hübner N, Weidinger S, Magnusson PKE, Jorgenson E, Lee YA, Boomsma DI, Karlsson R, Almqvist C, Koppelman GH, Paternoster L. Eleven loci with new reproducible genetic associations with allergic disease risk. J Allergy Clin Immunol 2019; 143:691-699. [PMID: 29679657 PMCID: PMC7189804 DOI: 10.1016/j.jaci.2018.03.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/01/2018] [Accepted: 03/19/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND A recent genome-wide association study (GWAS) identified 99 loci that contain genetic risk variants shared between asthma, hay fever, and eczema. Many more risk loci shared between these common allergic diseases remain to be discovered, which could point to new therapeutic opportunities. OBJECTIVE We sought to identify novel risk loci shared between asthma, hay fever, and eczema by applying a gene-based test of association to results from a published GWAS that included data from 360,838 subjects. METHODS We used approximate conditional analysis to adjust the results from the published GWAS for the effects of the top risk variants identified in that study. We then analyzed the adjusted GWAS results with the EUGENE gene-based approach, which combines evidence for association with disease risk across regulatory variants identified in different tissues. Novel gene-based associations were followed up in an independent sample of 233,898 subjects from the UK Biobank study. RESULTS Of the 19,432 genes tested, 30 had a significant gene-based association at a Bonferroni-corrected P value of 2.5 × 10-6. Of these, 20 were also significantly associated (P < .05/30 = .0016) with disease risk in the replication sample, including 19 that were located in 11 loci not reported to contain allergy risk variants in previous GWASs. Among these were 9 genes with a known function that is directly relevant to allergic disease: FOSL2, VPRBP, IPCEF1, PRR5L, NCF4, APOBR, IL27, ATXN2L, and LAT. For 4 genes (eg, ATXN2L), a genetically determined decrease in gene expression was associated with decreased allergy risk, and therefore drugs that inhibit gene expression or function are predicted to ameliorate disease symptoms. The opposite directional effect was observed for 14 genes, including IL27, a cytokine known to suppress TH2 responses. CONCLUSION Using a gene-based approach, we identified 11 risk loci for allergic disease that were not reported in previous GWASs. Functional studies that investigate the contribution of the 19 associated genes to the pathophysiology of allergic disease and assess their therapeutic potential are warranted.
Collapse
Affiliation(s)
- Manuel A R Ferreira
- Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Australia.
| | - Judith M Vonk
- Epidemiology, University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Hansjörg Baurecht
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ingo Marenholz
- Max Delbrück Center (MDC) for Molecular Medicine, Berlin, Germany; Clinic for Pediatric Allergy, Experimental and Clinical Research Center of Charité Universitätsmedizin Berlin and Max Delbrück Center, Berlin, Germany
| | | | - Joshua D Hoffman
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, Calif
| | - Quinta Helmer
- Department Biological Psychology, Netherlands Twin Register, Vrije University, Amsterdam, The Netherlands
| | - Annika Tillander
- Department of Medical Epidemiology and Biostatistics and the Swedish Twin Registry, Karolinska Institutet, Stockholm, Sweden
| | - Vilhelmina Ullemar
- Department of Medical Epidemiology and Biostatistics and the Swedish Twin Registry, Karolinska Institutet, Stockholm, Sweden
| | - Yi Lu
- Department of Medical Epidemiology and Biostatistics and the Swedish Twin Registry, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Norbert Hübner
- Max Delbrück Center (MDC) for Molecular Medicine, Berlin, Germany
| | - Stephan Weidinger
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics and the Swedish Twin Registry, Karolinska Institutet, Stockholm, Sweden
| | - Eric Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, Calif
| | - Young-Ae Lee
- Max Delbrück Center (MDC) for Molecular Medicine, Berlin, Germany; Clinic for Pediatric Allergy, Experimental and Clinical Research Center of Charité Universitätsmedizin Berlin and Max Delbrück Center, Berlin, Germany
| | - Dorret I Boomsma
- Department Biological Psychology, Netherlands Twin Register, Vrije University, Amsterdam, The Netherlands
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics and the Swedish Twin Registry, Karolinska Institutet, Stockholm, Sweden
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics and the Swedish Twin Registry, Karolinska Institutet, Stockholm, Sweden; Pediatric Allergy and Pulmonology Unit at Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Gerard H Koppelman
- University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, Pediatric Pulmonology and Pediatric Allergology, and University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
154
|
Gettler K, Giri M, Kenigsberg E, Martin J, Chuang LS, Hsu NY, Denson LA, Hyams JS, Griffiths A, Noe JD, Crandall WV, Mack DR, Kellermayer R, Abraham C, Hoffman G, Kugathasan S, Cho JH. Prioritizing Crohn's disease genes by integrating association signals with gene expression implicates monocyte subsets. Genes Immun 2019; 20:577-588. [PMID: 30692607 DOI: 10.1038/s41435-019-0059-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/27/2018] [Accepted: 01/07/2019] [Indexed: 12/19/2022]
Abstract
Genome-wide association studies have identified ~170 loci associated with Crohn's disease (CD) and defining which genes drive these association signals is a major challenge. The primary aim of this study was to define which CD locus genes are most likely to be disease related. We developed a gene prioritization regression model (GPRM) by integrating complementary mRNA expression datasets, including bulk RNA-Seq from the terminal ileum of 302 newly diagnosed, untreated CD patients and controls, and in stimulated monocytes. Transcriptome-wide association and co-expression network analyses were performed on the ileal RNA-Seq datasets, identifying 40 genome-wide significant genes. Co-expression network analysis identified a single gene module, which was substantially enriched for CD locus genes and most highly expressed in monocytes. By including expression-based and epigenetic information, we refined likely CD genes to 2.5 prioritized genes per locus from an average of 7.8 total genes. We validated our model structure using cross-validation and our prioritization results by protein-association network analyses, which demonstrated significantly higher CD gene interactions for prioritized compared with non-prioritized genes. Although individual datasets cannot convey all of the information relevant to a disease, combining data from multiple relevant expression-based datasets improves prediction of disease genes and helps to further understanding of disease pathogenesis.
Collapse
Affiliation(s)
- Kyle Gettler
- Department of Genetics, Yale University, New Haven, Connecticut, 06510, USA
| | - Mamta Giri
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Ephraim Kenigsberg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Jerome Martin
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Ling-Shiang Chuang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Nai-Yun Hsu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Lee A Denson
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Oio, USA
| | - Jeffrey S Hyams
- Division of Digestive Diseases, Hepatology, and Nutrition, Connecticut Children's Medical Center, Hartford, Connecticut, USA
| | - Anne Griffiths
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Joshua D Noe
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Wallace V Crandall
- Department of Pediatric Gastroenterology, Nationwide Children's Hospital, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - David R Mack
- Department of Pediatrics, Children's Hospital of Eastern Ontario IBD Centre and University of Ottawa, Ottawa, Ontario, Canada
| | - Richard Kellermayer
- Section of Pediatric Gastroenterology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Clara Abraham
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, Connecticut, 06510, USA
| | - Gabriel Hoffman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Subra Kugathasan
- Division of Pediatric Gastroenterology, Emory University School of Medicine, Atlanta, Georgia, USA.,Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Judy H Cho
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA. .,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.
| |
Collapse
|
155
|
Comparison of methods for multivariate gene-based association tests for complex diseases using common variants. Eur J Hum Genet 2019; 27:811-823. [PMID: 30683923 PMCID: PMC6461986 DOI: 10.1038/s41431-018-0327-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 10/30/2018] [Accepted: 12/04/2018] [Indexed: 12/29/2022] Open
Abstract
Complex diseases are usually associated with multiple correlated phenotypes, and the analysis of composite scores or disease status may not fully capture the complexity (or multidimensionality). Joint analysis of multiple disease-related phenotypes in genetic tests could potentially increase power to detect association of a disease with common SNPs (or genes). Gene-based tests are designed to identify genes containing multiple risk variants that individually are weakly associated with a univariate trait. We combined three multivariate association tests (O'Brien method, TATES, and MultiPhen) with two gene-based association tests (GATES and VEGAS) and compared performance (type I error and power) of six multivariate gene-based methods using simulated data. Data (n = 2000) for genetic sequence and correlated phenotypes were simulated by varying causal variant proportions and phenotype correlations for various scenarios. These simulations showed that two multivariate association tests (TATES and MultiPhen, but not O'Brien) paired with VEGAS have inflated type I error in all scenarios, while the three multivariate association tests paired with GATES have correct type I error. MultiPhen paired with GATES has higher power than competing methods if the correlations among phenotypes are low (r < 0.57). We applied these gene-based association methods to a GWAS dataset from the Alzheimer's Disease Genetics Consortium containing three neuropathological traits related to Alzheimer disease (neuritic plaque, neurofibrillary tangles, and cerebral amyloid angiopathy) measured in 3500 autopsied brains. Gene-level significant evidence (P < 2.7 × 10-6) was identified in a region containing three contiguous genes (TRAPPC12, TRAPPC12-AS1, ADI1) using O'Brien and VEGAS. Gene-wide significant associations were not observed in univariate gene-based tests.
Collapse
|
156
|
Lin WY, Huang CC, Liu YL, Tsai SJ, Kuo PH. Genome-Wide Gene-Environment Interaction Analysis Using Set-Based Association Tests. Front Genet 2019; 9:715. [PMID: 30693016 PMCID: PMC6339974 DOI: 10.3389/fgene.2018.00715] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 12/20/2018] [Indexed: 12/22/2022] Open
Abstract
The identification of gene-environment interactions (G × E) may eventually guide health-related choices and medical interventions for complex diseases. More powerful methods must be developed to identify G × E. The “adaptive combination of Bayes factors method” (ADABF) has been proposed as a powerful genome-wide polygenic approach to detect G × E. In this work, we evaluate its performance when serving as a gene-based G × E test. We compare ADABF with six tests including the “Set-Based gene-EnviRonment InterAction test” (SBERIA), “gene-environment set association test” (GESAT), etc. With extensive simulations, SBERIA and ADABF are found to be more powerful than other G × E tests. However, SBERIA suffers from a power loss when 50% SNP main effects are in the same direction with the SNP × E interaction effects while 50% are in the opposite direction. We further applied these seven G × E methods to the Taiwan Biobank data to explore gene× alcohol interactions on blood pressure levels. The ADAMTS7P1 gene at chromosome 15q25.2 was detected to interact with alcohol consumption on diastolic blood pressure (p = 9.5 × 10−7, according to the GESAT test). At this gene, the P-values provided by other six tests all reached the suggestive significance level (p < 5 × 10−5). Regarding the computation time required for a genome-wide G × E analysis, SBERIA is the fastest method, followed by ADABF. Considering the validity, power performance, robustness, and computation time, ADABF is recommended for genome-wide G × E analyses.
Collapse
Affiliation(s)
- Wan-Yu Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Ching-Chieh Huang
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Psychiatry, National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsiu Kuo
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
157
|
Xiang B, Yang BZ, Zhou H, Kranzler HR, Gelernter J. GWAS and network analysis of co-occurring nicotine and alcohol dependence identifies significantly associated alleles and network. Am J Med Genet B Neuropsychiatr Genet 2019; 180:3-11. [PMID: 30488612 PMCID: PMC6918694 DOI: 10.1002/ajmg.b.32692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 08/02/2018] [Accepted: 09/26/2018] [Indexed: 12/11/2022]
Abstract
Alcohol dependence (AD) and nicotine dependence (ND) co-occur frequently (AD+ND). We integrated SNP-based, gene-based, and protein-protein interaction network analyses to identify shared risk genes or gene subnetworks for AD+ND in African Americans (AAs, N = 2,094) and European Americans (EAs, N = 1,207). The DSM-IV criterion counts for AD and ND were modeled as two dependent variables in a multivariate linear mixed model, and analyzed separately for the two populations. The most significant SNP was rs6579845 in EAs (p < 1.29 × 10-8 ) in GM2A, which encodes GM2 ganglioside activator, and is a cis-expression quantitative locus that affects GM2A expression in blood and brain tissues. However, this SNP was not replicated in our another small sample (N = 678). We identified a subnetwork of 24 genes that contributed to the AD+ND criterion counts. In the gene-set analysis for the subnetwork in an independent sample, the Study of Addiction: Genetics and Environment project (predominately EAs), these 24 genes as a set differed in AD+ND versus control subjects in EAs (p = .041). Functional enrichment analysis for this subnetwork revealed that the gene enrichment involved primarily nerve growth factor pathways, and cocaine and amphetamine addiction. In conclusion, we identified a genome-wide significant variant at GM2A and a gene subnetwork underlying the genetic trait of shared AD+ND. These results increase our understanding of the shared (pleiotropic) genetic risk that underlies AD+ND.
Collapse
Affiliation(s)
- Bo Xiang
- Department of Psychiatry, Yale University School of Medicine, New Haven, and VA CT Healthcare Center, West Haven, CT, USA,Department of Psychiatry, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Bao-Zhu Yang
- Department of Psychiatry, Yale University School of Medicine, New Haven, and VA CT Healthcare Center, West Haven, CT, USA
| | - Hang Zhou
- Department of Psychiatry, Yale University School of Medicine, New Haven, and VA CT Healthcare Center, West Haven, CT, USA
| | - Henry R. Kranzler
- Department of Psychiatry, Center for Studies of Addiction, University of Pennsylvania and VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, and VA CT Healthcare Center, West Haven, CT, USA,Departments of Genetics and Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
158
|
Ostrom QT, Coleman W, Huang W, Rubin JB, Lathia JD, Berens ME, Speyer G, Liao P, Wrensch MR, Eckel-Passow JE, Armstrong G, Rice T, Wiencke JK, McCoy LS, Hansen HM, Amos CI, Bernstein JL, Claus EB, Houlston RS, Il’yasova D, Jenkins RB, Johansen C, Lachance DH, Lai RK, Merrell RT, Olson SH, Sadetzki S, Schildkraut JM, Shete S, Andersson U, Rajaraman P, Chanock SJ, Linet MS, Wang Z, Yeager M, Melin B, Bondy ML, Barnholtz-Sloan JS. Sex-specific gene and pathway modeling of inherited glioma risk. Neuro Oncol 2019; 21:71-82. [PMID: 30124908 PMCID: PMC6303471 DOI: 10.1093/neuonc/noy135] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background To date, genome-wide association studies (GWAS) have identified 25 risk variants for glioma, explaining 30% of heritable risk. Most histologies occur with significantly higher incidence in males, and this difference is not explained by currently known risk factors. A previous GWAS identified sex-specific glioma risk variants, and this analysis aims to further elucidate risk variation by sex using gene- and pathway-based approaches. Methods Results from the Glioma International Case-Control Study were used as a testing set, and results from 3 GWAS were combined via meta-analysis and used as a validation set. Using summary statistics for nominally significant autosomal SNPs (P < 0.01 in a previous meta-analysis) and nominally significant X-chromosome SNPs (P < 0.01), 3 algorithms (Pascal, BimBam, and GATES) were used to generate gene scores, and Pascal was used to generate pathway scores. Results were considered statistically significant in the discovery set when P < 3.3 × 10-6 and in the validation set when P < 0.001 in 2 of 3 algorithms. Results Twenty-five genes within 5 regions and 19 genes within 6 regions reached statistical significance in at least 2 of 3 algorithms in males and females, respectively. EGFR was significantly associated with all glioma and glioblastoma in males only and a female-specific association in TERT, all of which remained nominally significant after conditioning on known risk loci. There were nominal associations with the BioCarta telomeres pathway in both males and females. Conclusions These results provide additional evidence that there may be differences by sex in genetic risk for glioma. Additional analyses may further elucidate the biological processes through which this risk is conferred.
Collapse
Affiliation(s)
- Quinn T Ostrom
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | - William Huang
- Case Western Reserve University, Cleveland, Ohio, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, USA; Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, USA
| | - Justin D Lathia
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Michael E Berens
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Gil Speyer
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Peter Liao
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Margaret R Wrensch
- Department of Neurological Surgery, School of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jeanette E Eckel-Passow
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Georgina Armstrong
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Terri Rice
- Department of Neurological Surgery, School of Medicine, University of California San Francisco, San Francisco, California, USA
| | - John K Wiencke
- Department of Neurological Surgery, School of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Lucie S McCoy
- Department of Neurological Surgery, School of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Helen M Hansen
- Department of Neurological Surgery, School of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Jonine L Bernstein
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elizabeth B Claus
- School of Public Health, Yale University, New Haven, Connecticut, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - Dora Il’yasova
- Department of Epidemiology and Biostatistics, School of Public Health, Georgia State University, Atlanta, Georgia, USA
- Cancer Control and Prevention Program, Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina, USA
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Christoffer Johansen
- Oncology Clinic, Finsen Center, Rigshospitalet and Survivorship Research Unit, The Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Daniel H Lachance
- Department of Neurology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Rose K Lai
- Departments of Neurology and Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ryan T Merrell
- Department of Neurology, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Sara H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Siegal Sadetzki
- Cancer and Radiation Epidemiology Unit, Gertner Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joellen M Schildkraut
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | | - Ulrika Andersson
- Department of Radiation Sciences, Faculty of Medicine, Umeå University, Umeå, Sweden
| | - Preetha Rajaraman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
- Core Genotyping Facility, National Cancer Institute, SAIC-Frederick, Inc, Gaithersburg, Maryland, USA
| | - Martha S Linet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
- Core Genotyping Facility, National Cancer Institute, SAIC-Frederick, Inc, Gaithersburg, Maryland, USA
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
- Core Genotyping Facility, National Cancer Institute, SAIC-Frederick, Inc, Gaithersburg, Maryland, USA
| | - Beatrice Melin
- Department of Radiation Sciences, Faculty of Medicine, Umeå University, Umeå, Sweden
| | - Melissa L Bondy
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Jill S Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
159
|
|
160
|
Picomolar concentrations of oligomeric alpha-synuclein sensitizes TLR4 to play an initiating role in Parkinson's disease pathogenesis. Acta Neuropathol 2019; 137:103-120. [PMID: 30225556 PMCID: PMC6338693 DOI: 10.1007/s00401-018-1907-y] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 09/03/2018] [Accepted: 09/03/2018] [Indexed: 12/11/2022]
Abstract
Despite the wealth of genomic and transcriptomic data in Parkinson’s disease (PD), the initial molecular events are unknown. Using LD score regression analysis, we show significant enrichment in PD heritability within regulatory sites for LPS-activated monocytes and that TLR4 expression is highest within human substantia nigra, the most affected brain region, suggesting a role for TLR4 inflammatory responses. We then performed extended incubation of cells with physiological concentrations of small alpha-synuclein oligomers observing the development of a TLR4-dependent sensitized inflammatory response with time, including TNF-α production. ROS and cell death in primary neuronal cultures were significantly reduced by TLR4 antagonists revealing that an indirect inflammatory mechanism involving cytokines produced by glial cells makes a major contribution to neuronal death. Prolonged exposure to low levels of alpha-synuclein oligomers sensitizes TLR4 responsiveness in astrocytes and microglial, explaining how they become pro-inflammatory, and may be an early causative event in PD.
Collapse
|
161
|
White MJ, Yaspan BL, Veatch OJ, Goddard P, Risse-Adams OS, Contreras MG. Strategies for Pathway Analysis Using GWAS and WGS Data. CURRENT PROTOCOLS IN HUMAN GENETICS 2019; 100:e79. [PMID: 30387919 PMCID: PMC6391732 DOI: 10.1002/cphg.79] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Single-allele study designs, commonly used in genome-wide association studies (GWAS) as well as the more recently developed whole genome sequencing (WGS) studies, are a standard approach for investigating the relationship of common variation within the human genome to a given phenotype of interest. However, single-allele association results published for many GWAS studies represent only the tip of the iceberg for the information that can be extracted from these datasets. The primary analysis strategy for GWAS entails association analysis in which only the single nucleotide polymorphisms (SNPs) with the strongest p-values are declared statistically significant due to issues arising from multiple testing and type I errors. Factors such as locus heterogeneity, epistasis, and multiple genes conferring small effects contribute to the complexity of the genetic models underlying phenotype expression. Thus, many biologically meaningful associations having lower effect sizes at individual genes are overlooked, making it difficult to separate true associations from a sea of false-positive associations. Organizing these individual SNPs into biologically meaningful groups to look at the overall effects of minor perturbations to genes and pathways is desirable. This pathway-based approach provides researchers with insight into the functional foundations of the phenotype being studied and allows testing of various genetic scenarios. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Marquitta J White
- Department of Medicine, Lung Biology Center, University of California San Francisco, San Francisco, California
| | | | - Olivia J Veatch
- Center for Sleep and Circadian Neurobiology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Pagé Goddard
- Department of Medicine, Lung Biology Center, University of California San Francisco, San Francisco, California
| | - Oona S Risse-Adams
- Department of Medicine, Lung Biology Center, University of California San Francisco, San Francisco, California
| | - Maria G Contreras
- Department of Medicine, Lung Biology Center, University of California San Francisco, San Francisco, California
- MARC, San Francisco State University, San Francisco, California
| |
Collapse
|
162
|
A combined analysis of genetically correlated traits identifies 187 loci and a role for neurogenesis and myelination in intelligence. Mol Psychiatry 2019; 24:169-181. [PMID: 29326435 PMCID: PMC6344370 DOI: 10.1038/s41380-017-0001-5] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/30/2017] [Accepted: 11/03/2017] [Indexed: 12/13/2022]
Abstract
Intelligence, or general cognitive function, is phenotypically and genetically correlated with many traits, including a wide range of physical, and mental health variables. Education is strongly genetically correlated with intelligence (rg = 0.70). We used these findings as foundations for our use of a novel approach-multi-trait analysis of genome-wide association studies (MTAG; Turley et al. 2017)-to combine two large genome-wide association studies (GWASs) of education and intelligence, increasing statistical power and resulting in the largest GWAS of intelligence yet reported. Our study had four goals: first, to facilitate the discovery of new genetic loci associated with intelligence; second, to add to our understanding of the biology of intelligence differences; third, to examine whether combining genetically correlated traits in this way produces results consistent with the primary phenotype of intelligence; and, finally, to test how well this new meta-analytic data sample on intelligence predicts phenotypic intelligence in an independent sample. By combining datasets using MTAG, our functional sample size increased from 199,242 participants to 248,482. We found 187 independent loci associated with intelligence, implicating 538 genes, using both SNP-based and gene-based GWAS. We found evidence that neurogenesis and myelination-as well as genes expressed in the synapse, and those involved in the regulation of the nervous system-may explain some of the biological differences in intelligence. The results of our combined analysis demonstrated the same pattern of genetic correlations as those from previous GWASs of intelligence, providing support for the meta-analysis of these genetically-related phenotypes.
Collapse
|
163
|
Gene-Based Nonparametric Testing of Interactions Using Distance Correlation Coefficient in Case-Control Association Studies. Genes (Basel) 2018; 9:genes9120608. [PMID: 30563156 PMCID: PMC6316506 DOI: 10.3390/genes9120608] [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: 11/06/2018] [Revised: 11/24/2018] [Accepted: 11/27/2018] [Indexed: 12/12/2022] Open
Abstract
Among the various statistical methods for identifying gene⁻gene interactions in qualitative genome-wide association studies (GWAS), gene-based methods have recently grown in popularity because they confer advantages in both statistical power and biological interpretability. However, most of these methods make strong assumptions about the form of the relationship between traits and single-nucleotide polymorphisms, which result in limited statistical power. In this paper, we propose a gene-based method based on the distance correlation coefficient called gene-based gene-gene interaction via distance correlation coefficient (GBDcor). The distance correlation (dCor) is a measurement of the dependency between two random vectors with arbitrary, and not necessarily equal, dimensions. We used the difference in dCor in case and control datasets as an indicator of gene⁻gene interaction, which was based on the assumption that the joint distribution of two genes in case subjects and in control subjects should not be significantly different if the two genes do not interact. We designed a permutation-based statistical test to evaluate the difference between dCor in cases and controls for a pair of genes, and we provided the p-value for the statistic to represent the significance of the interaction between the two genes. In experiments with both simulated and real-world data, our method outperformed previous approaches in detecting interactions accurately.
Collapse
|
164
|
Zhang H, Wheeler W, Song L, Yu K. Proper joint analysis of summary association statistics requires the adjustment of heterogeneity in SNP coverage pattern. Brief Bioinform 2018; 19:1337-1343. [PMID: 28981575 DOI: 10.1093/bib/bbx072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Indexed: 11/12/2022] Open
Abstract
As meta-analysis results published by consortia of genome-wide association studies (GWASs) become increasingly available, many association summary statistics-based multi-locus tests have been developed to jointly evaluate multiple single-nucleotide polymorphisms (SNPs) to reveal novel genetic architectures of various complex traits. The validity of these approaches relies on the accurate estimate of z-score correlations at considered SNPs, which in turn requires knowledge on the set of SNPs assessed by each study participating in the meta-analysis. However, this exact SNP coverage information is usually unavailable from the meta-analysis results published by GWAS consortia. In the absence of the coverage information, researchers typically estimate the z-score correlations by making oversimplified coverage assumptions. We show through real studies that such a practice can generate highly inflated type I errors, and we demonstrate the proper way to incorporate correct coverage information into multi-locus analyses. We advocate that consortia should make SNP coverage information available when posting their meta-analysis results, and that investigators who develop analytic tools for joint analyses based on summary data should pay attention to the variation in SNP coverage and adjust for it appropriately.
Collapse
Affiliation(s)
- Han Zhang
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, USA
| | | | - Lei Song
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., USA
| | - Kai Yu
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, USA
| |
Collapse
|
165
|
Liu H, Wang W, Zhang C, Xu C, Duan H, Tian X, Zhang D. Heritability and Genome-Wide Association Study of Plasma Cholesterol in Chinese Adult Twins. Front Endocrinol (Lausanne) 2018; 9:677. [PMID: 30498476 PMCID: PMC6249314 DOI: 10.3389/fendo.2018.00677] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022] Open
Abstract
Dyslipidemia represents a strong and independent risk factor for cardiovascular disease. Plasma cholesterol, such as total cholesterol (TC), low density lipoprotein cholesterol (LDL-C), and high density lipoprotein cholesterol (HDL-C), is the common indicator of diagnosing dyslipidemia. Here based on 382 Chinese twin pairs, we explored the magnitude of genetic impact on TC, HDL-C, LDL-C variation and further searched for genetic susceptibility loci for them using genome-wide association study (GWAS). The ACE model was the best fit model with additive genetic parameter (A) accounting for 26.6%, common or shared environmental parameter (C) accounting for 47.8%, unique/non-shared environmental parameter (E) accounting for 25.6% for the variance in HDL-C. The AE model was the best fit model for TC (A: 61.4%; E: 38.6%) and LDL-C (A: 65.5%; E: 34.5%). While no SNPs reached the genome-wide significance level (P < 5 × 10-8), 8, 14, 9 SNPs exceeded the suggestive significance level (P < 1 × 10-5) for TC, HDL-C, LDL-C, respectively. The promising genetic regions for TC, HDL-C, LDL-C were on chromosome 11 around rs7107698, chromosome 5 around rs12518218, chromosome 2 around rs10490120, respectively. Gene-based analysis found 1038, 1033 and 1090 genes nominally associated with TC, HDL-C, LDL-C (P < 0.05), especially FAF1, KLKB1 for TC, KLKB1 for HDL-C, and NTRK1, FAF1, SNTB2 for LDL-C, respectively. The number of common related genes among TC, HDL-C and LDL-C was 71, including FAF1, KLKB1, etc. Pathway enrichment analysis discovered known related pathways-zinc transporters, metal ion SLC transporters for TC, cell adhesion molecules CAMs, IL-6 signaling for HDL, FC epsilon RI signaling pathway, NFAT pathway for LDL, respectively. In conclusion, the TC and LDL-C level is moderately heritable and the HDL-C level is lowly heritable in Chinese population. The genomic loci, functional genes and pathways are identified to account for the heritability of plasma cholesterol level. Our findings provide important insights into plasma cholesterol molecular physiology and expect future research to replicate and validate our results.
Collapse
Affiliation(s)
- Hui Liu
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Caixia Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Chunsheng Xu
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, China
| | - Haiping Duan
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, China
| | - Xiaocao Tian
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| |
Collapse
|
166
|
Torgerson DG, Ballard PL, Keller RL, Oh SS, Huntsman S, Hu D, Eng C, Burchard EG, Ballard RA. Ancestry and genetic associations with bronchopulmonary dysplasia in preterm infants. Am J Physiol Lung Cell Mol Physiol 2018; 315:L858-L869. [PMID: 30113228 PMCID: PMC6295513 DOI: 10.1152/ajplung.00073.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/12/2018] [Accepted: 08/15/2018] [Indexed: 01/19/2023] Open
Abstract
Bronchopulmonary dysplasia in premature infants is a common and often severe lung disease with long-term sequelae. A genetic component is suspected but not fully defined. We performed an ancestry and genome-wide association study to identify variants, genes, and pathways associated with survival without bronchopulmonary dysplasia in 387 high-risk infants treated with inhaled nitric oxide in the Trial of Late Surfactant study. Global African genetic ancestry was associated with increased survival without bronchopulmonary dysplasia among infants of maternal self-reported Hispanic white race/ethnicity [odds ratio (OR) = 4.5, P = 0.01]. Admixture mapping found suggestive outcome associations with local African ancestry at chromosome bands 18q21 and 10q22 among infants of maternal self-reported African-American race/ethnicity. For all infants, the top individual variant identified was within the intron of NBL1, which is expressed in midtrimester lung and is an antagonist of bone morphogenetic proteins ( rs372271081 , OR = 0.17, P = 7.4 × 10-7). The protective allele of this variant was significantly associated with lower nitric oxide metabolites in the urine of non-Hispanic white infants ( P = 0.006), supporting a role in the racial differential response to nitric oxide. Interrogating genes upregulated in bronchopulmonary dysplasia lungs indicated association with variants in CCL18, a cytokine associated with fibrosis and interstitial lung disease, and pathway analyses implicated variation in genes involved in immune/inflammatory processes in response to infection and mechanical ventilation. Our results suggest that genetic variation related to lung development, drug metabolism, and immune response contribute to individual and racial/ethnic differences in respiratory outcomes following inhaled nitric oxide treatment of high-risk premature infants.
Collapse
Affiliation(s)
- Dara G Torgerson
- Department of Pediatrics, University of California , San Francisco, California
| | - Philip L Ballard
- Department of Pediatrics, University of California , San Francisco, California
| | - Roberta L Keller
- Department of Pediatrics, University of California , San Francisco, California
| | - Sam S Oh
- Department of Medicine, University of California , San Francisco, California
| | - Scott Huntsman
- Department of Medicine, University of California , San Francisco, California
| | - Donglei Hu
- Department of Medicine, University of California , San Francisco, California
| | - Celeste Eng
- Department of Medicine, University of California , San Francisco, California
| | - Esteban G Burchard
- Department of Medicine, University of California , San Francisco, California
- Department of Bioengineering and Therapeutic Sciences, University of California , San Francisco, California
| | - Roberta A Ballard
- Department of Pediatrics, University of California , San Francisco, California
| |
Collapse
|
167
|
Choi S, Lee S, Kim Y, Hwang H, Park T. HisCoM-GGI: Hierarchical structural component analysis of gene-gene interactions. J Bioinform Comput Biol 2018; 16:1840026. [PMID: 30567476 DOI: 10.1142/s0219720018400267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although genome-wide association studies (GWAS) have successfully identified thousands of single nucleotide polymorphisms (SNPs) associated with common diseases, these observations are limited for fully explaining "missing heritability". Determining gene-gene interactions (GGI) are one possible avenue for addressing the missing heritability problem. While many statistical approaches have been proposed to detect GGI, most of these focus primarily on SNP-to-SNP interactions. While there are many advantages of gene-based GGI analyses, such as reducing the burden of multiple-testing correction, and increasing power by aggregating multiple causal signals across SNPs in specific genes, only a few methods are available. In this study, we proposed a new statistical approach for gene-based GGI analysis, "Hierarchical structural CoMponent analysis of Gene-Gene Interactions" (HisCoM-GGI). HisCoM-GGI is based on generalized structured component analysis, and can consider hierarchical structural relationships between genes and SNPs. For a pair of genes, HisCoM-GGI first effectively summarizes all possible pairwise SNP-SNP interactions into a latent variable, from which it then performs GGI analysis. HisCoM-GGI can evaluate both gene-level and SNP-level interactions. Through simulation studies, HisCoM-GGI demonstrated higher statistical power than existing gene-based GGI methods, in analyzing a GWAS of a Korean population for identifying GGI associated with body mass index. Resultantly, HisCoM-GGI successfully identified 14 potential GGI, two of which, (NCOR2 <mml:math xmlns:mml="http://www.w3.org/1998/Math/MathML"><mml:mo>×</mml:mo></mml:math> SPOCK1) and (LINGO2 <mml:math xmlns:mml="http://www.w3.org/1998/Math/MathML"><mml:mo>×</mml:mo></mml:math> ZNF385D) were successfully replicated in independent datasets. We conclude that HisCoM-GGI method may be a valuable tool for genome to identify GGI in missing heritability, allowing us to better understand the biological genetic mechanisms of complex traits. We conclude that HisCoM-GGI method may be a valuable tool for genome to identify GGI in missing heritability, allowing us to better understand biological genetic mechanisms of complex traits. An implementation of HisCoM-GGI can be downloaded from the website ( http://statgen.snu.ac.kr/software/hiscom-ggi ).
Collapse
Affiliation(s)
- Sungkyoung Choi
- Department of Pharmacology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sungyoung Lee
- Center for Precision Medicine, Seoul National University Hospital, 71 Daehak-ro Jongno-gu, Seoul 03082, Republic of Korea
| | - Yongkang Kim
- Department of Statistics, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul 08826, Republic of Korea.,Department of Psychology, McGill University, 2001 Avenue McGill College, Montreal, Quebec H3A 1G1, Canada
| | - Heungsun Hwang
- Department of Psychology, McGill University, 2001 Avenue McGill College, Montreal, Quebec H3A 1G1, Canada
| | - Taesung Park
- Department of Statistics, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul 08826, Republic of Korea.,Interdisciplinary Program in Bioinformatics, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul 08826, Republic of Korea
| |
Collapse
|
168
|
Duan X, Yang S, Zhang L, Yang T. V-ATPases and osteoclasts: ambiguous future of V-ATPases inhibitors in osteoporosis. Theranostics 2018; 8:5379-5399. [PMID: 30555553 PMCID: PMC6276090 DOI: 10.7150/thno.28391] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022] Open
Abstract
Vacuolar ATPases (V-ATPases) play a critical role in regulating extracellular acidification of osteoclasts and bone resorption. The deficiencies of subunit a3 and d2 of V-ATPases result in increased bone density in humans and mice. One of the traditional drug design strategies in treating osteoporosis is the use of subunit a3 inhibitor. Recent findings connect subunits H and G1 with decreased bone density. Given the controversial effects of ATPase subunits on bone density, there is a critical need to review the subunits of V-ATPase in osteoclasts and their functions in regulating osteoclasts and bone remodeling. In this review, we comprehensively address the following areas: information about all V-ATPase subunits and their isoforms; summary of V-ATPase subunits associated with human genetic diseases; V-ATPase subunits and osteopetrosis/osteoporosis; screening of all V-ATPase subunits variants in GEFOS data and in-house data; spectrum of V-ATPase subunits during osteoclastogenesis; direct and indirect roles of subunits of V-ATPases in osteoclasts; V-ATPase-associated signaling pathways in osteoclasts; interactions among V-ATPase subunits in osteoclasts; osteoclast-specific V-ATPase inhibitors; perspective of future inhibitors or activators targeting V-ATPase subunits in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Xiaohong Duan
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral Biology, Clinic of Oral Rare and Genetic Diseases, School of Stomatology, the Fourth Military Medical University, 145 West Changle Road, Xi'an 710032, P. R. China
| | - Shaoqing Yang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral Biology, Clinic of Oral Rare and Genetic Diseases, School of Stomatology, the Fourth Military Medical University, 145 West Changle Road, Xi'an 710032, P. R. China
| | - Lei Zhang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, P. R. China
| | - Tielin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, 28 West Xianning Road, Xi'an 710049, People's Republic of China
| |
Collapse
|
169
|
Zhu X, Stephens M. Large-scale genome-wide enrichment analyses identify new trait-associated genes and pathways across 31 human phenotypes. Nat Commun 2018; 9:4361. [PMID: 30341297 PMCID: PMC6195536 DOI: 10.1038/s41467-018-06805-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/29/2018] [Indexed: 12/27/2022] Open
Abstract
Genome-wide association studies (GWAS) aim to identify genetic factors associated with phenotypes. Standard analyses test variants for associations individually. However, variant-level associations are hard to identify and can be difficult to interpret biologically. Enrichment analyses help address both problems by targeting sets of biologically related variants. Here we introduce a new model-based enrichment method that requires only GWAS summary statistics. Applying this method to interrogate 4,026 gene sets in 31 human phenotypes identifies many previously-unreported enrichments, including enrichments of endochondral ossification pathway for height, NFAT-dependent transcription pathway for rheumatoid arthritis, brain-related genes for coronary artery disease, and liver-related genes for Alzheimer’s disease. A key feature of our method is that inferred enrichments automatically help identify new trait-associated genes. For example, accounting for enrichment in lipid transport genes highlights association between MTTP and low-density lipoprotein levels, whereas conventional analyses of the same data found no significant variants near this gene. In genome-wide association studies, variant-level associations are hard to identify and can be difficult to interpret biologically. Here, the authors develop a new model-based enrichment analysis method, and apply it to identify new associated genes, pathways and tissues across 31 human phenotypes.
Collapse
Affiliation(s)
- Xiang Zhu
- Department of Statistics, Stanford University, Stanford, 94305, CA, USA. .,Department of Statistics, The University of Chicago, Chicago, 60637, IL, USA.
| | - Matthew Stephens
- Department of Statistics, The University of Chicago, Chicago, 60637, IL, USA. .,Department of Human Genetics, The University of Chicago, Chicago, 60637, IL, USA.
| |
Collapse
|
170
|
Dizier MH, Margaritte-Jeannin P, Pain L, Sarnowski C, Brossard M, Mohamdi H, Lavielle N, Babron MCC, Just J, Lathrop M, Laprise C, Bouzigon E, Demenais F, Nadif R. Interactive effect between ATPase-related genes and early-life tobacco smoke exposure on bronchial hyper-responsiveness detected in asthma-ascertained families. Thorax 2018; 74:254-260. [PMID: 30282721 DOI: 10.1136/thoraxjnl-2018-211797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/21/2018] [Accepted: 08/20/2018] [Indexed: 01/29/2023]
Abstract
BACKGROUND A positional cloning study of bronchial hyper-responsiveness (BHR) at the 17p11 locus in the French Epidemiological study on the Genetics and Environment of Asthma (EGEA) families showed significant interaction between early-life environmental tobacco smoke (ETS) exposure and genetic variants located in DNAH9. This gene encodes the heavy chain subunit of axonemal dynein, which is involved with ATP in the motile cilia function.Our goal was to identify genetic variants at other genes interacting with ETS in BHR by investigating all genes belonging to the 'ATP-binding' and 'ATPase activity' pathways which include DNAH9, are targets of cigarette smoke and play a crucial role in the airway inflammation. METHODS Family-based interaction tests between ETS-exposed and unexposed BHR siblings were conducted in 388 EGEA families. Twenty single-nucleotide polymorphisms (SNP) showing interaction signals (p≤5.10-3) were tested in the 253 Saguenay-Lac-Saint-Jean (SLSJ) families. RESULTS One of these SNPs was significantly replicated for interaction with ETS in SLSJ families (p=0.003). Another SNP reached the significance threshold after correction for multiple testing in the combined analysis of the two samples (p=10-5). Results were confirmed using both a robust log-linear test and a gene-based interaction test. CONCLUSION The SNPs showing interaction with ETS belong to the ATP8A1 and ABCA1 genes, which play a role in the maintenance of asymmetry and homeostasis of lung membrane lipids.
Collapse
Affiliation(s)
- Marie-Hélène Dizier
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Patricia Margaritte-Jeannin
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Lucile Pain
- Département des Sciences Fondamentales, Université du Québec, Chicoutimi, Quebec, Canada
| | - Chloé Sarnowski
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Myriam Brossard
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Hamida Mohamdi
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Nolwenn Lavielle
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Marie-Claude C Babron
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Jocelyne Just
- Service d'Allergologie Pédiatrique, Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand-Trousseau (APHP), UPMC Paris 06, Paris, France
| | - Mark Lathrop
- Department of Human Genetics, McGill University and Genome Quebec's Innovation Centre, Montréal, Québec, Canada
| | - Catherine Laprise
- Département des Sciences Fondamentales, Université du Québec, Chicoutimi, Quebec, Canada
| | - Emmanuelle Bouzigon
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Florence Demenais
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Rachel Nadif
- Aging and Chronic Diseases-Epidemiological and Public Health Approaches (VIMA), Inserm, U1168, Villejuif, France.,UMR-S 1168, Université de Versailles Saint-Quentin-en-Yvelines, Paris, France
| |
Collapse
|
171
|
Bonham LW, Steele NZR, Karch CM, Manzoni C, Geier EG, Wen N, Ofori-Kuragu A, Momeni P, Hardy J, Miller ZA, Hess CP, Lewis P, Miller BL, Seeley WW, Baranzini SE, Desikan RS, Ferrari R, Yokoyama JS. Protein network analysis reveals selectively vulnerable regions and biological processes in FTD. Neurol Genet 2018; 4:e266. [PMID: 30283816 PMCID: PMC6167176 DOI: 10.1212/nxg.0000000000000266] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/31/2018] [Indexed: 11/15/2022]
Abstract
OBJECTIVE The neuroanatomical profile of behavioral variant frontotemporal dementia (bvFTD) suggests a common biological etiology of disease despite disparate pathologic causes; we investigated the genetic underpinnings of this selective regional vulnerability to identify new risk factors for bvFTD. METHODS We used recently developed analytical techniques designed to address the limitations of genome-wide association studies to generate a protein interaction network of 63 bvFTD risk genes. We characterized this network using gene expression data from healthy and diseased human brain tissue, evaluating regional network expression patterns across the lifespan as well as the cell types and biological processes most affected in bvFTD. RESULTS We found that bvFTD network genes show enriched expression across the human lifespan in vulnerable neuronal populations, are implicated in cell signaling, cell cycle, immune function, and development, and are differentially expressed in pathologically confirmed frontotemporal lobar degeneration cases. Five of the genes highlighted by our differential expression analyses, BAIAP2, ERBB3, POU2F2, SMARCA2, and CDC37, appear to be novel bvFTD risk loci. CONCLUSIONS Our findings suggest that the cumulative burden of common genetic variation in an interacting protein network expressed in specific brain regions across the lifespan may influence susceptibility to bvFTD.
Collapse
Affiliation(s)
- Luke W Bonham
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Natasha Z R Steele
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Celeste M Karch
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Claudia Manzoni
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Ethan G Geier
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Natalie Wen
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Aaron Ofori-Kuragu
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Parastoo Momeni
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - John Hardy
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Zachary A Miller
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Christopher P Hess
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Patrick Lewis
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Bruce L Miller
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - William W Seeley
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Sergio E Baranzini
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Rahul S Desikan
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Raffaele Ferrari
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| | - Jennifer S Yokoyama
- Department of Neurology (L.W.B., N.Z.R.S., E.G.G., Z.A.M., B.L.M., W.W.S., J.S.Y.), Memory and Aging Center, University of California, San Francisco; Department of Psychiatry (C.M.K., N.W., A.O.-K.), Washington University, St. Louis, MO; School of Pharmacy (C.M., P.L.), University of Reading, Reading, UK; Laboratory of Neurogenetics (P.M.), Texas Tech University Health Science Center, Lubbock; Department of Molecular Neuroscience (C.M., J.H., P.L., R.F.), UCL Institute of Neurology, London, UK; Department of Radiology and Biomedical Imaging, Neuroradiology Section (C.P.H., R.S.D.), University of California, San Francisco; and Department of Neurology (S.E.B.), University of California, San Francisco
| |
Collapse
|
172
|
Abstract
SIGNIFICANCE Reductionist studies have contributed greatly to our understanding of the basic biology of aging in recent years but we still do not understand fundamental mechanisms for many identified drugs and pathways. Use of systems approaches will help us move forward in our understanding of aging. Recent Advances: Recent work described here has illustrated the power of systems biology to inform our understanding of aging through the study of (i) diet restriction, (ii) neurodegenerative disease, and (iii) biomarkers of aging. CRITICAL ISSUES Although we do not understand all of the individual genes and pathways that affect aging, as we continue to uncover more of them, we have now also begun to synthesize existing data using systems-level approaches, often to great effect. The three examples noted here all benefit from computational approaches that were unknown a few years ago, and from biological insights gleaned from multiple model systems, from aging laboratories as well as many other areas of biology. FUTURE DIRECTIONS Many new technologies, such as single-cell sequencing, advances in epigenetics beyond the methylome (specifically, assay for transposase-accessible chromatin with high throughput sequencing ), and multiomic network studies, will increase the reach of systems biologists. This suggests that approaches similar to those described here will continue to lead to striking findings, and to interventions that may allow us to delay some of the many age-associated diseases in humans; perhaps sooner that we expect. Antioxid. Redox Signal. 29, 973-984.
Collapse
Affiliation(s)
| | - Daniel E L Promislow
- 2 Department of Pathology, University of Washington , Seattle, Washington.,3 Department of Biology, University of Washington , Seattle, Washington
| |
Collapse
|
173
|
Chang X, Lima LDA, Liu Y, Li J, Li Q, Sleiman PMA, Hakonarson H. Common and Rare Genetic Risk Factors Converge in Protein Interaction Networks Underlying Schizophrenia. Front Genet 2018; 9:434. [PMID: 30323833 PMCID: PMC6172705 DOI: 10.3389/fgene.2018.00434] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 09/12/2018] [Indexed: 11/25/2022] Open
Abstract
Hundreds of genomic loci have been identified with the recent advances of schizophrenia in genome-wide association studies (GWAS) and sequencing studies. However, the functional interactions among those genes remain largely unknown. We developed a network-based approach to integrate multiple genetic risk factors, which lead to the discovery of new susceptibility genes and causal sub-networks, or pathways in schizophrenia. We identified significantly and consistently over-represented pathways in the largest schizophrenia GWA studies, which are highly relevant to synaptic plasticity, neural development and signaling transduction, such as long-term potentiation, neurotrophin signaling pathway, and the ERBB signaling pathway. We also demonstrated that genes targeted by common SNPs are more likely to interact with genes harboring de novo mutations (DNMs) in the protein-protein interaction (PPI) network, suggesting a mutual interplay of both common and rare variants in schizophrenia. We further developed an edge-based search algorithm to identify the top-ranked gene modules associated with schizophrenia risk. Our results suggest that the N-methyl-D-aspartate receptor (NMDAR) interactome may play a leading role in the pathology of schizophrenia, as it is highly targeted by multiple types of genetic risk factors.
Collapse
Affiliation(s)
- Xiao Chang
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Leandro de Araujo Lima
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Yichuan Liu
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jin Li
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States.,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Qingqin Li
- Janssen Research & Development, LLC, Titusville, NJ, United States
| | - Patrick M A Sleiman
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
174
|
Sharma A, Kitsak M, Cho MH, Ameli A, Zhou X, Jiang Z, Crapo JD, Beaty TH, Menche J, Bakke PS, Santolini M, Silverman EK. Integration of Molecular Interactome and Targeted Interaction Analysis to Identify a COPD Disease Network Module. Sci Rep 2018; 8:14439. [PMID: 30262855 PMCID: PMC6160419 DOI: 10.1038/s41598-018-32173-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 08/20/2018] [Indexed: 12/21/2022] Open
Abstract
The polygenic nature of complex diseases offers potential opportunities to utilize network-based approaches that leverage the comprehensive set of protein-protein interactions (the human interactome) to identify new genes of interest and relevant biological pathways. However, the incompleteness of the current human interactome prevents it from reaching its full potential to extract network-based knowledge from gene discovery efforts, such as genome-wide association studies, for complex diseases like chronic obstructive pulmonary disease (COPD). Here, we provide a framework that integrates the existing human interactome information with experimental protein-protein interaction data for FAM13A, one of the most highly associated genetic loci to COPD, to find a more comprehensive disease network module. We identified an initial disease network neighborhood by applying a random-walk method. Next, we developed a network-based closeness approach (CAB) that revealed 9 out of 96 FAM13A interacting partners identified by affinity purification assays were significantly close to the initial network neighborhood. Moreover, compared to a similar method (local radiality), the CAB approach predicts low-degree genes as potential candidates. The candidates identified by the network-based closeness approach were combined with the initial network neighborhood to build a comprehensive disease network module (163 genes) that was enriched with genes differentially expressed between controls and COPD subjects in alveolar macrophages, lung tissue, sputum, blood, and bronchial brushing datasets. Overall, we demonstrate an approach to find disease-related network components using new laboratory data to overcome incompleteness of the current interactome.
Collapse
Affiliation(s)
- Amitabh Sharma
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA. .,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA. .,Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, 02115, USA. .,Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA.
| | - Maksim Kitsak
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, 02115, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA.,Pulmonary and Critical Care Division, Brigham and Women's Hospital and Harvard Medical School, Boston, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Asher Ameli
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA.,Department of Physics, Northeastern University, Boston, MA, 02115, United States
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Zhiqiang Jiang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA
| | - James D Crapo
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Terri H Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jörg Menche
- Department of Bioinformatics, CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, A-1090, Vienna, Austria
| | - Per S Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marc Santolini
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA.,Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, 02115, USA.,Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA. .,Pulmonary and Critical Care Division, Brigham and Women's Hospital and Harvard Medical School, Boston, USA. .,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
175
|
Takahashi KH, Ishimori M, Iwata H. HSP90 as a global genetic modifier for male genital morphology in Drosophila melanogaster. Evolution 2018; 72:2419-2434. [PMID: 30221481 DOI: 10.1111/evo.13598] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 09/03/2018] [Indexed: 02/06/2023]
Abstract
The molecular chaperone protein HSP90 has been proposed to modulate genotype-phenotype relationship in a broad range of organisms. We explore the proposed genetic modifier effect of HSP90 through a genomewide analysis. Here, we show that HSP90 functions as a genetic modifier of genital morphology in Drosophila melanogaster. We identified a large number of single-nucleotide polymorphisms (SNPs) with an HSP90-dependent effect by using genome wide association analysis. We classified the SNPs into the ones under capacitance effect (smaller allelic effect under HSP90 inhibition) or the ones under potentiation effect (larger allelic effect under HSP90 inhibition). Although the majority of SNPs are under capacitance, there are a large number of SNPs under potentiation. This observation provides support for a model in which Hsp90 is not described exclusively as a "genetic capacitor," but is described more broadly as a "genetic modifier." Because the majority of the candidate genes estimated from SNPs with an HSP90-dependent effect in the current study has never been reported to interact with HSP90 directly, the global genetic modifier effect of HSP90 may be exhibited through epistatic interactions in gene regulatory networks.
Collapse
Affiliation(s)
- Kazuo H Takahashi
- Graduate School of Environmental and Life Science, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-si, Okayama-ken, 700-8530, Japan
| | - Motoyuki Ishimori
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo, Tokyo 113-8657, Japan
| | - Hiroyoshi Iwata
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo, Tokyo 113-8657, Japan
| |
Collapse
|
176
|
Genomic Analyses of Visual Cognition: Perceptual Rivalry and Top-Down Control. J Neurosci 2018; 38:9668-9678. [PMID: 30242048 DOI: 10.1523/jneurosci.1970-17.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 11/21/2022] Open
Abstract
Visual cognition in humans has traditionally been studied with cognitive behavioral methods and brain imaging, but much less with genetic methods. Perceptual rivalry, an important phenomenon in visual cognition, is the spontaneous perceptual alternation that occurs between two distinct interpretations of a physically constant visual stimulus (e.g., binocular rivalry stimuli) or a perceptually ambiguous stimulus (e.g., the Necker cube). The switching rate varies dramatically across individuals and can be voluntarily modulated by observers. Here, we adopted a genomic approach to systematically investigate the genetics underlying binocular rivalry, Necker cube rivalry and voluntary modulation of Necker cube rivalry in young Chinese adults (Homo sapiens, 81% female, 20 ± 1 years old) at multiple levels, including common single nucleotide polymorphism (SNP)-based heritability estimation, SNP-based genome-wide association study (GWAS), gene-based analysis, and pathway analysis. We performed a pilot GWAS in 2441 individuals and replicated it in an independent cohort of 943 individuals. Common SNP-based heritability was estimated to be 25% for spontaneous perceptual rivalry. SNPs rs184765639 and rs75595941 were associated with voluntary modulation, and imaging data suggested genotypic difference of rs184765639 in the surface area of the left caudal-middle frontal cortex. Additionally, converging evidence from multilevel analyses associated genes such as PRMT1 with perceptual switching rate, and MIR1178 with voluntary modulation strength. In summary, this study discovered specific genetic contributions to perceptual rivalry and its voluntary modulation in human beings. These findings may promote our understanding of psychiatric disorders, as perceptual rivalry is a potential psychiatric biomarker.SIGNIFICANCE STATEMENT Perceptual rivalry is an important visual phenomenon in which our perception of a physically constant visual input spontaneously switches between two different states. There are individual variations in perceptual switching rate and voluntary modulation strength. Our genomic analyses reveal several loci associated with these two kinds of variation. Because perceptual rivalry is thought to be relevant to and potentially an endophenotype for psychiatric disorders, these results may help understand not only visual cognition, but also psychiatric disorders.
Collapse
|
177
|
Cui P, Ma X, Li H, Lang W, Hao J. Shared Biological Pathways Between Alzheimer's Disease and Ischemic Stroke. Front Neurosci 2018; 12:605. [PMID: 30245614 PMCID: PMC6137293 DOI: 10.3389/fnins.2018.00605] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/10/2018] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) and ischemic stroke (IS) are an immense socioeconomic burden worldwide. There is a possibility that shared genetic factors lead to their links at epidemiological and pathophysiological levels. Although recent genome-wide association studies (GWAS) have provided profound insights into the genetics of AD and IS, no shared genetic variants have been identified to date. This prompted us to initiate this study, which sought to identify shared pathways linking AD and IS. We took advantage of large-scale GWAS summary data of AD (17,008 AD cases and 37,154 controls) and IS (10,307 cases and 19,326 controls) to conduct pathway analyses using genetic pathways from multiple well-studied databases, including GO, KEGG, PANTHER, Reactome, and Wikipathways. Collectively, we discovered that AD and IS shared 179 GO categories (56 biological processes, 95 cellular components, and 28 molecular functions); and the following pathways: six KEGG pathways; two PANTHER pathways; four Reactome pathways; and one in Wikipathways pathway. The more fine-grained GO terms were mainly summarized into different functional categories: transcriptional and post-transcriptional regulation, synapse, endocytic membrane traffic through the endosomal system, signaling transduction, immune process, multi-organism process, protein catabolic metabolism, and cell adhesion. The shared pathways were roughly classified into three categories: immune system; cancer (NSCLC and glioma); and signal transduction pathways involving the cadherin signaling pathway, Wnt signaling pathway, G-protein signaling and downstream signaling mediated by phosphoinositides (PIPs). The majority of these common pathways linked to both AD and IS were supported by convincing evidence from the literature. In conclusion, our findings contribute to a better understanding of common biological mechanisms underlying AD and IS and serve as a guide to direct future research.
Collapse
Affiliation(s)
- Pan Cui
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin, China
| | - Xiaofeng Ma
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin, China
| | - He Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin, China
| | - Wenjing Lang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin, China
| | - Junwei Hao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin, China
| |
Collapse
|
178
|
Cai Z, Guldbrandtsen B, Lund MS, Sahana G. Prioritizing candidate genes post-GWAS using multiple sources of data for mastitis resistance in dairy cattle. BMC Genomics 2018; 19:656. [PMID: 30189836 PMCID: PMC6127918 DOI: 10.1186/s12864-018-5050-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/31/2018] [Indexed: 12/31/2022] Open
Abstract
Background Improving resistance to mastitis, one of the costliest diseases in dairy production, has become an important objective in dairy cattle breeding. However, mastitis resistance is influenced by many genes involved in multiple processes, including the response to infection, inflammation, and post-infection healing. Low genetic heritability, environmental variations, and farm management differences further complicate the identification of links between genetic variants and mastitis resistance. Consequently, studies of the genetics of variation in mastitis resistance in dairy cattle lack agreement about the responsible genes. Results We associated 15,552,968 imputed whole-genome sequencing markers for 5147 Nordic Holstein cattle with mastitis resistance in a genome-wide association study (GWAS). Next, we augmented P-values for markers in genes in the associated regions using Gene Ontology terms, Kyoto Encyclopedia of Genes and Genomes pathway analysis, and mammalian phenotype database. To confirm results of gene-based analyses, we used gene expression data from E. coli-challenged cow udders. We identified 22 independent quantitative trait loci (QTL) that collectively explained 14% of the variance in breeding values for resistance to clinical mastitis (CM). Using association test statistics with multiple pieces of independent information on gene function and differential expression during bacterial infection, we suggested putative causal genes with biological relevance for 12 QTL affecting resistance to CM in dairy cattle. Conclusion Combining information on the nearest positional genes, gene-based analyses, and differential gene expression data from RNA-seq, we identified putative causal genes (candidate genes with biological evidence) in QTL for mastitis resistance in Nordic Holstein cattle. The same strategy can be applied for other traits. Electronic supplementary material The online version of this article (10.1186/s12864-018-5050-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zexi Cai
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830, Tjele, Denmark.
| | - Bernt Guldbrandtsen
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830, Tjele, Denmark
| | - Mogens Sandø Lund
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830, Tjele, Denmark
| | - Goutam Sahana
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830, Tjele, Denmark
| |
Collapse
|
179
|
Chen YC, Xu C, Zhang JG, Zeng CP, Wang XF, Zhou R, Lin X, Ao ZX, Lu JM, Shen J, Deng HW. Multivariate analysis of genomics data to identify potential pleiotropic genes for type 2 diabetes, obesity and dyslipidemia using Meta-CCA and gene-based approach. PLoS One 2018; 13:e0201173. [PMID: 30110382 PMCID: PMC6093635 DOI: 10.1371/journal.pone.0201173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/10/2018] [Indexed: 11/19/2022] Open
Abstract
Previous studies have demonstrated the genetic correlations between type 2 diabetes, obesity and dyslipidemia, and indicated that many genes have pleiotropic effects on them. However, these pleiotropic genes have not been well-defined. It is essential to identify pleiotropic genes using systematic approaches because systematically analyzing correlated traits is an effective way to enhance their statistical power. To identify potential pleiotropic genes for these three disorders, we performed a systematic analysis by incorporating GWAS (genome-wide associated study) datasets of six correlated traits related to type 2 diabetes, obesity and dyslipidemia using Meta-CCA (meta-analysis using canonical correlation analysis). Meta-CCA is an emerging method to systematically identify potential pleiotropic genes using GWAS summary statistics of multiple correlated traits. 2,720 genes were identified as significant genes after multiple testing (Bonferroni corrected p value < 0.05). Further, to refine the identified genes, we tested their relationship to the six correlated traits using VEGAS-2 (versatile gene-based association study-2). Only the genes significantly associated (Bonferroni corrected p value < 0.05) with more than one trait were kept. Finally, 25 genes (including two confirmed pleiotropic genes and eleven novel pleiotropic genes) were identified as potential pleiotropic genes. They were enriched in 5 pathways including the statin pathway and the PPAR (peroxisome proliferator-activated receptor) Alpha pathway. In summary, our study identified potential pleiotropic genes and pathways of type 2 diabetes, obesity and dyslipidemia, which may shed light on the common biological etiology and pathogenesis of these three disorders and provide promising insights for new therapies.
Collapse
Affiliation(s)
- Yuan-Cheng Chen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, GuangDong, PR China
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Chao Xu
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Ji-Gang Zhang
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Chun-Ping Zeng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, GuangDong, PR China
| | - Xia-Fang Wang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, GuangDong, PR China
| | - Rou Zhou
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, GuangDong, PR China
| | - Xu Lin
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, GuangDong, PR China
| | - Zeng-Xin Ao
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, GuangDong, PR China
| | - Jun-Min Lu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, GuangDong, PR China
| | - Jie Shen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, GuangDong, PR China
| | - Hong-Wen Deng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, GuangDong, PR China
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
- School of Basic Medical Sciences, Central South University, Changsha, HuNan, PR China
| |
Collapse
|
180
|
Gossmann A, Zille P, Calhoun V, Wang YP. FDR-Corrected Sparse Canonical Correlation Analysis With Applications to Imaging Genomics. IEEE TRANSACTIONS ON MEDICAL IMAGING 2018; 37:1761-1774. [PMID: 29993802 DOI: 10.1109/tmi.2018.2815583] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Reducing the number of false discoveries is presently one of the most pressing issues in the life sciences. It is of especially great importance for many applications in neuroimaging and genomics, where data sets are typically high-dimensional, which means that the number of explanatory variables exceeds the sample size. The false discovery rate (FDR) is a criterion that can be employed to address that issue. Thus it has gained great popularity as a tool for testing multiple hypotheses. Canonical correlation analysis (CCA) is a statistical technique that is used to make sense of the cross-correlation of two sets of measurements collected on the same set of samples (e.g., brain imaging and genomic data for the same mental illness patients), and sparse CCA extends the classical method to high-dimensional settings. Here, we propose a way of applying the FDR concept to sparse CCA, and a method to control the FDR. The proposed FDR correction directly influences the sparsity of the solution, adapting it to the unknown true sparsity level. Theoretical derivation as well as simulation studies show that our procedure indeed keeps the FDR of the canonical vectors below a user-specified target level. We apply the proposed method to an imaging genomics data set from the Philadelphia Neurodevelopmental Cohort. Our results link the brain connectivity profiles derived from brain activity during an emotion identification task, as measured by functional magnetic resonance imaging, to the corresponding subjects' genomic data.
Collapse
|
181
|
Gray KJ, Kovacheva VP, Mirzakhani H, Bjonnes AC, Almoguera B, DeWan AT, Triche EW, Saftlas AF, Hoh J, Bodian DL, Klein E, Huddleston KC, Ingles SA, Lockwood CJ, Hakonarson H, McElrath TF, Murray JC, Wilson ML, Norwitz ER, Karumanchi SA, Bateman BT, Keating BJ, Saxena R. Gene-Centric Analysis of Preeclampsia Identifies Maternal Association at PLEKHG1. Hypertension 2018; 72:408-416. [PMID: 29967039 PMCID: PMC6043396 DOI: 10.1161/hypertensionaha.117.10688] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 12/30/2017] [Accepted: 04/14/2018] [Indexed: 12/13/2022]
Abstract
The genetic susceptibility to preeclampsia, a pregnancy-specific complication with significant maternal and fetal morbidity, has been poorly characterized. To identify maternal genes associated with preeclampsia risk, we assembled 498 cases and 1864 controls of European ancestry from preeclampsia case-control collections in 5 different US sites (with additional matched population controls), genotyped samples on a cardiovascular gene-centric array composed of variants from ≈2000 genes selected based on prior genetic studies of cardiovascular and metabolic diseases and performed case-control genetic association analysis on 27 429 variants passing quality control. In silico replication testing of 9 lead signals with P<10-4 was performed in independent European samples from the SOPHIA (Study of Pregnancy Hypertension in Iowa) and Inova cohorts (212 cases, 456 controls). Multiethnic assessment of lead signals was then performed in samples of black (26 cases, 136 controls), Hispanic (132 cases, 468 controls), and East Asian (9 cases, 80 controls) ancestry. Multiethnic meta-analysis (877 cases, 3004 controls) revealed a study-wide statistically significant association of the rs9478812 variant in the pleiotropic PLEKHG1 gene (odds ratio, 1.40 [1.23-1.60]; Pmeta=5.90×10-7). The rs9478812 effect was even stronger in the subset of European cases with known early-onset preeclampsia (236 cases diagnosed <37 weeks, 1864 controls; odds ratio, 1.59 [1.27-1.98]; P=4.01×10-5). PLEKHG1 variants have previously been implicated in genome-wide association studies of blood pressure, body weight, and neurological disorders. Although larger studies are required to further define maternal preeclampsia heritability, this study identifies a novel maternal risk locus for further investigation.
Collapse
Affiliation(s)
- Kathryn J Gray
- From the Division of Maternal-Fetal Medicine (K.J.G., T.F.M.)
- Center for Genomic Medicine (K.J.G., A.C.B., R.S.)
- Massachusetts General Hospital, Boston; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (K.J.G., A.C.B., R.S.)
| | | | - Hooman Mirzakhani
- Brigham and Women's Hospital, Boston, MA; Department of Anesthesia, Critical Care and Pain Medicine (H.M., B.T.B., R.S.)
| | - Andrew C Bjonnes
- Center for Genomic Medicine (K.J.G., A.C.B., R.S.)
- Massachusetts General Hospital, Boston; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (K.J.G., A.C.B., R.S.)
| | - Berta Almoguera
- Center for Applied Genomics, Children's Hospital of Philadelphia, PA (B.A., H.H.)
| | | | - Elizabeth W Triche
- Yale School of Public Health, New Haven, CT; Center for Outcomes Research and Evaluation, Yale School of Medicine, New Haven, CT (E.W.T.)
| | - Audrey F Saftlas
- Department of Epidemiology, College of Public Health, University of Iowa (A.F.S.)
| | | | - Dale L Bodian
- Inova Translational Medicine Institute, Inova Health System, Falls Church, VA (D.L.B., E.K., K.C.H.)
| | - Elisabeth Klein
- Inova Translational Medicine Institute, Inova Health System, Falls Church, VA (D.L.B., E.K., K.C.H.)
| | - Kathi C Huddleston
- Inova Translational Medicine Institute, Inova Health System, Falls Church, VA (D.L.B., E.K., K.C.H.)
| | - Sue Ann Ingles
- Department of Preventative Medicine, University of Southern California, Keck School of Medicine, Los Angeles (S.A.I., M.L.W.)
| | - Charles J Lockwood
- University of South Florida, Morsani College of Medicine, Tampa (C.J.L.)
| | - Hakon Hakonarson
- Divisions of Human Genetics and Pulmonary Medicine, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (H.H.)
| | | | - Jeffrey C Murray
- Department of Pediatrics, Carver College of Medicine, University of Iowa (J.C.M.)
| | - Melissa L Wilson
- Department of Preventative Medicine, University of Southern California, Keck School of Medicine, Los Angeles (S.A.I., M.L.W.)
| | - Errol R Norwitz
- Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA (E.R.N.)
| | - S Ananth Karumanchi
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA (S.A.K.)
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (S.A.K.)
| | - Brian T Bateman
- Brigham and Women's Hospital, Boston, MA; Department of Anesthesia, Critical Care and Pain Medicine (H.M., B.T.B., R.S.)
| | - Brendan J Keating
- Department of Surgery and Pediatrics, University of Pennsylvania, Philadelphia (B.J.K.)
| | - Richa Saxena
- Brigham and Women's Hospital, Boston, MA; Department of Anesthesia, Critical Care and Pain Medicine (H.M., B.T.B., R.S.)
- Center for Genomic Medicine (K.J.G., A.C.B., R.S.)
- Massachusetts General Hospital, Boston; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (K.J.G., A.C.B., R.S.)
| |
Collapse
|
182
|
Flitcroft DI, Loughman J, Wildsoet CF, Williams C, Guggenheim JA. Novel Myopia Genes and Pathways Identified From Syndromic Forms of Myopia. Invest Ophthalmol Vis Sci 2018; 59:338-348. [PMID: 29346494 PMCID: PMC5773233 DOI: 10.1167/iovs.17-22173] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose To test the hypothesis that genes known to cause clinical syndromes featuring myopia also harbor polymorphisms contributing to nonsyndromic refractive errors. Methods Clinical phenotypes and syndromes that have refractive errors as a recognized feature were identified using the Online Mendelian Inheritance in Man (OMIM) database. One hundred fifty-four unique causative genes were identified, of which 119 were specifically linked with myopia and 114 represented syndromic myopia (i.e., myopia and at least one other clinical feature). Myopia was the only refractive error listed for 98 genes and hyperopia and the only refractive error noted for 28 genes, with the remaining 28 genes linked to phenotypes with multiple forms of refractive error. Pathway analysis was carried out to find biological processes overrepresented within these sets of genes. Genetic variants located within 50 kb of the 119 myopia-related genes were evaluated for involvement in refractive error by analysis of summary statistics from genome-wide association studies (GWAS) conducted by the CREAM Consortium and 23andMe, using both single-marker and gene-based tests. Results Pathway analysis identified several biological processes already implicated in refractive error development through prior GWAS analyses and animal studies, including extracellular matrix remodeling, focal adhesion, and axon guidance, supporting the research hypothesis. Novel pathways also implicated in myopia development included mannosylation, glycosylation, lens development, gliogenesis, and Schwann cell differentiation. Hyperopia was found to be linked to a different pattern of biological processes, mostly related to organogenesis. Comparison with GWAS findings further confirmed that syndromic myopia genes were enriched for genetic variants that influence refractive errors in the general population. Gene-based analyses implicated 21 novel candidate myopia genes (ADAMTS18, ADAMTS2, ADAMTSL4, AGK, ALDH18A1, ASXL1, COL4A1, COL9A2, ERBB3, FBN1, GJA1, GNPTG, IFIH1, KIF11, LTBP2, OCA2, POLR3B, POMT1, PTPN11, TFAP2A, ZNF469). Conclusions Common genetic variants within or nearby genes that cause syndromic myopia are enriched for variants that cause nonsyndromic, common myopia. Analysis of syndromic forms of refractive errors can provide new insights into the etiology of myopia and additional potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- D Ian Flitcroft
- Children's University Hospital and University College Dublin, Dublin, Ireland.,College of Sciences and Health, Dublin Institute of Technology, Dublin, Ireland
| | - James Loughman
- College of Sciences and Health, Dublin Institute of Technology, Dublin, Ireland
| | - Christine F Wildsoet
- Center for Eye Disease and Development, School of Optometry, University of California-Berkeley, Berkeley, California, United States
| | - Cathy Williams
- Bristol Eye Hospital and Bristol University, Bristol, United Kingdom
| | - Jeremy A Guggenheim
- School of Optometry & Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | | |
Collapse
|
183
|
Lee JJ, Wedow R, Okbay A, Kong E, Maghzian O, Zacher M, Nguyen-Viet TA, Bowers P, Sidorenko J, Karlsson Linnér R, Fontana MA, Kundu T, Lee C, Li H, Li R, Royer R, Timshel PN, Walters RK, Willoughby EA, Yengo L, Alver M, Bao Y, Clark DW, Day FR, Furlotte NA, Joshi PK, Kemper KE, Kleinman A, Langenberg C, Mägi R, Trampush JW, Verma SS, Wu Y, Lam M, Zhao JH, Zheng Z, Boardman JD, Campbell H, Freese J, Harris KM, Hayward C, Herd P, Kumari M, Lencz T, Luan J, Malhotra AK, Metspalu A, Milani L, Ong KK, Perry JRB, Porteous DJ, Ritchie MD, Smart MC, Smith BH, Tung JY, Wareham NJ, Wilson JF, Beauchamp JP, Conley DC, Esko T, Lehrer SF, Magnusson PKE, Oskarsson S, Pers TH, Robinson MR, Thom K, Watson C, Chabris CF, Meyer MN, Laibson DI, Yang J, Johannesson M, Koellinger PD, Turley P, Visscher PM, Benjamin DJ, Cesarini D. Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nat Genet 2018; 50:1112-1121. [PMID: 30038396 PMCID: PMC6393768 DOI: 10.1038/s41588-018-0147-3] [Citation(s) in RCA: 1449] [Impact Index Per Article: 207.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 04/30/2018] [Indexed: 02/06/2023]
Abstract
Here we conducted a large-scale genetic association analysis of educational attainment in a sample of approximately 1.1 million individuals and identify 1,271 independent genome-wide-significant SNPs. For the SNPs taken together, we found evidence of heterogeneous effects across environments. The SNPs implicate genes involved in brain-development processes and neuron-to-neuron communication. In a separate analysis of the X chromosome, we identify 10 independent genome-wide-significant SNPs and estimate a SNP heritability of around 0.3% in both men and women, consistent with partial dosage compensation. A joint (multi-phenotype) analysis of educational attainment and three related cognitive phenotypes generates polygenic scores that explain 11-13% of the variance in educational attainment and 7-10% of the variance in cognitive performance. This prediction accuracy substantially increases the utility of polygenic scores as tools in research.
Collapse
Affiliation(s)
- James J Lee
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Robbee Wedow
- Department of Sociology, University of Colorado Boulder, Boulder, CO, USA
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Institute of Behavioral Science, University of Colorado Boulder, Boulder, CO, USA
| | - Aysu Okbay
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Department of Economics, School of Business and Economics, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - Edward Kong
- Department of Economics, Harvard University, Cambridge, MA, USA
| | - Omeed Maghzian
- Department of Economics, Harvard University, Cambridge, MA, USA
| | - Meghan Zacher
- Department of Sociology, Harvard University, Cambridge, MA, USA
| | - Tuan Anh Nguyen-Viet
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
| | - Peter Bowers
- Department of Economics, Harvard University, Cambridge, MA, USA
| | - Julia Sidorenko
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Richard Karlsson Linnér
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Economics, School of Business and Economics, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Institute for Behavior and Biology, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Mark Alan Fontana
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
- Center for the Advancement of Value in Musculoskeletal Care, Hospital for Special Surgery, New York, NY, USA
| | - Tushar Kundu
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
| | - Chanwook Lee
- Department of Economics, Harvard University, Cambridge, MA, USA
| | - Hui Li
- Department of Economics, Harvard University, Cambridge, MA, USA
| | - Ruoxi Li
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
| | - Rebecca Royer
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
| | - Pascal N Timshel
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
- Statens Serum Institut, Department of Epidemiology Research, Copenhagen, Denmark
| | - Raymond K Walters
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Emily A Willoughby
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Loïc Yengo
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Maris Alver
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Yanchun Bao
- Institute for Social and Economic Research, University of Essex, Colchester, UK
| | - David W Clark
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Felix R Day
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | | | - Peter K Joshi
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
- Institute of Social and Preventive Medicine, University Hospital of Lausanne, Lausanne, Switzerland
| | - Kathryn E Kemper
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | | | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Joey W Trampush
- BrainWorkup, LLC, Santa Monica, CA, USA
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shefali Setia Verma
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, PA, USA
| | - Yang Wu
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Max Lam
- Institute of Mental Health, Singapore, Singapore
- Genome Institute, Singapore, Singapore
| | - Jing Hua Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Zhili Zheng
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Jason D Boardman
- Department of Sociology, University of Colorado Boulder, Boulder, CO, USA
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Institute of Behavioral Science, University of Colorado Boulder, Boulder, CO, USA
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Jeremy Freese
- Department of Sociology, Stanford University, Stanford, CA, USA
| | - Kathleen Mullan Harris
- Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Pamela Herd
- Institute for Social and Economic Research, University of Essex, Colchester, UK
- La Follette School of Public Affairs, University of Wisconsin-Madison, Madison, WI, USA
| | - Meena Kumari
- Institute for Social and Economic Research, University of Essex, Colchester, UK
| | - Todd Lencz
- Departments of Psychiatry and Molecular Medicine, Hofstra Northwell School of Medicine, Hempstead, NY, USA
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, NY, USA
- Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, CA, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Anil K Malhotra
- Departments of Psychiatry and Molecular Medicine, Hofstra Northwell School of Medicine, Hempstead, NY, USA
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, NY, USA
- Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, CA, USA
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Ken K Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - John R B Perry
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Marylyn D Ritchie
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, PA, USA
| | - Melissa C Smart
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Blair H Smith
- Division of Population Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Medical Research Institute, University of Dundee, Dundee, UK
| | | | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - James F Wilson
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | | | - Dalton C Conley
- Department of Sociology, Princeton University, Princeton, NJ, USA
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Steven F Lehrer
- School of Policy Studies, Queen's University, Kingston, Ontario, Canada
- Department of Economics, New York University Shanghai, Pudong, Shanghai, China
- National Bureau of Economic Research, Cambridge, MA, USA
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sven Oskarsson
- Department of Government, Uppsala University, Uppsala, Sweden
| | - Tune H Pers
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
- Statens Serum Institut, Department of Epidemiology Research, Copenhagen, Denmark
| | - Matthew R Robinson
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Kevin Thom
- Department of Economics, New York University, New York, NY, USA
| | - Chelsea Watson
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
| | - Christopher F Chabris
- Autism and Developmental Medicine Institute, Geisinger Health System, Lewisburg, PA, USA
| | - Michelle N Meyer
- Center for Translational Bioethics and Health Care Policy, Geisinger Health System, Danville, PA, USA
| | - David I Laibson
- Department of Economics, Harvard University, Cambridge, MA, USA
| | - Jian Yang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Magnus Johannesson
- Department of Economics, Stockholm School of Economics, Stockholm, Sweden
| | - Philipp D Koellinger
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Economics, School of Business and Economics, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Institute for Behavior and Biology, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Patrick Turley
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Peter M Visscher
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
| | - Daniel J Benjamin
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA.
- National Bureau of Economic Research, Cambridge, MA, USA.
- Department of Economics, University of Southern California, Los Angeles, CA, USA.
| | - David Cesarini
- National Bureau of Economic Research, Cambridge, MA, USA
- Department of Economics, New York University, New York, NY, USA
- Center for Experimental Social Science, New York University, New York, NY, USA
| |
Collapse
|
184
|
Pemberton TJ, Verdu P, Becker NS, Willer CJ, Hewlett BS, Le Bomin S, Froment A, Rosenberg NA, Heyer E. A genome scan for genes underlying adult body size differences between Central African hunter-gatherers and farmers. Hum Genet 2018; 137:487-509. [PMID: 30008065 DOI: 10.1007/s00439-018-1902-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 07/03/2018] [Indexed: 12/16/2022]
Abstract
The evolutionary and biological bases of the Central African "pygmy" phenotype, a characteristic of rainforest hunter-gatherers defined by reduced body size compared with neighboring farmers, remain largely unknown. Here, we perform a joint investigation in Central African hunter-gatherers and farmers of adult standing height, sitting height, leg length, and body mass index (BMI), considering 358 hunter-gatherers and 169 farmers with genotypes for 153,798 SNPs. In addition to reduced standing heights, hunter-gatherers have shorter sitting heights and leg lengths and higher sitting/standing height ratios than farmers and lower BMI for males. Standing height, sitting height, and leg length are strongly correlated with inferred levels of farmer genetic ancestry, whereas BMI is only weakly correlated, perhaps reflecting greater contributions of non-genetic factors to body weight than to height. Single- and multi-marker association tests identify one region and eight genes associated with hunter-gatherer/farmer status, and 24 genes associated with the height-related traits. Many of these genes have putative functions consistent with roles in determining their associated traits and the pygmy phenotype, and they include three associated with standing height in non-Africans (PRKG1, DSCAM, MAGI2). We find evidence that European height-associated SNPs or variants in linkage disequilibrium with them contribute to standing- and sitting-height determination in Central Africans, but not to the differential status of hunter-gatherers and farmers. These findings provide new insights into the biological basis of the pygmy phenotype, and they highlight the potential of cross-population studies for exploring the genetic basis of phenotypes that vary naturally across populations.
Collapse
Affiliation(s)
- Trevor J Pemberton
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.
| | - Paul Verdu
- CNRS-MNHN-Université Paris Diderot, UMR 7206 Eco-Anthropologie et Ethnobiologie, Paris, France.
| | - Noémie S Becker
- Division of Evolutionary Biology, Faculty of Biology, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Cristen J Willer
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Barry S Hewlett
- Department of Anthropology, Washington State University, Vancouver, WA, USA
| | - Sylvie Le Bomin
- CNRS-MNHN-Université Paris Diderot, UMR 7206 Eco-Anthropologie et Ethnobiologie, Paris, France
| | | | | | - Evelyne Heyer
- CNRS-MNHN-Université Paris Diderot, UMR 7206 Eco-Anthropologie et Ethnobiologie, Paris, France.
| |
Collapse
|
185
|
Pierzynski JA, Ye Y, Lippman SM, Rodriguez MA, Wu X, Hildebrandt MAT. Socio-demographic, Clinical, and Genetic Determinants of Quality of Life in Lung Cancer Patients. Sci Rep 2018; 8:10640. [PMID: 30006595 PMCID: PMC6045646 DOI: 10.1038/s41598-018-25712-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 01/12/2018] [Indexed: 12/15/2022] Open
Abstract
Patient reported health-related quality of life (QOL) is a major component of the overall well-being of cancer patients, with links to prognosis. In 6,420 lung cancer patients, we identified patient characteristics and genetic determinants of QOL. Patient responses from the SF-12 questionnaire was used to calculate normalized Physical Component Summary (PCS) and Mental Component Summary (MCS) scores. Further, we analyzed 218 single nucleotide polymorphisms (SNPs) in the p38 MAPK signaling pathway, a key mediator of response to cellular and environmental stress, as genetic determinants of QOL in a subset of the study population (N = 641). Trends among demographic factors for mean PCS and MCS included smoking status (PCS Ptrend < 0.001, MCS Ptrend < 0.001) and education (PCS Ptrend < 0.001, MCS Ptrend < 0.001). Similar relationships were seen for MCS. The homozygous rare genotype of MEF2B: rs2040562 showed an increased risk of a poor MCS (OR: 3.06, 95% CI: 1.05–8.92, P = 0.041). Finally, survival analysis showed that a low PCS or a MCS was associated with increased risks of five-year mortality (HR = 1.63, 95% CI: 1.51–1.77, HR = 1.23, 95% CI: 1.16–1.32, respectively) and there was a significant reduction in median survival time (Plog-rank < 0.001). These findings suggest that multiple factors contribute to QOL in lung cancer patients, and baseline QOL can impact survival.
Collapse
Affiliation(s)
- Jeanne A Pierzynski
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yuanqing Ye
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Scott M Lippman
- Department of Medicine, University of California at San Diego Moores Cancer Center, La Jolla, California, USA
| | - Maria A Rodriguez
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Michelle A T Hildebrandt
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
186
|
Readhead B, Haure-Mirande JV, Funk CC, Richards MA, Shannon P, Haroutunian V, Sano M, Liang WS, Beckmann ND, Price ND, Reiman EM, Schadt EE, Ehrlich ME, Gandy S, Dudley JT. Multiscale Analysis of Independent Alzheimer's Cohorts Finds Disruption of Molecular, Genetic, and Clinical Networks by Human Herpesvirus. Neuron 2018; 99:64-82.e7. [PMID: 29937276 PMCID: PMC6551233 DOI: 10.1016/j.neuron.2018.05.023] [Citation(s) in RCA: 476] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/05/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022]
Abstract
Investigators have long suspected that pathogenic microbes might contribute to the onset and progression of Alzheimer's disease (AD) although definitive evidence has not been presented. Whether such findings represent a causal contribution, or reflect opportunistic passengers of neurodegeneration, is also difficult to resolve. We constructed multiscale networks of the late-onset AD-associated virome, integrating genomic, transcriptomic, proteomic, and histopathological data across four brain regions from human post-mortem tissue. We observed increased human herpesvirus 6A (HHV-6A) and human herpesvirus 7 (HHV-7) from subjects with AD compared with controls. These results were replicated in two additional, independent and geographically dispersed cohorts. We observed regulatory relationships linking viral abundance and modulators of APP metabolism, including induction of APBB2, APPBP2, BIN1, BACE1, CLU, PICALM, and PSEN1 by HHV-6A. This study elucidates networks linking molecular, clinical, and neuropathological features with viral activity and is consistent with viral activity constituting a general feature of AD.
Collapse
Affiliation(s)
- Ben Readhead
- Departments of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute of Genomic Sciences and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Institute for Next Generation Healthcare, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ 85287-5001, USA
| | - Jean-Vianney Haure-Mirande
- Department of Neurology, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Cory C Funk
- Institute for Systems Biology, Seattle, WA, 98109-5263, USA
| | | | - Paul Shannon
- Institute for Systems Biology, Seattle, WA, 98109-5263, USA
| | - Vahram Haroutunian
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; James J. Peters VA Medical Center, 130 West Kingsbridge Road, New York, NY 10468, USA
| | - Mary Sano
- James J. Peters VA Medical Center, 130 West Kingsbridge Road, New York, NY 10468, USA; Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Winnie S Liang
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Noam D Beckmann
- Departments of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute of Genomic Sciences and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nathan D Price
- Institute for Systems Biology, Seattle, WA, 98109-5263, USA
| | - Eric M Reiman
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA; Department of Psychiatry, University of Arizona, Phoenix, AZ 85721, USA; Banner Alzheimer's Institute, Phoenix, AZ 85006, USA
| | - Eric E Schadt
- Departments of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute of Genomic Sciences and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Sema4, Stamford, CT 06902, USA
| | - Michelle E Ehrlich
- Departments of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute of Genomic Sciences and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neurology, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sam Gandy
- Department of Neurology, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; James J. Peters VA Medical Center, 130 West Kingsbridge Road, New York, NY 10468, USA; Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for NFL Neurological Care, Department of Neurology, New York, NY 10029, USA
| | - Joel T Dudley
- Departments of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute of Genomic Sciences and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Institute for Next Generation Healthcare, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ 85287-5001, USA.
| |
Collapse
|
187
|
Multi-level genomic analyses suggest new genetic variants involved in human memory. Eur J Hum Genet 2018; 26:1668-1678. [PMID: 29970928 DOI: 10.1038/s41431-018-0201-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 05/05/2018] [Accepted: 05/22/2018] [Indexed: 12/29/2022] Open
Abstract
Development of high-throughput genotyping platforms provides an opportunity to identify new genetic elements related to complex cognitive functions. Taking advantage of multi-level genomic analysis, here we studied the genetic basis of human short-term (STM, n = 1623) and long-term (LTM, n = 1522) memory functions. Heritability estimation based on single nucleotide polymorphism showed moderate (61%, standard error 35%) heritability of short-term memory but almost zero heritability of long-term memory. We further performed a two-step genome-wide association study, but failed to find any SNPs that could pass genome-wide significance and survive replication at the same time. However, suggestive significance for rs7011450 was found in the shared component of the two STM tasks. Further inspections on its nearby gene zinc finger and at-hook domain containing and SNPs around this gene showed suggestive association with STM. In LTM, a polymorphism within branched chain amino acid transaminase 2 showed suggestive significance in the discovery cohort and has been replicated in another independent population of 1862. Furthermore, we performed a pathway analysis based on the current genomic data and found pathways including mTOR signaling and axon guidance significantly associated with STM capacity. These findings warrant further replication in other larger populations.
Collapse
|
188
|
Sharma A, Halu A, Decano JL, Padi M, Liu YY, Prasad RB, Fadista J, Santolini M, Menche J, Weiss ST, Vidal M, Silverman EK, Aikawa M, Barabási AL, Groop L, Loscalzo J. Controllability in an islet specific regulatory network identifies the transcriptional factor NFATC4, which regulates Type 2 Diabetes associated genes. NPJ Syst Biol Appl 2018; 4:25. [PMID: 29977601 PMCID: PMC6028434 DOI: 10.1038/s41540-018-0057-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 04/09/2018] [Accepted: 05/04/2018] [Indexed: 01/14/2023] Open
Abstract
Probing the dynamic control features of biological networks represents a new frontier in capturing the dysregulated pathways in complex diseases. Here, using patient samples obtained from a pancreatic islet transplantation program, we constructed a tissue-specific gene regulatory network and used the control centrality (Cc) concept to identify the high control centrality (HiCc) pathways, which might serve as key pathobiological pathways for Type 2 Diabetes (T2D). We found that HiCc pathway genes were significantly enriched with modest GWAS p-values in the DIAbetes Genetics Replication And Meta-analysis (DIAGRAM) study. We identified variants regulating gene expression (expression quantitative loci, eQTL) of HiCc pathway genes in islet samples. These eQTL genes showed higher levels of differential expression compared to non-eQTL genes in low, medium, and high glucose concentrations in rat islets. Among genes with highly significant eQTL evidence, NFATC4 belonged to four HiCc pathways. We asked if the expressions of T2D-associated candidate genes from GWAS and literature are regulated by Nfatc4 in rat islets. Extensive in vitro silencing of Nfatc4 in rat islet cells displayed reduced expression of 16, and increased expression of four putative downstream T2D genes. Overall, our approach uncovers the mechanistic connection of NFATC4 with downstream targets including a previously unknown one, TCF7L2, and establishes the HiCc pathways' relationship to T2D.
Collapse
Affiliation(s)
- Amitabh Sharma
- 1Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA.,2Center for Complex Network Research and Department of Physics, Northeastern University, Boston, MA 02115 USA.,3Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215 USA.,4Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215 USA
| | - Arda Halu
- 1Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA.,4Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215 USA
| | - Julius L Decano
- 4Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215 USA
| | - Megha Padi
- 5Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721 USA
| | - Yang-Yu Liu
- 1Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Rashmi B Prasad
- 6Lund University Diabetes Center, Department of Clinical Sciences, Diabetes & Endocrinology, Skåne University Hospital Malmö, Lund University, Malmö, 20502 Sweden
| | - Joao Fadista
- 6Lund University Diabetes Center, Department of Clinical Sciences, Diabetes & Endocrinology, Skåne University Hospital Malmö, Lund University, Malmö, 20502 Sweden
| | - Marc Santolini
- 1Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA.,2Center for Complex Network Research and Department of Physics, Northeastern University, Boston, MA 02115 USA
| | - Jörg Menche
- 2Center for Complex Network Research and Department of Physics, Northeastern University, Boston, MA 02115 USA.,7 CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, 1090 Austria
| | - Scott T Weiss
- 1Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Marc Vidal
- 3Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215 USA.,8Department of Genetics, Harvard Medical School, Boston, MA 02115 USA
| | - Edwin K Silverman
- 1Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Masanori Aikawa
- 4Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215 USA
| | - Albert-László Barabási
- 1Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA.,2Center for Complex Network Research and Department of Physics, Northeastern University, Boston, MA 02115 USA.,3Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215 USA.,9Center for Network Science, Central European University, Nador u. 9, 1051 Budapest, Hungary
| | - Leif Groop
- 6Lund University Diabetes Center, Department of Clinical Sciences, Diabetes & Endocrinology, Skåne University Hospital Malmö, Lund University, Malmö, 20502 Sweden.,10Department of Clinical Sciences, Islet cell physiology, Skåne University Hospital Malmö, Lund University, Malmö, 20502 Sweden
| | - Joseph Loscalzo
- 11Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA
| |
Collapse
|
189
|
Yamaguchi-Kabata Y, Morihara T, Ohara T, Ninomiya T, Takahashi A, Akatsu H, Hashizume Y, Hayashi N, Shigemizu D, Boroevich KA, Ikeda M, Kubo M, Takeda M, Tsunoda T. Integrated analysis of human genetic association study and mouse transcriptome suggests LBH and SHF genes as novel susceptible genes for amyloid-β accumulation in Alzheimer's disease. Hum Genet 2018; 137:521-533. [PMID: 30006735 PMCID: PMC6061045 DOI: 10.1007/s00439-018-1906-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 07/06/2018] [Indexed: 12/04/2022]
Abstract
Alzheimer's disease (AD) is a common neurological disease that causes dementia in humans. Although the reports of associated pathological genes have been increasing, the molecular mechanism leading to the accumulation of amyloid-β (Aβ) in human brain is still not well understood. To identify novel genes that cause accumulation of Aβ in AD patients, we conducted an integrative analysis by combining a human genetic association study and transcriptome analysis in mouse brain. First, we examined genome-wide gene expression levels in the hippocampus, comparing them to amyloid Aβ level in mice with mixed genetic backgrounds. Next, based on a GWAS statistics obtained by a previous study with human AD subjects, we obtained gene-based statistics from the SNP-based statistics. We combined p values from the two types of analysis across orthologous gene pairs in human and mouse into one p value for each gene to evaluate AD susceptibility. As a result, we found five genes with significant p values in this integrated analysis among the 373 genes analyzed. We also examined the gene expression level of these five genes in the hippocampus of independent human AD cases and control subjects. Two genes, LBH and SHF, showed lower expression levels in AD cases than control subjects. This is consistent with the gene expression levels of both the genes in mouse which were negatively correlated with Aβ accumulation. These results, obtained from the integrative approach, suggest that LBH and SHF are associated with the AD pathogenesis.
Collapse
Affiliation(s)
- Yumi Yamaguchi-Kabata
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
| | - Takashi Morihara
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Tomoyuki Ohara
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Atsushi Takahashi
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Department of Genomic Medicine, Research Institute, National Cerebral and Cardiovascular Center, Osaka, 565-8565, Japan
| | - Hiroyasu Akatsu
- Graduate School of Medical Sciences and Medical School, Nagoya City University, Nagoya, 467-8601, Japan
- Institute of Neuropathology, Fukushimura Hospital, Toyohashi-shi, Aichi, 441-8124, Japan
| | - Yoshio Hashizume
- Institute of Neuropathology, Fukushimura Hospital, Toyohashi-shi, Aichi, 441-8124, Japan
| | - Noriyuki Hayashi
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Daichi Shigemizu
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
- Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Division of Genomic Medicine, Medical Genome Center, National Center for Geriastrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi, 474-8511, Japan
| | - Keith A Boroevich
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Masatoshi Takeda
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Tatsuhiko Tsunoda
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan.
- Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
190
|
Fernández E, Collins MO, Frank RAW, Zhu F, Kopanitsa MV, Nithianantharajah J, Lemprière SA, Fricker D, Elsegood KA, McLaughlin CL, Croning MDR, Mclean C, Armstrong JD, Hill WD, Deary IJ, Cencelli G, Bagni C, Fromer M, Purcell SM, Pocklington AJ, Choudhary JS, Komiyama NH, Grant SGN. Arc Requires PSD95 for Assembly into Postsynaptic Complexes Involved with Neural Dysfunction and Intelligence. Cell Rep 2018; 21:679-691. [PMID: 29045836 PMCID: PMC5656750 DOI: 10.1016/j.celrep.2017.09.045] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 08/03/2017] [Accepted: 09/13/2017] [Indexed: 12/12/2022] Open
Abstract
Arc is an activity-regulated neuronal protein, but little is known about its interactions, assembly into multiprotein complexes, and role in human disease and cognition. We applied an integrated proteomic and genetic strategy by targeting a tandem affinity purification (TAP) tag and Venus fluorescent protein into the endogenous Arc gene in mice. This allowed biochemical and proteomic characterization of native complexes in wild-type and knockout mice. We identified many Arc-interacting proteins, of which PSD95 was the most abundant. PSD95 was essential for Arc assembly into 1.5-MDa complexes and activity-dependent recruitment to excitatory synapses. Integrating human genetic data with proteomic data showed that Arc-PSD95 complexes are enriched in schizophrenia, intellectual disability, autism, and epilepsy mutations and normal variants in intelligence. We propose that Arc-PSD95 postsynaptic complexes potentially affect human cognitive function. TAP tag and purification of endogenous Arc protein complexes from the mouse brain PSD95 is the major Arc binding protein, and both assemble into 1.5-MDa supercomplexes PSD95 is essential for recruitment of Arc to synapses Mutations and genetic variants in Arc-PSD95 are linked to cognition
Collapse
Affiliation(s)
- Esperanza Fernández
- Genes to Cognition Programme, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK; KU Leuven, Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), and VIB Center for the Biology of Disease, Leuven, Belgium
| | - Mark O Collins
- Proteomic Mass Spectrometry, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - René A W Frank
- Genes to Cognition Programme, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK; Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Fei Zhu
- Genes to Cognition Programme, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK; Genes to Cognition Programme, Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK
| | - Maksym V Kopanitsa
- Genes to Cognition Programme, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK; Synome Ltd., Moneta Building, Babraham Research Campus, Cambridge, UK
| | - Jess Nithianantharajah
- Genes to Cognition Programme, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK; Genes to Cognition Programme, Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK
| | - Sarah A Lemprière
- Genes to Cognition Programme, Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK
| | - David Fricker
- Genes to Cognition Programme, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK; Synome Ltd., Moneta Building, Babraham Research Campus, Cambridge, UK
| | - Kathryn A Elsegood
- Genes to Cognition Programme, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK; Genes to Cognition Programme, Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK
| | - Catherine L McLaughlin
- Genes to Cognition Programme, Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK
| | - Mike D R Croning
- Genes to Cognition Programme, Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK
| | - Colin Mclean
- School of Informatics, Institute for Adaptive and Neural Computation, University of Edinburgh, UK
| | - J Douglas Armstrong
- School of Informatics, Institute for Adaptive and Neural Computation, University of Edinburgh, UK
| | - W David Hill
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, UK
| | - Giulia Cencelli
- KU Leuven, Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), and VIB Center for the Biology of Disease, Leuven, Belgium; Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Claudia Bagni
- KU Leuven, Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), and VIB Center for the Biology of Disease, Leuven, Belgium; Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Menachem Fromer
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Psychiatric Genomics, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shaun M Purcell
- Division of Psychiatric Genomics, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrew J Pocklington
- Institute of Psychological Medicine & Clinical Neurosciences, University of Cardiff, Cardiff, Wales, UK
| | - Jyoti S Choudhary
- Proteomic Mass Spectrometry, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Noboru H Komiyama
- Genes to Cognition Programme, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK; Genes to Cognition Programme, Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK
| | - Seth G N Grant
- Genes to Cognition Programme, The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK; Genes to Cognition Programme, Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
191
|
Novel Methods for Family-Based Genetic Studies. Methods Mol Biol 2018. [PMID: 29876895 DOI: 10.1007/978-1-4939-7868-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The recent development of microarray and sequencing technology allows identification of disease susceptibility genes. Although the genome-wide association studies (GWAS) have successfully identified many genetic markers related to human diseases, the traditional statistical methods are not powerful to detect rare genetic markers. The rare genetic markers are usually grouped together and tested at the set level. One of such methods is the sequence kernel association test (SKAT), which has been commonly used in the rare genetic marker analysis. In recent publications, SKAT has been extended to be applicable for family-based rare variant analysis. Here, I present three published statistical approaches for family-based rare variant analysis for: 1. continuous traits, 2. binary traits, and 3. multiple correlated traits.
Collapse
|
192
|
Dai M, Ming J, Cai M, Liu J, Yang C, Wan X, Xu Z. IGESS: a statistical approach to integrating individual-level genotype data and summary statistics in genome-wide association studies. Bioinformatics 2018; 33:2882-2889. [PMID: 28498950 DOI: 10.1093/bioinformatics/btx314] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 05/10/2017] [Indexed: 01/24/2023] Open
Abstract
Motivation Results from genome-wide association studies (GWAS) suggest that a complex phenotype is often affected by many variants with small effects, known as 'polygenicity'. Tens of thousands of samples are often required to ensure statistical power of identifying these variants with small effects. However, it is often the case that a research group can only get approval for the access to individual-level genotype data with a limited sample size (e.g. a few hundreds or thousands). Meanwhile, summary statistics generated using single-variant-based analysis are becoming publicly available. The sample sizes associated with the summary statistics datasets are usually quite large. How to make the most efficient use of existing abundant data resources largely remains an open question. Results In this study, we propose a statistical approach, IGESS, to increasing statistical power of identifying risk variants and improving accuracy of risk prediction by i ntegrating individual level ge notype data and s ummary s tatistics. An efficient algorithm based on variational inference is developed to handle the genome-wide analysis. Through comprehensive simulation studies, we demonstrated the advantages of IGESS over the methods which take either individual-level data or summary statistics data as input. We applied IGESS to perform integrative analysis of Crohns Disease from WTCCC and summary statistics from other studies. IGESS was able to significantly increase the statistical power of identifying risk variants and improve the risk prediction accuracy from 63.2% ( ±0.4% ) to 69.4% ( ±0.1% ) using about 240 000 variants. Availability and implementation The IGESS software is available at https://github.com/daviddaigithub/IGESS . Contact zbxu@xjtu.edu.cn or xwan@comp.hkbu.edu.hk or eeyang@hkbu.edu.hk. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Mingwei Dai
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an, China.,Department of Mathematics, Hong Kong Baptist University, Hong Kong
| | - Jingsi Ming
- Department of Mathematics, Hong Kong Baptist University, Hong Kong
| | - Mingxuan Cai
- Department of Mathematics, Hong Kong Baptist University, Hong Kong
| | - Jin Liu
- Centre of Quantitative Medicine, Duke-NUS Medical School, Singapore
| | - Can Yang
- Department of Mathematics, Hong Kong Baptist University, Hong Kong
| | - Xiang Wan
- Department of Computer Science, Hong Kong Baptist University, Hong Kong
| | - Zongben Xu
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
193
|
Baker E, Schmidt KM, Sims R, O'Donovan MC, Williams J, Holmans P, Escott‐Price V, Consortium WTGERAD. POLARIS: Polygenic LD-adjusted risk score approach for set-based analysis of GWAS data. Genet Epidemiol 2018; 42:366-377. [PMID: 29532500 PMCID: PMC6001515 DOI: 10.1002/gepi.22117] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 01/19/2018] [Accepted: 01/30/2018] [Indexed: 11/11/2022]
Abstract
Polygenic risk scores (PRSs) are a method to summarize the additive trait variance captured by a set of SNPs, and can increase the power of set-based analyses by leveraging public genome-wide association study (GWAS) datasets. PRS aims to assess the genetic liability to some phenotype on the basis of polygenic risk for the same or different phenotype estimated from independent data. We propose the application of PRSs as a set-based method with an additional component of adjustment for linkage disequilibrium (LD), with potential extension of the PRS approach to analyze biologically meaningful SNP sets. We call this method POLARIS: POlygenic Ld-Adjusted RIsk Score. POLARIS identifies the LD structure of SNPs using spectral decomposition of the SNP correlation matrix and replaces the individuals' SNP allele counts with LD-adjusted dosages. Using a raw genotype dataset together with SNP effect sizes from a second independent dataset, POLARIS can be used for set-based analysis. MAGMA is an alternative set-based approach employing principal component analysis to account for LD between markers in a raw genotype dataset. We used simulations, both with simple constructed and real LD-structure, to compare the power of these methods. POLARIS shows more power than MAGMA applied to the raw genotype dataset only, but less or comparable power to combined analysis of both datasets. POLARIS has the advantages that it produces a risk score per person per set using all available SNPs, and aims to increase power by leveraging the effect sizes from the discovery set in a self-contained test of association in the test dataset.
Collapse
Affiliation(s)
- Emily Baker
- Medical Research Council Centre for Neuropsychiatric Genetics and GenomicsInstitute of Psychological Medicine and Clinical Neurosciences, Cardiff UniversityUnited Kingdom
| | | | - Rebecca Sims
- Medical Research Council Centre for Neuropsychiatric Genetics and GenomicsInstitute of Psychological Medicine and Clinical Neurosciences, Cardiff UniversityUnited Kingdom
| | - Michael C. O'Donovan
- Medical Research Council Centre for Neuropsychiatric Genetics and GenomicsInstitute of Psychological Medicine and Clinical Neurosciences, Cardiff UniversityUnited Kingdom
| | - Julie Williams
- Medical Research Council Centre for Neuropsychiatric Genetics and GenomicsInstitute of Psychological Medicine and Clinical Neurosciences, Cardiff UniversityUnited Kingdom
| | - Peter Holmans
- Medical Research Council Centre for Neuropsychiatric Genetics and GenomicsInstitute of Psychological Medicine and Clinical Neurosciences, Cardiff UniversityUnited Kingdom
| | - Valentina Escott‐Price
- Medical Research Council Centre for Neuropsychiatric Genetics and GenomicsInstitute of Psychological Medicine and Clinical Neurosciences, Cardiff UniversityUnited Kingdom
| | - with the GERAD Consortium
- Medical Research Council Centre for Neuropsychiatric Genetics and GenomicsInstitute of Psychological Medicine and Clinical Neurosciences, Cardiff UniversityUnited Kingdom
| |
Collapse
|
194
|
Li W, Fan CC, Mäki-Marttunen T, Thompson WK, Schork AJ, Bettella F, Schizophrenia Working Group of the Psychiatric Genomics Consortium, Djurovic S, Dale AM, Andreassen OA, Wang Y. A molecule-based genetic association approach implicates a range of voltage-gated calcium channels associated with schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2018; 177:454-467. [PMID: 29704319 PMCID: PMC7093061 DOI: 10.1002/ajmg.b.32634] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 02/13/2018] [Accepted: 03/23/2018] [Indexed: 01/06/2023]
Abstract
Traditional genome-wide association studies (GWAS) have successfully detected genetic variants associated with schizophrenia. However, only a small fraction of heritability can be explained. Gene-set/pathway-based methods can overcome limitations arising from single nucleotide polymorphism (SNP)-based analysis, but most of them place constraints on size which may exclude highly specific and functional sets, like macromolecules. Voltage-gated calcium (Cav ) channels, belonging to macromolecules, are composed of several subunits whose encoding genes are located far away or even on different chromosomes. We combined information about such molecules with GWAS data to investigate how functional channels associated with schizophrenia. We defined a biologically meaningful SNP-set based on channel structure and performed an association study by using a validated method: SNP-set (sequence) kernel association test. We identified eight subtypes of Cav channels significantly associated with schizophrenia from a subsample of published data (N = 56,605), including the L-type channels (Cav 1.1, Cav 1.2, Cav 1.3), P-/Q-type Cav 2.1, N-type Cav 2.2, R-type Cav 2.3, T-type Cav 3.1, and Cav 3.3. Only genes from Cav 1.2 and Cav 3.3 have been implicated by the largest GWAS (N = 82,315). Each subtype of Cav channels showed relatively high chip heritability, proportional to the size of its constituent gene regions. The results suggest that abnormalities of Cav channels may play an important role in the pathophysiology of schizophrenia and these channels may represent appropriate drug targets for therapeutics. Analyzing subunit-encoding genes of a macromolecule in aggregate is a complementary way to identify more genetic variants of polygenic diseases. This study offers the potential of power for discovery the biological mechanisms of schizophrenia.
Collapse
Affiliation(s)
- Wen Li
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo 0424 Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Chun Chieh Fan
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA,Multimodal Imaging Laboratory, University of California, San Diego, La Jolla, CA 92093, USA
| | - Tuomo Mäki-Marttunen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo 0424 Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Wesley K. Thompson
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA,Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Mental Health Services, Copenhagen, Denmark,The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark
| | - Andrew J. Schork
- Department of Cognitive Sciences, University of California, San Diego, La Jolla, CA92093, USA
| | - Francesco Bettella
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo 0424 Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | | | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, 0407 Oslo, Norway,NORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Anders M. Dale
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA,Multimodal Imaging Laboratory, University of California, San Diego, La Jolla, CA 92093, USA,Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA,Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ole A. Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo 0424 Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway
| | - Yunpeng Wang
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo 0424 Oslo, Norway,Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo, Norway,Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA,Multimodal Imaging Laboratory, University of California, San Diego, La Jolla, CA 92093, USA,The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Copenhagen, Denmark,Corresponding author information: Dr. Yunpeng Wang, NORMENT, KG Jebsen Centre, Building 49, Oslo University Hospital, Ullevål, Kirkeveien 166, PO Box 4956 Nydalen, 0424 Oslo, Norway, , Phone +47 46 55 96 52, Fax: +47 23 02 73 33
| |
Collapse
|
195
|
Reinbold CS, Forstner AJ, Hecker J, Fullerton JM, Hoffmann P, Hou L, Heilbronner U, Degenhardt F, Adli M, Akiyama K, Akula N, Ardau R, Arias B, Backlund L, Benabarre A, Bengesser S, Bhattacharjee AK, Biernacka JM, Birner A, Marie-Claire C, Cervantes P, Chen GB, Chen HC, Chillotti C, Clark SR, Colom F, Cousins DA, Cruceanu C, Czerski PM, Dayer A, Étain B, Falkai P, Frisén L, Gard S, Garnham JS, Goes FS, Grof P, Gruber O, Hashimoto R, Hauser J, Herms S, Jamain S, Jiménez E, Kahn JP, Kassem L, Kittel-Schneider S, Kliwicki S, König B, Kusumi I, Lackner N, Laje G, Landén M, Lavebratt C, Leboyer M, Leckband SG, López Jaramillo CA, MacQueen G, Manchia M, Martinsson L, Mattheisen M, McCarthy MJ, McElroy SL, Mitjans M, Mondimore FM, Monteleone P, Nievergelt CM, Ösby U, Ozaki N, Perlis RH, Pfennig A, Reich-Erkelenz D, Rouleau GA, Schofield PR, Schubert KO, Schweizer BW, Seemüller F, Severino G, Shekhtman T, Shilling PD, Shimoda K, Simhandl C, Slaney CM, Smoller JW, Squassina A, Stamm TJ, Stopkova P, Tighe SK, Tortorella A, Turecki G, Volkert J, Witt SH, Wright AJ, Young LT, Zandi PP, Potash JB, DePaulo JR, Bauer M, Reininghaus E, Novák T, Aubry JM, et alReinbold CS, Forstner AJ, Hecker J, Fullerton JM, Hoffmann P, Hou L, Heilbronner U, Degenhardt F, Adli M, Akiyama K, Akula N, Ardau R, Arias B, Backlund L, Benabarre A, Bengesser S, Bhattacharjee AK, Biernacka JM, Birner A, Marie-Claire C, Cervantes P, Chen GB, Chen HC, Chillotti C, Clark SR, Colom F, Cousins DA, Cruceanu C, Czerski PM, Dayer A, Étain B, Falkai P, Frisén L, Gard S, Garnham JS, Goes FS, Grof P, Gruber O, Hashimoto R, Hauser J, Herms S, Jamain S, Jiménez E, Kahn JP, Kassem L, Kittel-Schneider S, Kliwicki S, König B, Kusumi I, Lackner N, Laje G, Landén M, Lavebratt C, Leboyer M, Leckband SG, López Jaramillo CA, MacQueen G, Manchia M, Martinsson L, Mattheisen M, McCarthy MJ, McElroy SL, Mitjans M, Mondimore FM, Monteleone P, Nievergelt CM, Ösby U, Ozaki N, Perlis RH, Pfennig A, Reich-Erkelenz D, Rouleau GA, Schofield PR, Schubert KO, Schweizer BW, Seemüller F, Severino G, Shekhtman T, Shilling PD, Shimoda K, Simhandl C, Slaney CM, Smoller JW, Squassina A, Stamm TJ, Stopkova P, Tighe SK, Tortorella A, Turecki G, Volkert J, Witt SH, Wright AJ, Young LT, Zandi PP, Potash JB, DePaulo JR, Bauer M, Reininghaus E, Novák T, Aubry JM, Maj M, Baune BT, Mitchell PB, Vieta E, Frye MA, Rybakowski JK, Kuo PH, Kato T, Grigoroiu-Serbanescu M, Reif A, Del Zompo M, Bellivier F, Schalling M, Wray NR, Kelsoe JR, Alda M, McMahon FJ, Schulze TG, Rietschel M, Nöthen MM, Cichon S. Analysis of the Influence of microRNAs in Lithium Response in Bipolar Disorder. Front Psychiatry 2018; 9:207. [PMID: 29904359 PMCID: PMC5991073 DOI: 10.3389/fpsyt.2018.00207] [Show More Authors] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/03/2018] [Indexed: 12/30/2022] Open
Abstract
Bipolar disorder (BD) is a common, highly heritable neuropsychiatric disease characterized by recurrent episodes of mania and depression. Lithium is the best-established long-term treatment for BD, even though individual response is highly variable. Evidence suggests that some of this variability has a genetic basis. This is supported by the largest genome-wide association study (GWAS) of lithium response to date conducted by the International Consortium on Lithium Genetics (ConLiGen). Recently, we performed the first genome-wide analysis of the involvement of miRNAs in BD and identified nine BD-associated miRNAs. However, it is unknown whether these miRNAs are also associated with lithium response in BD. In the present study, we therefore tested whether common variants at these nine candidate miRNAs contribute to the variance in lithium response in BD. Furthermore, we systematically analyzed whether any other miRNA in the genome is implicated in the response to lithium. For this purpose, we performed gene-based tests for all known miRNA coding genes in the ConLiGen GWAS dataset (n = 2,563 patients) using a set-based testing approach adapted from the versatile gene-based test for GWAS (VEGAS2). In the candidate approach, miR-499a showed a nominally significant association with lithium response, providing some evidence for involvement in both development and treatment of BD. In the genome-wide miRNA analysis, 71 miRNAs showed nominally significant associations with the dichotomous phenotype and 106 with the continuous trait for treatment response. A total of 15 miRNAs revealed nominal significance in both phenotypes with miR-633 showing the strongest association with the continuous trait (p = 9.80E-04) and miR-607 with the dichotomous phenotype (p = 5.79E-04). No association between miRNAs and treatment response to lithium in BD in either of the tested conditions withstood multiple testing correction. Given the limited power of our study, the investigation of miRNAs in larger GWAS samples of BD and lithium response is warranted.
Collapse
Affiliation(s)
- Céline S. Reinbold
- Human Genomics Research Group, Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Andreas J. Forstner
- Human Genomics Research Group, Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - Julian Hecker
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Janice M. Fullerton
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Per Hoffmann
- Human Genomics Research Group, Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Liping Hou
- Intramural Research Program, US Department of Health & Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Urs Heilbronner
- Department Psychiatry and Psychotherapy, Institute of Psychiatric Phenomics and Genomics, University Hospital, LMU Munich, Munich, Germany
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Mazda Adli
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
| | - Kazufumi Akiyama
- Department of Biological Psychiatry and Neuroscience, Dokkyo Medical University School of Medicine, Mibu, Japan
| | - Nirmala Akula
- Intramural Research Program, US Department of Health & Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Raffaella Ardau
- Unit of Clinical Pharmacology, University Hospital of Cagliari, Cagliari, Italy
| | - Bárbara Arias
- Zoology and Biological Anthropology Section (Department of Evolutive Biology, Ecology and Environmental Sciences), Facultat de Biologia and Institut de Biomedicina, CIBERSAM, Universitat de Barcelona, Barcelona, Spain
| | - Lena Backlund
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Antonio Benabarre
- Bipolar Disorder Program, Institute of Neuroscience, Hospital Clinic, CIBERSAM, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Susanne Bengesser
- Special Outpatient Center for Bipolar Affective Disorder, Medical University of Graz, Graz, Austria
| | | | - Joanna M. Biernacka
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
- Institut National de la Santé et de la Recherche Médicale, U1144, Paris, France
| | - Armin Birner
- Special Outpatient Center for Bipolar Affective Disorder, Medical University of Graz, Graz, Austria
| | - Cynthia Marie-Claire
- Université Paris Diderot, Sorbonne Paris Cité, UMR-S 1144, Paris, France
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Pablo Cervantes
- The Neuromodulation Unit, McGill University Health Centre, Montreal, QC, Canada
| | - Guo-Bo Chen
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Hsi-Chung Chen
- Department of Psychiatry & Center of Sleep Disorders, National Taiwan University Hospital, Taipei, Taiwan
| | - Caterina Chillotti
- Unit of Clinical Pharmacology, University Hospital of Cagliari, Cagliari, Italy
| | - Scott R. Clark
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
| | - Francesc Colom
- Mental Health Research Group, IMIM-Hospital del Mar, Barcelona, Spain
| | - David A. Cousins
- Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Cristiana Cruceanu
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Piotr M. Czerski
- Psychiatric Genetic Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Alexandre Dayer
- Mood Disorders Unit, HUG - Geneva University Hospitals, Geneva, Switzerland
| | - Bruno Étain
- Université Paris Diderot, Sorbonne Paris Cité, UMR-S 1144, Paris, France
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
- AP-HP, GH Saint-Louis – Lariboisière – F. Widal, Département de Psychiatrie et de Médecine Addictologique, Paris, France
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Louise Frisén
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Sébastien Gard
- Service de Psychiatrie, Hôpital Charles Perrens, Bordeaux, France
| | - Julie S. Garnham
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Fernando S. Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Paul Grof
- Mood Disorders Center of Ottawa, Ottawa, ON, Canada
| | - Oliver Gruber
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August University Göttingen, Göttingen, Germany
| | - Ryota Hashimoto
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Joanna Hauser
- Psychiatric Genetic Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Stefan Herms
- Human Genomics Research Group, Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Stéphane Jamain
- Institut National de la Santé et de la Recherche Médicale U955, Psychiatrie Translationnelle, Créteil, France
| | - Esther Jiménez
- Bipolar Disorder Program, Institute of Neuroscience, Hospital Clinic, CIBERSAM, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Jean-Pierre Kahn
- Service de Psychiatrie et Psychologie Clinique, Centre Psychothérapique de Nancy - Université de Lorraine, Nancy, France
| | - Layla Kassem
- Intramural Research Program, US Department of Health & Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Sebastian Kliwicki
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Barbara König
- Department of Psychiatry and Psychotherapeuthic Medicine, Landesklinikum Neunkirchen, Neunkirchen, Austria
| | - Ichiro Kusumi
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Nina Lackner
- Special Outpatient Center for Bipolar Affective Disorder, Medical University of Graz, Graz, Austria
| | - Gonzalo Laje
- Intramural Research Program, US Department of Health & Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Mikael Landén
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the Gothenburg University, Gothenburg, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Marion Leboyer
- Assistance Publique-Hôpitaux de Paris, Hôpital Albert Chenevier - Henri Mondor, Pôle de Psychiatrie, Créteil, France
| | - Susan G. Leckband
- Department of Pharmacy, VA San Diego Healthcare System, San Diego, CA, United States
| | | | - Glenda MacQueen
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Lina Martinsson
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
| | | | - Michael J. McCarthy
- Department of Psychiatry, VA San Diego Healthcare System, San Diego, CA, United States
| | - Susan L. McElroy
- Department of Psychiatry, Lindner Center of Hope/University of Cincinnati, Mason, OH, United States
| | - Marina Mitjans
- Zoology and Biological Anthropology Section (Department of Evolutive Biology, Ecology and Environmental Sciences), Facultat de Biologia and Institut de Biomedicina, CIBERSAM, Universitat de Barcelona, Barcelona, Spain
| | - Francis M. Mondimore
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Palmiero Monteleone
- Neurosciences Section, Department of Medicine and Surgery, University of Salerno, Salerno, Italy
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | | | - Urban Ösby
- Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Roy H. Perlis
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Andrea Pfennig
- Department of Psychiatry and Psychotherapy, Medical Faculty, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Daniela Reich-Erkelenz
- Department Psychiatry and Psychotherapy, Institute of Psychiatric Phenomics and Genomics, University Hospital, LMU Munich, Munich, Germany
| | - Guy A. Rouleau
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Peter R. Schofield
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
- Mental Illness, Neuroscience Research Australia, Sydney, NSW, Australia
| | - K. Oliver Schubert
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
| | - Barbara W. Schweizer
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Florian Seemüller
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Giovanni Severino
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | | | - Paul D. Shilling
- Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | - Kazutaka Shimoda
- Department of Psychiatry, Dokkyo Medical University School of Medicine, Mibu, Japan
| | - Christian Simhandl
- Medical school, Sigmund Freud University, Vienna, Austria
- Bipolar Center Wiener Neustadt, Vienna, Austria
| | - Claire M. Slaney
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Jordan W. Smoller
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Alessio Squassina
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Thomas J. Stamm
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical School Brandenburg, Neuruppin, Germany
| | - Pavla Stopkova
- Department of Psychiatry, National Institute of Mental Health, Klecany, Czechia
| | - Sarah K. Tighe
- Department of Psychiatry, University of Iowa, Iowa, IA, United States
| | - Alfonso Tortorella
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Gustavo Turecki
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Julia Volkert
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Stephanie H. Witt
- Department of Genetic Epidemiology in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Adam J. Wright
- School of Psychiatry, University of New South Wales, and Black Dog Institute, Sydney, NSW, Australia
| | - L. Trevor Young
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Peter P. Zandi
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - James B. Potash
- Department of Psychiatry, University of Iowa, Iowa, IA, United States
| | - J. Raymond DePaulo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Michael Bauer
- Department of Psychiatry and Psychotherapy, Medical Faculty, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Eva Reininghaus
- Special Outpatient Center for Bipolar Affective Disorder, Medical University of Graz, Graz, Austria
| | - Tomáš Novák
- Department of Psychiatry, National Institute of Mental Health, Klecany, Czechia
| | - Jean-Michel Aubry
- Mood Disorders Unit, HUG - Geneva University Hospitals, Geneva, Switzerland
| | - Mario Maj
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Bernhard T. Baune
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
| | - Philip B. Mitchell
- School of Psychiatry, University of New South Wales, and Black Dog Institute, Sydney, NSW, Australia
| | - Eduard Vieta
- Bipolar Disorder Program, Institute of Neuroscience, Hospital Clinic, CIBERSAM, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Mark A. Frye
- Institut National de la Santé et de la Recherche Médicale, U1144, Paris, France
| | - Janusz K. Rybakowski
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Po-Hsiu Kuo
- Department of Public Health and Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Saitama, Japan
| | - Maria Grigoroiu-Serbanescu
- Biometric Psychiatric Genetics Research Unit, Alexandru Obregia Clinical Psychiatric Hospital, Bucharest, Romania
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Maria Del Zompo
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Frank Bellivier
- Université Paris Diderot, Sorbonne Paris Cité, UMR-S 1144, Paris, France
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
- AP-HP, GH Saint-Louis – Lariboisière – F. Widal, Département de Psychiatrie et de Médecine Addictologique, Paris, France
| | - Martin Schalling
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Naomi R. Wray
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - John R. Kelsoe
- Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
- Department of Psychiatry, National Institute of Mental Health, Klecany, Czechia
| | - Francis J. McMahon
- Intramural Research Program, US Department of Health & Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Thomas G. Schulze
- Intramural Research Program, US Department of Health & Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
- Department Psychiatry and Psychotherapy, Institute of Psychiatric Phenomics and Genomics, University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, United States
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August University Göttingen, Göttingen, Germany
- Department of Genetic Epidemiology in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Markus M. Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Sven Cichon
- Human Genomics Research Group, Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Research Centre Jülich, Institute of Neuroscience and Medicine (INM-1), Jülich, Germany
| |
Collapse
|
196
|
Association Study and Fine-Mapping Major Histocompatibility Complex Analysis of Pemphigus Vulgaris in a Han Chinese Population. J Invest Dermatol 2018; 138:2307-2314. [PMID: 29857070 DOI: 10.1016/j.jid.2018.05.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 05/08/2018] [Accepted: 05/13/2018] [Indexed: 12/15/2022]
Abstract
To identify possible additional genetic susceptibility loci for pemphigus vulgaris (PV), we performed a genome-wide association study of 240 PV patients and 1,031 control individuals, and we selected the top single nucleotide polymorphisms for replication in independent samples, with 252 patient samples and 1,852 control samples. We identified rs11218708 (P = 3.1 × 10-8, odds ratio = 1.54) at chromosome locus 11q24.1 as significantly associated with PV. A fine-mapping analysis of PV risk in the major histocompatibility complex region showed three independent variants predisposed to PV using stepwise analysis: HLA-DRB1*14:04 (P = 2.47 × 10-38, odds ratio = 6.28), rs7454108 at the TAP2 gene (P = 2.78 × 10-12, odds ratio = 3.25), and rs1051336 at the HLA-DRA gene (P = 3.06 × 10-6, odds ratio = 0.33). A systematic evaluation using gene- and pathway-based analyses showed a high tendency for PV susceptibility genes to be associated with autoimmunity. Our study highlights the involvement of immune-mediated processes in the pathophysiology of PV and illustrates the value of imputation to identify variants in the major histocompatibility complex region.
Collapse
|
197
|
Davies G, Lam M, Harris SE, Trampush JW, Luciano M, Hill WD, Hagenaars SP, Ritchie SJ, Marioni RE, Fawns-Ritchie C, Liewald DCM, Okely JA, Ahola-Olli AV, Barnes CLK, Bertram L, Bis JC, Burdick KE, Christoforou A, DeRosse P, Djurovic S, Espeseth T, Giakoumaki S, Giddaluru S, Gustavson DE, Hayward C, Hofer E, Ikram MA, Karlsson R, Knowles E, Lahti J, Leber M, Li S, Mather KA, Melle I, Morris D, Oldmeadow C, Palviainen T, Payton A, Pazoki R, Petrovic K, Reynolds CA, Sargurupremraj M, Scholz M, Smith JA, Smith AV, Terzikhan N, Thalamuthu A, Trompet S, van der Lee SJ, Ware EB, Windham BG, Wright MJ, Yang J, Yu J, Ames D, Amin N, Amouyel P, Andreassen OA, Armstrong NJ, Assareh AA, Attia JR, Attix D, Avramopoulos D, Bennett DA, Böhmer AC, Boyle PA, Brodaty H, Campbell H, Cannon TD, Cirulli ET, Congdon E, Conley ED, Corley J, Cox SR, Dale AM, Dehghan A, Dick D, Dickinson D, Eriksson JG, Evangelou E, Faul JD, Ford I, Freimer NA, Gao H, Giegling I, Gillespie NA, Gordon SD, Gottesman RF, Griswold ME, Gudnason V, Harris TB, Hartmann AM, Hatzimanolis A, Heiss G, Holliday EG, Joshi PK, Kähönen M, Kardia SLR, Karlsson I, Kleineidam L, et alDavies G, Lam M, Harris SE, Trampush JW, Luciano M, Hill WD, Hagenaars SP, Ritchie SJ, Marioni RE, Fawns-Ritchie C, Liewald DCM, Okely JA, Ahola-Olli AV, Barnes CLK, Bertram L, Bis JC, Burdick KE, Christoforou A, DeRosse P, Djurovic S, Espeseth T, Giakoumaki S, Giddaluru S, Gustavson DE, Hayward C, Hofer E, Ikram MA, Karlsson R, Knowles E, Lahti J, Leber M, Li S, Mather KA, Melle I, Morris D, Oldmeadow C, Palviainen T, Payton A, Pazoki R, Petrovic K, Reynolds CA, Sargurupremraj M, Scholz M, Smith JA, Smith AV, Terzikhan N, Thalamuthu A, Trompet S, van der Lee SJ, Ware EB, Windham BG, Wright MJ, Yang J, Yu J, Ames D, Amin N, Amouyel P, Andreassen OA, Armstrong NJ, Assareh AA, Attia JR, Attix D, Avramopoulos D, Bennett DA, Böhmer AC, Boyle PA, Brodaty H, Campbell H, Cannon TD, Cirulli ET, Congdon E, Conley ED, Corley J, Cox SR, Dale AM, Dehghan A, Dick D, Dickinson D, Eriksson JG, Evangelou E, Faul JD, Ford I, Freimer NA, Gao H, Giegling I, Gillespie NA, Gordon SD, Gottesman RF, Griswold ME, Gudnason V, Harris TB, Hartmann AM, Hatzimanolis A, Heiss G, Holliday EG, Joshi PK, Kähönen M, Kardia SLR, Karlsson I, Kleineidam L, Knopman DS, Kochan NA, Konte B, Kwok JB, Le Hellard S, Lee T, Lehtimäki T, Li SC, Lill CM, Liu T, Koini M, London E, Longstreth WT, Lopez OL, Loukola A, Luck T, Lundervold AJ, Lundquist A, Lyytikäinen LP, Martin NG, Montgomery GW, Murray AD, Need AC, Noordam R, Nyberg L, Ollier W, Papenberg G, Pattie A, Polasek O, Poldrack RA, Psaty BM, Reppermund S, Riedel-Heller SG, Rose RJ, Rotter JI, Roussos P, Rovio SP, Saba Y, Sabb FW, Sachdev PS, Satizabal CL, Schmid M, Scott RJ, Scult MA, Simino J, Slagboom PE, Smyrnis N, Soumaré A, Stefanis NC, Stott DJ, Straub RE, Sundet K, Taylor AM, Taylor KD, Tzoulaki I, Tzourio C, Uitterlinden A, Vitart V, Voineskos AN, Kaprio J, Wagner M, Wagner H, Weinhold L, Wen KH, Widen E, Yang Q, Zhao W, Adams HHH, Arking DE, Bilder RM, Bitsios P, Boerwinkle E, Chiba-Falek O, Corvin A, De Jager PL, Debette S, Donohoe G, Elliott P, Fitzpatrick AL, Gill M, Glahn DC, Hägg S, Hansell NK, Hariri AR, Ikram MK, Jukema JW, Vuoksimaa E, Keller MC, Kremen WS, Launer L, Lindenberger U, Palotie A, Pedersen NL, Pendleton N, Porteous DJ, Räikkönen K, Raitakari OT, Ramirez A, Reinvang I, Rudan I, Dan Rujescu, Schmidt R, Schmidt H, Schofield PW, Schofield PR, Starr JM, Steen VM, Trollor JN, Turner ST, Van Duijn CM, Villringer A, Weinberger DR, Weir DR, Wilson JF, Malhotra A, McIntosh AM, Gale CR, Seshadri S, Mosley TH, Bressler J, Lencz T, Deary IJ. Study of 300,486 individuals identifies 148 independent genetic loci influencing general cognitive function. Nat Commun 2018; 9:2098. [PMID: 29844566 PMCID: PMC5974083 DOI: 10.1038/s41467-018-04362-x] [Show More Authors] [Citation(s) in RCA: 423] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 04/23/2018] [Indexed: 11/15/2022] Open
Abstract
General cognitive function is a prominent and relatively stable human trait that is associated with many important life outcomes. We combine cognitive and genetic data from the CHARGE and COGENT consortia, and UK Biobank (total N = 300,486; age 16-102) and find 148 genome-wide significant independent loci (P < 5 × 10-8) associated with general cognitive function. Within the novel genetic loci are variants associated with neurodegenerative and neurodevelopmental disorders, physical and psychiatric illnesses, and brain structure. Gene-based analyses find 709 genes associated with general cognitive function. Expression levels across the cortex are associated with general cognitive function. Using polygenic scores, up to 4.3% of variance in general cognitive function is predicted in independent samples. We detect significant genetic overlap between general cognitive function, reaction time, and many health variables including eyesight, hypertension, and longevity. In conclusion we identify novel genetic loci and pathways contributing to the heritability of general cognitive function.
Collapse
Affiliation(s)
- Gail Davies
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Max Lam
- Institute of Mental Health, Singapore, 539747, Singapore
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Medical Genetics Section, Centre for Genomic & Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Joey W Trampush
- BrainWorkup, LLC, Los Angeles, 90033, CA, USA
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, 90033, CA, USA
| | - Michelle Luciano
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - W David Hill
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Saskia P Hagenaars
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
| | - Stuart J Ritchie
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Riccardo E Marioni
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Medical Genetics Section, Centre for Genomic & Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Chloe Fawns-Ritchie
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - David C M Liewald
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Judith A Okely
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Ari V Ahola-Olli
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, 20520, Finland
- Department of Internal Medicine, Satakunta Central Hospital, Pori, 28100, Finland
| | - Catriona L K Barnes
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - Lars Bertram
- Max Planck Institute for Molecular Genetics, Berlin, 14195, Germany
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, 98101, Washington, USA
| | - Katherine E Burdick
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
- Mental Illness Research, Education, and Clinical Center (VISN 3), James J. Peters VA Medical Center, Bronx, 10468, NY, USA
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA
| | - Andrea Christoforou
- NORMENT, K.G. Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, 5020, Norway
| | - Pamela DeRosse
- Institute of Mental Health, Singapore, 539747, Singapore
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, 11030, NY, USA
| | - Srdjan Djurovic
- NORMENT, K.G. Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
- Department of Medical Genetics, Oslo University Hospital, University of Bergen, Oslo, 0424, Norway
| | - Thomas Espeseth
- Department of Psychology, University of Oslo, Oslo, 0373, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, 0315, Norway
| | - Stella Giakoumaki
- Department of Psychology, University of Crete, Crete, GR-74100, Greece
| | - Sudheer Giddaluru
- NORMENT, K.G. Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, 5020, Norway
| | - Daniel E Gustavson
- Department of Psychiatry, University of California, San Diego, 92093, CA, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, 92093, CA, USA
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Edith Hofer
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University of Graz, Graz, 8036, Austria
- Institute of Medical Informatics Statistics and Documentation, Medical University of Graz, Graz, 8036, Austria
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, 3015, The Netherlands
- Department of Neurology, Erasmus University Medical Center, Rotterdam, xxxxxx, The Netherlands
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Emma Knowles
- Department of Psychiatry, Yale University School of Medicine, New Haven, 06511, CT, USA
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
- Helsinki Collegium for Advanced Studies, University of Helsinki, Helsinki, 00014, Finland
| | - Markus Leber
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, D-50937, Germany
| | - Shuo Li
- Department of Biostatistics, Boston University School of Public Health, Boston, 02118, MA, USA
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, 2031, Australia
| | - Ingrid Melle
- NORMENT, K.G. Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
- Department of Psychology, University of Oslo, Oslo, 0373, Norway
| | - Derek Morris
- Neuroimaging, Cognition & Genomics (NICOG) Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - Christopher Oldmeadow
- Medical Research Institute and Faculty of Health, University of Newcastle, New South Wa0les, 2308, Australia
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, FI-00014, Finland
| | - Antony Payton
- Centre for EpidemiologyDivision of Population Health, Health Services Research & Primary Care, The University of Manchester, Manchester, M13 9PL, UK
| | - Raha Pazoki
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W2 1PG, UK
| | - Katja Petrovic
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University of Graz, Graz, 8036, Austria
| | - Chandra A Reynolds
- Department of Psychology, University of California Riverside, Riverside, 92521, CA, USA
| | - Muralidharan Sargurupremraj
- University of Bordeaux, Bordeaux Population Health Research Center, INSERM UMR 1219, F-33000, Bordeaux, France
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, 04107, Germany
- LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, 04107, Germany
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, IS-201, Iceland
- University of Iceland, Reykjavik, 101, Iceland
| | - Natalie Terzikhan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015, The Netherlands
- Department of Respiratory Medicine, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, 2031, Australia
| | - Stella Trompet
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, 2333, The Netherlands
| | - Sven J van der Lee
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015, The Netherlands
| | - Erin B Ware
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - B Gwen Windham
- Department of Medicine, Division of Geriatrics, University of Mississippi Medical Center, Jackson, 39216, MS, USA
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, 4072, Australia
- Centre for Advanced Imaging, University of Queensland, Brisbane, 4072, Australia
| | - Jingyun Yang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, 60612, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, 60612, IL, USA
| | - Jin Yu
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, 11030, NY, USA
| | - David Ames
- National Ageing Research Institute, Royal Melbourne Hospital, Victoria, 3052, Australia
- Academic Unit for Psychiatry of Old Age, University of Melbourne, St George's Hospital, Kew, 3010, Australia
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015, The Netherlands
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-LabEx DISTALZ, F-59000, Lille, France
| | - Ole A Andreassen
- Department of Psychology, University of Oslo, Oslo, 0373, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, 0372, Norway
| | | | - Amelia A Assareh
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, 2031, Australia
| | - John R Attia
- Hunter Medical Research Institute and Faculty of Health, University of Newcastle, New South Wales, 2305, Australia
| | - Deborah Attix
- Department of NeurologyBryan Alzheimer's Disease Research Center, and Center for Genomic and Computational Biology, Duke University Medical Center, Durham, 27708, NC, USA
- Psychiatry and Behavioral Sciences, Division of Medical Psychology, and Department of Neurology, Duke University Medical Center, Durham, 27708, NC, USA
| | - Dimitrios Avramopoulos
- Department of Psychiatry, Johns Hopkins University School of Medicine, MD, Baltimore, 21287, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, MD, Baltimore, 21287, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, 60612, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, 60612, IL, USA
| | - Anne C Böhmer
- Institute of Human Genetics, University of Bonn, Bonn, 53113, Germany
- Department of Genomics, Life and Brain Center, University of Bonn, Bonn, 53113, Germany
| | - Patricia A Boyle
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, 60612, IL, USA
- Departments of Behavioral Sciences, Rush University Medical Center, Chicago, 60612, IL, USA
| | - Henry Brodaty
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, 2031, Australia
- Dementia Centre for Research Collaboration, University of New South Wales, Sydney, 2031, NSW, Australia
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - Tyrone D Cannon
- Department of Psychology, Yale University, New Haven, 06520, CT, USA
| | | | - Eliza Congdon
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, 90024, CA, USA
| | | | - Janie Corley
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Simon R Cox
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Anders M Dale
- Department of Psychiatry, University of California, San Diego, 92093, CA, USA
- Department of Cognitive Science, University of California, San Diego, La Jolla, 92093, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, 92093, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, 92093, CA, USA
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W2 1PG, UK
- MRC-PHE Centre for Environment, School of Public Health, Imperial College London, London, W2 1PG, UK
| | - Danielle Dick
- Department of Psychology, Virginia Commonwealth University, Richmond, 23284, VA, USA
| | - Dwight Dickinson
- Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institute of Health, Bethesda, 20892, MD, USA
| | - Johan G Eriksson
- National Institute for Health and Welfare, Helsinki, FI-00271, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, 00290, Finland
- Helsinki University Central Hospital, Unit of General Practice, Helsinki, FI-00029, Finland
- Folkhälsan Research Centre, Helsinki, 2018, Finland
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W2 1PG, UK
- National Institute for Health and Welfare, Helsinki, FI-00271, Finland
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Nelson A Freimer
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, 90024, CA, USA
| | - He Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W2 1PG, UK
| | - Ina Giegling
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, 06108, Germany
| | - Nathan A Gillespie
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, 23298, VA, USA
| | - Scott D Gordon
- QIMR Berghofer Medical Research Institute, Brisbane, 4029, Australia
| | - Rebecca F Gottesman
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, 21205, MD, USA
| | - Michael E Griswold
- Department of Data Science, University of Mississippi Medical Center, Jackson, 39216, MS, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, IS-201, Iceland
- University of Iceland, Reykjavik, 101, Iceland
| | - Tamara B Harris
- Intramural Research Program National Institutes on Aging, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Annette M Hartmann
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, 06108, Germany
| | - Alex Hatzimanolis
- Department of Psychiatry, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, 11528, Greece
- University Mental Health Research Institute, Athens, GR-156 01, Greece
- Neurobiology Research Institute, Theodor-Theohari Cozzika Foundation, Athens, 11521, Greece
| | - Gerardo Heiss
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, 27599, NC, USA
| | - Elizabeth G Holliday
- Hunter Medical Research Institute and Faculty of Health, University of Newcastle, New South Wales, 2305, Australia
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, and Finnish Cardiovascular Research Center, Tampere, FI-33014, Finland
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33521, Finland
- Department of Clinical Physiology, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ida Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Luca Kleineidam
- Department of Psychiatry Medical Faculty, University of Cologne, Cologne, 50923, Germany
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, 53127, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53127, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, 53127, Germany
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, 55905, MN, USA
| | - Nicole A Kochan
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, 2031, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, 2031, Australia
| | - Bettina Konte
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, 06108, Germany
| | - John B Kwok
- Brain and Mind Centre-The University of Sydney, Camperdown, NSW, 2050, Australia
- School of Medical Sciences, University of New South Wales, Sydney, 2052, Australia
| | - Stephanie Le Hellard
- NORMENT, K.G. Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, 5020, Norway
| | - Teresa Lee
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, 2031, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, 2031, Australia
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
| | - Shu-Chen Li
- Max Planck Institute for Human Development, Berlin, 14195, Germany
- Technische Universität Dresden, Dresden, 01187, Germany
| | - Christina M Lill
- Genetic and Molecular Epidemiology Group, Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Tian Liu
- Max Planck Institute for Molecular Genetics, Berlin, 14195, Germany
- Max Planck Institute for Human Development, Berlin, 14195, Germany
| | - Marisa Koini
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University of Graz, Graz, 8036, Austria
| | - Edythe London
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, 90024, CA, USA
| | - Will T Longstreth
- Department of Neurology, School of Medicine, University of Washington, Seattle, 98195-6465, WA, USA
- Department of Epidemiology, University of Washington, Seattle, 98195, WA, USA
| | - Oscar L Lopez
- Department of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, 15213, PA, USA
| | - Anu Loukola
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, FI-00014, Finland
| | - Tobias Luck
- LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, 04107, Germany
- Institute of Social Medicine, Occupational Health and Public Health (ISAP), University of Leipzig, Leipzig, 04103, Germany
| | - Astri J Lundervold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, 5009, Norway
- K. G. Jebsen Center for Neuropsychiatry, University of Bergen, Bergen, N-5009, Norway
| | - Anders Lundquist
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, SE-901 87, Sweden
- Department of Statistics, USBE Umeå University, S-907 97, Umeå, Sweden
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, 4029, Australia
| | - Grant W Montgomery
- QIMR Berghofer Medical Research Institute, Brisbane, 4029, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, 4072, Australia
| | - Alison D Murray
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
- The Institute of Medical Sciences, Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Anna C Need
- Division of Brain Sciences, Department of Medicine, Imperial College, London, SW7 2AZ, UK
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, 2333, The Netherlands
| | - Lars Nyberg
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, SE-901 87, Sweden
- Department of Radiation Sciences, Umeå University, Umeå, SE-901 87, Sweden
- Department of Integrative Medical Biology, Umeå University, Umeå, SE-901 87, Sweden
| | - William Ollier
- Centre for Integrated Genomic Medical Research, Institute of Population Health, University of Manchester, Manchester, M13 9PT, UK
| | - Goran Papenberg
- Max Planck Institute for Human Development, Berlin, 14195, Germany
- Karolinska Institutet, Aging Research Center, Stockholm University, Stockholm, SE-113 30, Sweden
| | - Alison Pattie
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Ozren Polasek
- Gen-Info LLC, Zagreb, 10000, Croatia
- Faculty of Medicine, University of Split, Split, 21000, Croatia
| | - Russell A Poldrack
- Department of Psychology, Stanford University, Palo Alto, 94305-2130, CA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, 98101, Washington, USA
- Deparment of Health Services, University of Washington, Seattle, 98195-7660, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, 98101, WA, USA
| | - Simone Reppermund
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, 2031, Australia
- Department of Developmental Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney, 2052, Australia
| | - Steffi G Riedel-Heller
- Institute of Social Medicine, Occupational Health and Public Health (ISAP), University of Leipzig, Leipzig, 04103, Germany
| | - Richard J Rose
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405-7007, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, 90509, CA, USA
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
- Mental Illness Research, Education, and Clinical Center (VISN 2), James J. Peters VA Medical Center, Bronx, 10468, NY, USA
| | - Suvi P Rovio
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, 20520, Finland
| | - Yasaman Saba
- Institute of Molecular Biology and Biochemistry, Centre for Molecular Medicine, Medical University of Graz, Graz, 8036, Austria
| | - Fred W Sabb
- Robert and Beverly Lewis Center for Neuroimaging, University of Oregon, Eugene, 97403, OR, USA
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, 2031, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, 2031, Australia
| | - Claudia L Satizabal
- Department of Neurology, Boston University School of Medicine, Boston, 02118, MA, USA
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, 01702-5827, MA, USA
| | - Matthias Schmid
- Department of Medical Biometry, Informatics and Epidemiology, University Hospital, Bonn, D-53012, Germany
| | - Rodney J Scott
- Hunter Medical Research Institute and Faculty of Health, University of Newcastle, New South Wales, 2305, Australia
| | - Matthew A Scult
- Laboratory of NeuroGenetics, Department of Psychology & Neuroscience, Duke University, Durham, 27708-0086, NC, USA
| | - Jeannette Simino
- Department of Data Science, University of Mississippi Medical Center, Jackson, 39216, MS, USA
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, 2333, The Netherlands
| | - Nikolaos Smyrnis
- Department of Psychiatry, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, 11528, Greece
- University Mental Health Research Institute, Athens, GR-156 01, Greece
| | - Aïcha Soumaré
- University of Bordeaux, Bordeaux Population Health Research Center, INSERM UMR 1219, F-33000, Bordeaux, France
| | - Nikos C Stefanis
- Department of Psychiatry, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, 11528, Greece
- University Mental Health Research Institute, Athens, GR-156 01, Greece
- Neurobiology Research Institute, Theodor-Theohari Cozzika Foundation, Athens, 11521, Greece
| | - David J Stott
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Richard E Straub
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, 21205, MD, USA
| | - Kjetil Sundet
- Department of Psychology, University of Oslo, Oslo, 0373, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, 0315, Norway
| | - Adele M Taylor
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, 90509, CA, USA
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W2 1PG, UK
- MRC-PHE Centre for Environment, School of Public Health, Imperial College London, London, W2 1PG, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Christophe Tzourio
- University of Bordeaux, Bordeaux Population Health Research Center, INSERM UMR 1219, F-33000, Bordeaux, France
- Department of Public Health, University Hospital of Bordeaux, Bordeaux, 33076, France
| | - André Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, 3015, The Netherlands
| | - Veronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Aristotle N Voineskos
- Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, M5T 1L8, Canada
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, FI-00014, Finland
- National Institute for Health and Welfare, Helsinki, FI-00271, Finland
- Department of Public Health, University of Helsinki, Helsinki, 00014, Finland
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53127, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, 53127, Germany
| | - Holger Wagner
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, 53127, Germany
| | - Leonie Weinhold
- Department of Medical Biometry, Informatics and Epidemiology, University Hospital, Bonn, D-53012, Germany
| | - K Hoyan Wen
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015, The Netherlands
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, FI-00014, Finland
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, 02118, MA, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hieab H H Adams
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015, The Netherlands
- Department of Radiology, Erasmus MC, Rotterdam, 3015, The Netherlands
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, MD, Baltimore, 21287, USA
| | - Robert M Bilder
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, 90024, CA, USA
| | - Panos Bitsios
- Department of Psychiatry and Behavioral Sciences, Faculty of Medicine, University of Crete, Heraklion, GR-71003, Greece
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, 77030, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, 77030-3411, TX, USA
| | - Ornit Chiba-Falek
- Department of NeurologyBryan Alzheimer's Disease Research Center, and Center for Genomic and Computational Biology, Duke University Medical Center, Durham, 27708, NC, USA
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, DO2 AY89, Ireland
| | - Philip L De Jager
- Center for Translational and Systems Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, 10032, NY, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, 02142, MA, USA
| | - Stéphanie Debette
- University of Bordeaux, Bordeaux Population Health Research Center, INSERM UMR 1219, F-33000, Bordeaux, France
- Department of Neurology, University Hospital of Bordeaux, Bordeaux, 33000, France
| | - Gary Donohoe
- Neuroimaging, Cognition & Genomics (NICOG) Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W2 1PG, UK
- MRC-PHE Centre for Environment, School of Public Health, Imperial College London, London, W2 1PG, UK
| | - Annette L Fitzpatrick
- Department of Epidemiology, University of Washington, Seattle, 98195, WA, USA
- Department of Global Health, University of Washington, Seattle, 98104, WA, USA
| | - Michael Gill
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, DO2 AY89, Ireland
| | - David C Glahn
- Department of Psychiatry, Yale University School of Medicine, New Haven, 06511, CT, USA
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Narelle K Hansell
- Queensland Brain Institute, University of Queensland, Brisbane, 4072, Australia
| | - Ahmad R Hariri
- Laboratory of NeuroGenetics, Department of Psychology & Neuroscience, Duke University, Durham, 27708-0086, NC, USA
| | - M Kamran Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015, The Netherlands
- Department of Neurology, Erasmus University Medical Center, Rotterdam, xxxxxx, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, 2333, The Netherlands
| | - Eero Vuoksimaa
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, FI-00014, Finland
- Department of Public Health, University of Helsinki, Helsinki, 00014, Finland
| | - Matthew C Keller
- Institute for Behavioral Genetics, University of Colorado, Boulder, 80309, CO, USA
| | - William S Kremen
- Department of Psychiatry, University of California, San Diego, 92093, CA, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, 92093, CA, USA
| | - Lenore Launer
- Intramural Research Program National Institutes on Aging, National Institutes of Health, Bethesda, 20892, MD, USA
| | | | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, FI-00014, Finland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, CB10 1SA, UK
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, 00014, Finland
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Neil Pendleton
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Manchester Academic Health Science Centre, and Manchester Medical School, Institute of Brain, Behaviour, and Mental Health, University of Manchester, Manchester, M13 9PL, UK
| | - David J Porteous
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Medical Genetics Section, Centre for Genomic & Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, 20520, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, 20520, Finland
| | - Alfredo Ramirez
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, D-50937, Germany
- Institute of Human Genetics, University of Bonn, Bonn, 53113, Germany
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, 53127, Germany
| | - Ivar Reinvang
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, 0315, Norway
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - Dan Rujescu
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, 06108, Germany
| | - Reinhold Schmidt
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University of Graz, Graz, 8036, Austria
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry, Centre for Molecular Medicine, Medical University of Graz, Graz, 8036, Austria
| | - Peter W Schofield
- School of Medicine and Public Health, University of Newcastle, New South Wales, 2308, Australia
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, 2031, Australia
- Faculty of Medicine, University of New South Wales, Sydney, 2052, Australia
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Vidar M Steen
- NORMENT, K.G. Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, 5020, Norway
| | - Julian N Trollor
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, 2031, Australia
- Department of Developmental Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney, 2052, Australia
| | - Steven T Turner
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Cornelia M Van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015, The Netherlands
| | - Arno Villringer
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, 04103, Germany
- Day Clinic for Cognitive Neurology, University Hospital Leipzig, Leipzig, 04103, Germany
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, 21205, MD, USA
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - James F Wilson
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Anil Malhotra
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, 11030, NY, USA
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, 11004, NY, USA
- Department of Psychiatry, Hofstra Northwell School of Medicine, Hempstead, 11549, NY, USA
| | - Andrew M McIntosh
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Division of Psychiatry, University of Edinburgh, Edinburgh, EH10 5HF, UK
| | - Catharine R Gale
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, SO16 6YD, UK
| | - Sudha Seshadri
- Robert and Beverly Lewis Center for Neuroimaging, University of Oregon, Eugene, 97403, OR, USA
- Department of Neurology, Boston University School of Medicine, Boston, 02118, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, 78229, TX, USA
| | - Thomas H Mosley
- Department of Medicine, Division of Geriatrics, University of Mississippi Medical Center, Jackson, 39216, MS, USA
| | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, 77030, TX, USA
| | - Todd Lencz
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, 11030, NY, USA
- Division of Psychiatry, University of Edinburgh, Edinburgh, EH10 5HF, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, EH8 9JZ, UK.
| |
Collapse
|
198
|
Iglesias AI, Mishra A, Vitart V, Bykhovskaya Y, Höhn R, Springelkamp H, Cuellar-Partida G, Gharahkhani P, Bailey JNC, Willoughby CE, Li X, Yazar S, Nag A, Khawaja AP, Polašek O, Siscovick D, Mitchell P, Tham YC, Haines JL, Kearns LS, Hayward C, Shi Y, van Leeuwen EM, Taylor KD, Bonnemaijer P, Rotter JI, Martin NG, Zeller T, Mills RA, Souzeau E, Staffieri SE, Jonas JB, Schmidtmann I, Boutin T, Kang JH, Lucas SEM, Wong TY, Beutel ME, Wilson JF, Uitterlinden AG, Vithana EN, Foster PJ, Hysi PG, Hewitt AW, Khor CC, Pasquale LR, Montgomery GW, Klaver CCW, Aung T, Pfeiffer N, Mackey DA, Hammond CJ, Cheng CY, Craig JE, Rabinowitz YS, Wiggs JL, Burdon KP, van Duijn CM, MacGregor S. Cross-ancestry genome-wide association analysis of corneal thickness strengthens link between complex and Mendelian eye diseases. Nat Commun 2018; 9:1864. [PMID: 29760442 PMCID: PMC5951816 DOI: 10.1038/s41467-018-03646-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 03/02/2018] [Indexed: 12/17/2022] Open
Abstract
Central corneal thickness (CCT) is a highly heritable trait associated with complex eye diseases such as keratoconus and glaucoma. We perform a genome-wide association meta-analysis of CCT and identify 19 novel regions. In addition to adding support for known connective tissue-related pathways, pathway analyses uncover previously unreported gene sets. Remarkably, >20% of the CCT-loci are near or within Mendelian disorder genes. These included FBN1, ADAMTS2 and TGFB2 which associate with connective tissue disorders (Marfan, Ehlers-Danlos and Loeys-Dietz syndromes), and the LUM-DCN-KERA gene complex involved in myopia, corneal dystrophies and cornea plana. Using index CCT-increasing variants, we find a significant inverse correlation in effect sizes between CCT and keratoconus (r = -0.62, P = 5.30 × 10-5) but not between CCT and primary open-angle glaucoma (r = -0.17, P = 0.2). Our findings provide evidence for shared genetic influences between CCT and keratoconus, and implicate candidate genes acting in collagen and extracellular matrix regulation.
Collapse
MESH Headings
- ADAMTS Proteins/genetics
- ADAMTS Proteins/metabolism
- Asian People
- Cornea/abnormalities
- Cornea/metabolism
- Cornea/pathology
- Corneal Diseases/ethnology
- Corneal Diseases/genetics
- Corneal Diseases/metabolism
- Corneal Diseases/pathology
- Corneal Dystrophies, Hereditary/ethnology
- Corneal Dystrophies, Hereditary/genetics
- Corneal Dystrophies, Hereditary/metabolism
- Corneal Dystrophies, Hereditary/pathology
- Decorin/genetics
- Decorin/metabolism
- Ehlers-Danlos Syndrome/ethnology
- Ehlers-Danlos Syndrome/genetics
- Ehlers-Danlos Syndrome/metabolism
- Ehlers-Danlos Syndrome/pathology
- Eye Diseases, Hereditary/ethnology
- Eye Diseases, Hereditary/genetics
- Eye Diseases, Hereditary/metabolism
- Eye Diseases, Hereditary/pathology
- Fibrillin-1/genetics
- Fibrillin-1/metabolism
- Gene Expression
- Genome, Human
- Genome-Wide Association Study
- Glaucoma, Open-Angle/ethnology
- Glaucoma, Open-Angle/genetics
- Glaucoma, Open-Angle/metabolism
- Glaucoma, Open-Angle/pathology
- Humans
- Keratoconus/ethnology
- Keratoconus/genetics
- Keratoconus/metabolism
- Keratoconus/pathology
- Loeys-Dietz Syndrome/ethnology
- Loeys-Dietz Syndrome/genetics
- Loeys-Dietz Syndrome/metabolism
- Loeys-Dietz Syndrome/pathology
- Lumican/genetics
- Lumican/metabolism
- Marfan Syndrome/ethnology
- Marfan Syndrome/genetics
- Marfan Syndrome/metabolism
- Marfan Syndrome/pathology
- Mendelian Randomization Analysis
- Myopia/ethnology
- Myopia/genetics
- Myopia/metabolism
- Myopia/pathology
- Polymorphism, Single Nucleotide
- Proteoglycans/genetics
- Proteoglycans/metabolism
- Quantitative Trait Loci
- Quantitative Trait, Heritable
- Transforming Growth Factor beta2/genetics
- Transforming Growth Factor beta2/metabolism
- White People
Collapse
Affiliation(s)
- Adriana I Iglesias
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands
| | - Aniket Mishra
- University of Bordeaux, Bordeaux Population Health Research Center, INSERM UMR 1219, F-33000, Bordeaux, France
| | - Veronique Vitart
- Institute of Genetics and Molecular Medicine, Medical Research Council Human Genetics Unit, University of Edinburgh, EH42XU, Edinburgh, UK
| | - Yelena Bykhovskaya
- Regenerative Medicine Institute and Department of Surgery, Cedars-Sinai Medical Center, CA 90048, Los Angeles, CA, USA
- Cornea Genetic Eye Institute, CA 90048, Los Angeles, CA, USA
| | - René Höhn
- Department of Ophthalmology, University Medical Center Mainz, 55131, Mainz, Germany
- Department of Ophthalmology, Inselspital, University Hospital Bern, University of Bern, Bern, CH-3010, Switzerland
| | - Henriët Springelkamp
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands
| | - Gabriel Cuellar-Partida
- Statistical Genetics, QIMR Berghofer Medical Research Institute, QLD 4029, Brisbane, Australia
| | - Puya Gharahkhani
- Statistical Genetics, QIMR Berghofer Medical Research Institute, QLD 4029, Brisbane, Australia
| | - Jessica N Cooke Bailey
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, OH 44106, Cleveland, OH, USA
- Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Colin E Willoughby
- Biomedical Sciences Research Institute, Ulster University, BT52 1SA, Belfast, Northern Ireland, UK
- Royal Victoria Hospital, Belfast Health and Social Care Trust, BT12 6BA, Belfast, Northern Ireland, UK
| | - Xiaohui Li
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90509, CA, USA
- Division of Genomic Outcomes, Departments of Pediatrics and Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, CA, USA
| | - Seyhan Yazar
- Institute of Genetics and Molecular Medicine, Medical Research Council Human Genetics Unit, University of Edinburgh, EH42XU, Edinburgh, UK
- Centre for Ophthalmology and Visual Science, University of Western Australia, Lions Eye Institute, WA 6009, Perth, WA, Australia
| | - Abhishek Nag
- Department of Twin Research and Genetic Epidemiology, King's College London, WC2R 2LS, London, UK
| | - Anthony P Khawaja
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, CB2 0SR, Cambridge, UK
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, EC1V 9EL, London, UK
| | - Ozren Polašek
- Faculty of Medicine, University of Split, HR-21000, Split, Croatia
| | - David Siscovick
- Departments of Medicine and Epidemiology and Cardiovascular Health Research Unit, University of Washington, WA 98101, Washington, USA
- The New York Academy of Medicine, NY 10029, New York, NY, USA
| | - Paul Mitchell
- Centre for Vision Research, Department of Ophthalmology and Westmead Institute for Medical Research, University of Sydney, NSW 2145, Sydney, NSW, Australia
| | - Yih Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, 168751, Singapore, Singapore
| | - Jonathan L Haines
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, OH 44106, Cleveland, OH, USA
- Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Lisa S Kearns
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, VIC 3002, East Melbourne, Australia
| | - Caroline Hayward
- Institute of Genetics and Molecular Medicine, Medical Research Council Human Genetics Unit, University of Edinburgh, EH42XU, Edinburgh, UK
| | - Yuan Shi
- Singapore Eye Research Institute, Singapore National Eye Centre, 168751, Singapore, Singapore
| | | | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90509, CA, USA
- Division of Genomic Outcomes, Departments of Pediatrics and Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, CA, USA
| | - Pieter Bonnemaijer
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90509, CA, USA
- Division of Genomic Outcomes, Departments of Pediatrics and Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, CA, USA
| | - Nicholas G Martin
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, QLD 4029, Brisbane, Australia
| | - Tanja Zeller
- Department of General and Interventional Cardiology, University Heart Center Hamburg, 20251, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246, Hamburg, Germany
| | - Richard A Mills
- Department of Ophthalmology, Flinders University, SA 5042, Adelaide, Australia
| | - Emmanuelle Souzeau
- Department of Ophthalmology, Flinders University, SA 5042, Adelaide, Australia
| | - Sandra E Staffieri
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, VIC 3002, East Melbourne, Australia
| | - Jost B Jonas
- Department of Ophthalmology, Medical Faculty Mannheim of the Ruprecht-Karls-University of Heidelberg, 68167, Mannheim, Germany
| | - Irene Schmidtmann
- Institute for Medical Biostatistics, Epidemiology and Informatics, University Medical Center Mainz, 55131, Mainz, Germany
| | - Thibaud Boutin
- Institute of Genetics and Molecular Medicine, Medical Research Council Human Genetics Unit, University of Edinburgh, EH42XU, Edinburgh, UK
| | - Jae H Kang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA 02115, MA, USA
| | - Sionne E M Lucas
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7005, TAS, Australia
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, 168751, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, 169857, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117549, Singapore
| | - Manfred E Beutel
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center Mainz, Mainz, 55131, Germany
| | - James F Wilson
- Institute of Genetics and Molecular Medicine, Medical Research Council Human Genetics Unit, University of Edinburgh, EH42XU, Edinburgh, UK
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, EH16 4UX, Edinburgh, UK
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, 2593 HW, The Hague, The Netherlands
| | - Eranga N Vithana
- Singapore Eye Research Institute, Singapore National Eye Centre, 168751, Singapore, Singapore
| | - Paul J Foster
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, EC1V 9EL, London, UK
| | - Pirro G Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London, WC2R 2LS, London, UK
| | - Alex W Hewitt
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, VIC 3002, East Melbourne, Australia
- School of Medicine, Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7005, TAS, Australia
| | - Chiea Chuen Khor
- Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore
| | - Louis R Pasquale
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA 02115, MA, USA
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear Infirmary, Boston, MA 02114, MA, USA
| | - Grant W Montgomery
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, QLD 4029, Brisbane, Australia
- Institute for Molecular Bioscience, University of Queensland, QLD 4067, Brisbane, Australia
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, 168751, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, 169857, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117549, Singapore
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center Mainz, 55131, Mainz, Germany
| | - David A Mackey
- Centre for Ophthalmology and Visual Science, University of Western Australia, Lions Eye Institute, WA 6009, Perth, WA, Australia
| | - Christopher J Hammond
- Department of Twin Research and Genetic Epidemiology, King's College London, WC2R 2LS, London, UK
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, 168751, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, 169857, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117549, Singapore
| | - Jamie E Craig
- Department of Ophthalmology, Flinders University, SA 5042, Adelaide, Australia
| | - Yaron S Rabinowitz
- Regenerative Medicine Institute and Department of Surgery, Cedars-Sinai Medical Center, CA 90048, Los Angeles, CA, USA
- Cornea Genetic Eye Institute, CA 90048, Los Angeles, CA, USA
| | - Janey L Wiggs
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear Infirmary, Boston, MA 02114, MA, USA
| | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7005, TAS, Australia
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus Medical Center, 3000 CA, Rotterdam, The Netherlands
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, QLD 4029, Brisbane, Australia.
| |
Collapse
|
199
|
Abstract
Advances in omics technologies - such as genomics, transcriptomics, proteomics and metabolomics - have begun to enable personalized medicine at an extraordinarily detailed molecular level. Individually, these technologies have contributed medical advances that have begun to enter clinical practice. However, each technology individually cannot capture the entire biological complexity of most human diseases. Integration of multiple technologies has emerged as an approach to provide a more comprehensive view of biology and disease. In this Review, we discuss the potential for combining diverse types of data and the utility of this approach in human health and disease. We provide examples of data integration to understand, diagnose and inform treatment of diseases, including rare and common diseases as well as cancer and transplant biology. Finally, we discuss technical and other challenges to clinical implementation of integrative omics.
Collapse
Affiliation(s)
- Konrad J Karczewski
- Massachusetts General Hospital, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
200
|
Liu J, Su B. Integrated analysis supports ATXN1 as a schizophrenia risk gene. Schizophr Res 2018; 195:298-305. [PMID: 29055568 DOI: 10.1016/j.schres.2017.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 07/27/2017] [Accepted: 10/08/2017] [Indexed: 12/13/2022]
Abstract
Protein-protein interaction (PPI) is informative in identifying hidden disease risk genes that tend to interact with known risk genes usually working together in the same disease module. With the use of an integrated approach combining PPI information with pathway and expression analysis as well as genome-wide association study (GWAS), we intended to find new risk genes for schizophrenia (SCZ). We showed that ATXN1 was the only direct PPI partner of the know SCZ risk gene ZNF804A, and it also had direct PPIs with other 18 known SCZ risk genes. ATXN1 serves as one of the hub genes in the PPI network containing many known SCZ risk genes, and this network is significantly enriched for the MAPK signaling pathway. Further gene expression analysis indicated that ATXN1 is highly expressed in prefrontal cortex, and SCZ patients had significantly decreased expression compared with healthy controls. Finally, the published GWAS data supports an association of ATXN1 with SCZ as well as other psychiatric disorders though not reaching genome-wide significance. These convergent evidences support ATXN1 as a promising risk gene for SCZ, and the integrated approach serves as a useful tool for dissecting the genetic basis of psychiatric disorders.
Collapse
Affiliation(s)
- Jiewei Liu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, Yunnan, China
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China.
| |
Collapse
|