scispace - formally typeset
Search or ask a question

Showing papers by "Gonçalo R. Abecasis published in 2021"


Journal ArticleDOI
Daniel Taliun1, Daniel N. Harris2, Michael D. Kessler2, Jedidiah Carlson3  +202 moreInstitutions (61)
10 Feb 2021-Nature
TL;DR: The Trans-Omics for Precision Medicine (TOPMed) project as discussed by the authors aims to elucidate the genetic architecture and biology of heart, lung, blood and sleep disorders, with the ultimate goal of improving diagnosis, treatment and prevention of these diseases.
Abstract: The Trans-Omics for Precision Medicine (TOPMed) programme seeks to elucidate the genetic architecture and biology of heart, lung, blood and sleep disorders, with the ultimate goal of improving diagnosis, treatment and prevention of these diseases The initial phases of the programme focused on whole-genome sequencing of individuals with rich phenotypic data and diverse backgrounds Here we describe the TOPMed goals and design as well as the available resources and early insights obtained from the sequence data The resources include a variant browser, a genotype imputation server, and genomic and phenotypic data that are available through dbGaP (Database of Genotypes and Phenotypes)1 In the first 53,831 TOPMed samples, we detected more than 400 million single-nucleotide and insertion or deletion variants after alignment with the reference genome Additional previously undescribed variants were detected through assembly of unmapped reads and customized analysis in highly variable loci Among the more than 400 million detected variants, 97% have frequencies of less than 1% and 46% are singletons that are present in only one individual (53% among unrelated individuals) These rare variants provide insights into mutational processes and recent human evolutionary history The extensive catalogue of genetic variation in TOPMed studies provides unique opportunities for exploring the contributions of rare and noncoding sequence variants to phenotypic variation Furthermore, combining TOPMed haplotypes with modern imputation methods improves the power and reach of genome-wide association studies to include variants down to a frequency of approximately 001% The goals, resources and design of the NHLBI Trans-Omics for Precision Medicine (TOPMed) programme are described, and analyses of rare variants detected in the first 53,831 samples provide insights into mutational processes and recent human evolutionary history

801 citations


Journal ArticleDOI
TL;DR: RegenerIE as mentioned in this paper is a whole-genome regression method based on ridge regression that enables highly parallelized analysis of quantitative and binary traits in biobank-scale data with reduced computational requirements.
Abstract: Genome-wide association analysis of cohorts with thousands of phenotypes is computationally expensive, particularly when accounting for sample relatedness or population structure. Here we present a novel machine-learning method called REGENIE for fitting a whole-genome regression model for quantitative and binary phenotypes that is substantially faster than alternatives in multi-trait analyses while maintaining statistical efficiency. The method naturally accommodates parallel analysis of multiple phenotypes and requires only local segments of the genotype matrix to be loaded in memory, in contrast to existing alternatives, which must load genome-wide matrices into memory. This results in substantial savings in compute time and memory usage. We introduce a fast, approximate Firth logistic regression test for unbalanced case–control phenotypes. The method is ideally suited to take advantage of distributed computing frameworks. We demonstrate the accuracy and computational benefits of this approach using the UK Biobank dataset with up to 407,746 individuals. REGENIE is a whole-genome regression method based on ridge regression that enables highly parallelized analysis of quantitative and binary traits in biobank-scale data with reduced computational requirements.

239 citations


Journal ArticleDOI
18 Oct 2021-Nature
TL;DR: This paper used exome sequencing to explore protein altering variants and their consequences in 454,787 UK Biobank study participants and identified 12 million coding variants, including ~1 million loss-of-function and ~1.8 million deleterious missense variants.
Abstract: A major goal in human genetics is to use natural variation to understand the phenotypic consequences of altering each protein-coding gene in the genome. Here we used exome sequencing1 to explore protein altering variants and their consequences in 454,787 UK Biobank study participants2. We identified 12 million coding variants, including ~1 million loss-of-function and ~1.8 million deleterious missense variants. When these were tested for association with 3,994 health-related traits, we found 564 genes with trait associations at P≤2.18x10-11. Rare variant associations were enriched in GWAS loci, but most (91%) were independent of common variant signals. We discover several risk-increasing associations with traits related to liver disease, eye disease and cancer, among others, as well as novel risk-lowering associations for hypertension (SLC9A3R2), diabetes (MAP3K15, FAM234A) and asthma (SLC27A3). Six genes were associated with brain imaging phenotypes, including two involved in neural development (GBE1, PLD1). 81% of signals available and powered for replication were confirmed in an independent cohort; furthermore, association signals were generally consistent across European, Asian and African ancestry individuals. We illustrate the ability of exome sequencing to identify novel gene-trait associations, elucidate gene function, and pinpoint effector genes underlying GWAS signals at scale.

217 citations


Journal ArticleDOI
Ji Chen1, Ji Chen2, Cassandra N. Spracklen3, Cassandra N. Spracklen4  +475 moreInstitutions (146)
TL;DR: This paper aggregated genome-wide association studies comprising up to 281,416 individuals without diabetes (30% non-European ancestry) for whom fasting glucose, 2-h glucose after an oral glucose challenge, glycated hemoglobin and fasting insulin data were available.
Abstract: Glycemic traits are used to diagnose and monitor type 2 diabetes and cardiometabolic health. To date, most genetic studies of glycemic traits have focused on individuals of European ancestry. Here we aggregated genome-wide association studies comprising up to 281,416 individuals without diabetes (30% non-European ancestry) for whom fasting glucose, 2-h glucose after an oral glucose challenge, glycated hemoglobin and fasting insulin data were available. Trans-ancestry and single-ancestry meta-analyses identified 242 loci (99 novel; P < 5 × 10-8), 80% of which had no significant evidence of between-ancestry heterogeneity. Analyses restricted to individuals of European ancestry with equivalent sample size would have led to 24 fewer new loci. Compared with single-ancestry analyses, equivalent-sized trans-ancestry fine-mapping reduced the number of estimated variants in 99% credible sets by a median of 37.5%. Genomic-feature, gene-expression and gene-set analyses revealed distinct biological signatures for each trait, highlighting different underlying biological pathways. Our results increase our understanding of diabetes pathophysiology by using trans-ancestry studies for improved power and resolution.

178 citations


Journal ArticleDOI
02 Jul 2021-Science
TL;DR: In this paper, the authors sequenced the exomes of 645,626 individuals from the United Kingdom, the United States, and Mexico and estimated associations of rare coding variants with body mass index (BMI).
Abstract: Large-scale human exome sequencing can identify rare protein-coding variants with a large impact on complex traits such as body adiposity. We sequenced the exomes of 645,626 individuals from the United Kingdom, the United States, and Mexico and estimated associations of rare coding variants with body mass index (BMI). We identified 16 genes with an exome-wide significant association with BMI, including those encoding five brain-expressed G protein-coupled receptors (CALCR, MC4R, GIPR, GPR151, and GPR75). Protein-truncating variants in GPR75 were observed in ~4/10,000 sequenced individuals and were associated with 1.8 kilograms per square meter lower BMI and 54% lower odds of obesity in the heterozygous state. Knock out of Gpr75 in mice resulted in resistance to weight gain and improved glycemic control in a high-fat diet model. Inhibition of GPR75 may provide a therapeutic strategy for obesity.

99 citations


Journal ArticleDOI
TL;DR: LocusZoom as mentioned in this paper is a JavaScript library for creating interactive web-based visualizations of genetic association study results, which can display one or more traits in the context of relevant biological data (such as gene models and other genomic annotation), and allows interactive refinement of analysis models (by selecting linkage disequilibrium reference panels, identifying sets of likely causal variants, or comparisons to the GWAS catalog).
Abstract: LocusZoom.js is a JavaScript library for creating interactive web-based visualizations of genetic association study results. It can display one or more traits in the context of relevant biological data (such as gene models and other genomic annotation), and allows interactive refinement of analysis models (by selecting linkage disequilibrium reference panels, identifying sets of likely causal variants, or comparisons to the GWAS catalog). It can be embedded in web pages to enable data sharing and exploration. Views can be customized and extended to display other data types such as phenome-wide association study (PheWAS) results, chromatin co-accessibility, or eQTL measurements. A new web upload service harmonizes datasets, adds annotations, and makes it easy to explore user-provided result sets. Availability LocusZoom.js is open-source software under a permissive MIT license. Code and documentation are available at: https://github.com/statgen/locuszoom/. Installable packages for all versions are also distributed via NPM. Additional features are provided as standalone libraries to promote reuse. Use with your own GWAS results at https://my.locuszoom.org/. Supplementary information Supplementary data are available at Bioinformatics online.

91 citations


Posted ContentDOI
04 Jan 2021-bioRxiv
TL;DR: LocusZoom as discussed by the authors is a JavaScript library for creating interactive web-based visualizations of genetic association study results, which can display one or more traits in the context of relevant biological data (such as gene models and other genomic annotation), and allows interactive refinement of analysis models (by selecting linkage disequilibrium reference panels, identifying sets of likely causal variants, or comparisons to the GWAS catalog).
Abstract: LocusZoom.js is a JavaScript library for creating interactive web-based visualizations of genetic association study results. It can display one or more traits in the context of relevant biological data (such as gene models and other genomic annotation), and allows interactive refinement of analysis models (by selecting linkage disequilibrium reference panels, identifying sets of likely causal variants, or comparisons to the GWAS catalog). It can be embedded in web pages to enable data sharing and exploration. Views can be customized and extended to display other data types such as phenome-wide association study (PheWAS) results, chromatin co-accessibility, or eQTL measurements. A new web upload service harmonizes datasets, adds annotations, and makes it easy to explore user-provided result sets. Availability LocusZoom.js is open-source software under a permissive MIT license. Code and documentation are available at: https://github.com/statgen/locuszoom/. Installable packages are also distributed via NPM. Additional features are provided as standalone libraries to promote reuse. Use with your own GWAS results at https://my.locuszoom.org/. Contact locuszoom@googlegroups.com

71 citations


Journal ArticleDOI
Vasiliki Lagou1, Vasiliki Lagou2, Reedik Mägi3, Hottenga J-J.4  +251 moreInstitutions (89)
TL;DR: In this paper, the authors assess sex-dimorphic (73,089/50,404 women and 67,506/47,806 men) and sex-combined (151,188/105,056 individuals) fasting glucose/fasting insulin genetic effects via genome-wide association study meta-analyses.
Abstract: Differences between sexes contribute to variation in the levels of fasting glucose and insulin. Epidemiological studies established a higher prevalence of impaired fasting glucose in men and impaired glucose tolerance in women, however, the genetic component underlying this phenomenon is not established. We assess sex-dimorphic (73,089/50,404 women and 67,506/47,806 men) and sex-combined (151,188/105,056 individuals) fasting glucose/fasting insulin genetic effects via genome-wide association study meta-analyses in individuals of European descent without diabetes. Here we report sex dimorphism in allelic effects on fasting insulin at IRS1 and ZNF12 loci, the latter showing higher RNA expression in whole blood in women compared to men. We also observe sex-homogeneous effects on fasting glucose at seven novel loci. Fasting insulin in women shows stronger genetic correlations than in men with waist-to-hip ratio and anorexia nervosa. Furthermore, waist-to-hip ratio is causally related to insulin resistance in women, but not in men. These results position dissection of metabolic and glycemic health sex dimorphism as a steppingstone for understanding differences in genetic effects between women and men in related phenotypes.

69 citations


Journal ArticleDOI
TL;DR: In this article, the authors used exome sequence data to investigate associations between rare genetic variants and seven COVID-19 outcomes in 586,157 individuals, including 20,952 with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), a respiratory illness causing hospitalization or death.
Abstract: Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), a respiratory illness that can result in hospitalization or death. We used exome sequence data to investigate associations between rare genetic variants and seven COVID-19 outcomes in 586,157 individuals, including 20,952 with COVID-19. After accounting for multiple testing, we did not identify any clear associations with rare variants either exome wide or when specifically focusing on (1) 13 interferon pathway genes in which rare deleterious variants have been reported in individuals with severe COVID-19, (2) 281 genes located in susceptibility loci identified by the COVID-19 Host Genetics Initiative, or (3) 32 additional genes of immunologic relevance and/or therapeutic potential. Our analyses indicate there are no significant associations with rare protein-coding variants with detectable effect sizes at our current sample sizes. Analyses will be updated as additional data become available, and results are publicly available through the Regeneron Genetics Center COVID-19 Results Browser.

59 citations


Journal ArticleDOI
TL;DR: In this article, the authors conducted a genome-wide association study with 53,400 cases and 433,201 controls and replicated significant associations in a 23andMe panel (205,252 cases and 1,384,055 controls).
Abstract: Irritable bowel syndrome (IBS) results from disordered brain-gut interactions. Identifying susceptibility genes could highlight the underlying pathophysiological mechanisms. We designed a digestive health questionnaire for UK Biobank and combined identified cases with IBS with independent cohorts. We conducted a genome-wide association study with 53,400 cases and 433,201 controls and replicated significant associations in a 23andMe panel (205,252 cases and 1,384,055 controls). Our study identified and confirmed six genetic susceptibility loci for IBS. Implicated genes included NCAM1, CADM2, PHF2/FAM120A, DOCK9, CKAP2/TPTE2P3 and BAG6. The first four are associated with mood and anxiety disorders, expressed in the nervous system, or both. Mirroring this, we also found strong genome-wide correlation between the risk of IBS and anxiety, neuroticism and depression (rg > 0.5). Additional analyses suggested this arises due to shared pathogenic pathways rather than, for example, anxiety causing abdominal symptoms. Implicated mechanisms require further exploration to help understand the altered brain-gut interactions underlying IBS.

58 citations


Journal ArticleDOI
TL;DR: In this paper, the association between CHIP, epigenetic clocks, and health outcomes was investigated. And the association was strongly associated with epigenetic age acceleration, defined as the residual after regressing epigenetic clock age on chronological age, in several clocks, ranging from 1.31 years to 5.5 years.
Abstract: Clonal hematopoiesis of indeterminate potential (CHIP) is a common precursor state for blood cancers that most frequently occurs due to mutations in the DNA-methylation modifying enzymes DNMT3A or TET2. We used DNA-methylation array and whole-genome sequencing data from four cohorts together comprising 5522 persons to study the association between CHIP, epigenetic clocks, and health outcomes. CHIP was strongly associated with epigenetic age acceleration, defined as the residual after regressing epigenetic clock age on chronological age, in several clocks, ranging from 1.31 years (GrimAge, p 0 in both Hannum and GrimAge (referred to as AgeAccelHG+). This group was at high risk of all-cause mortality (hazard ratio 2.90, p < 4.1 × 10-8 ) and coronary heart disease (CHD) (hazard ratio 3.24, p < 9.3 × 10-6 ) compared to those who were CHIP-/AgeAccelHG-. In contrast, the other ~60% of CHIP carriers who were AgeAccelHG- were not at increased risk of these outcomes. In summary, CHIP is strongly linked to age acceleration in multiple clocks, and the combination of CHIP and epigenetic aging may be used to identify a population at high risk for adverse outcomes and who may be a target for clinical interventions.

Journal ArticleDOI
Mathias Gorski1, Mathias Gorski2, Bettina Jung2, Yong Li3  +189 moreInstitutions (57)
TL;DR: The identified loci for rapid kidney function decline may help prioritize therapeutic targets and identify mechanisms and individuals at risk for sustained deterioration of kidney function.

Journal ArticleDOI
Xingyi Guo1, Weiqiang Lin2, Wanqing Wen1, Jeroen R. Huyghe3, Stephanie A. Bien3, Qiuyin Cai1, Tabitha A. Harrison3, Zhishan Chen1, Conghui Qu3, Jiandong Bao1, Jirong Long1, Yuan Yuan2, Fangqin Wang2, Mengqiu Bai2, Gonçalo R. Abecasis4, Demetrius Albanes5, Sonja I. Berndt5, Stéphane Bézieau, D. Timothy Bishop6, Hermann Brenner7, Stephan Buch8, Andrea N. Burnett-Hartman9, Peter T. Campbell10, Sergi Castellví-Bel11, Andrew T. Chan12, Andrew T. Chan13, Jenny Chang-Claude7, Jenny Chang-Claude14, Stephen J. Chanock5, Sang-Hee Cho15, David V. Conti16, Albert de la Chapelle17, Edith J. M. Feskens18, Steven Gallinger19, Graham G. Giles20, Graham G. Giles21, Phyllis J. Goodman3, Andrea Gsur, Mark A. Guinter10, Marc J. Gunter22, Jochen Hampe8, Heather Hampel17, Richard B. Hayes23, Michael Hoffmeister7, Ellen Kampman18, Hyun Min Kang4, Temitope O. Keku24, Hyeong Rok Kim15, Loic Le Marchand25, Soo-Chin Lee26, Christopher I. Li3, Li Li27, Annika Lindblom28, Annika Lindblom29, Noralane M. Lindor30, Roger L. Milne20, Roger L. Milne21, Victor Moreno, Neil Murphy10, Polly A. Newcomb31, Polly A. Newcomb3, Deborah A. Nickerson31, Kenneth Offit32, Kenneth Offit33, Rachel Pearlman17, Paul D.P. Pharoah34, Elizabeth A. Platz35, John D. Potter3, Gad Rennert36, Lori C. Sakoda9, Lori C. Sakoda3, Clemens Schafmayer, Stephanie L. Schmit, Robert E. Schoen37, Fredrick R. Schumacher38, Martha L. Slattery39, Yu Ru Su3, Catherine M. Tangen3, Cornelia M. Ulrich40, Fränzel J.B. Van Duijnhoven18, Bethany Van Guelpen41, Kala Visvanathan35, Pavel Vodicka42, Pavel Vodicka43, Ludmila Vodickova43, Ludmila Vodickova42, Veronika Vymetalkova42, Veronika Vymetalkova43, Xiaoliang Wang3, Emily White31, Emily White3, Alicja Wolk28, Michael O. Woods44, Graham Casey27, Li Hsu3, Mark A. Jenkins20, Stephen B. Gruber16, Ulrike Peters3, Ulrike Peters31, Wei Zheng1 
TL;DR: A transcriptome-wide association study to identify putative susceptibility genes for colorectal cancer risk identified 25 genes and provides new insight into the biological mechanisms underlying CRC development.

Journal ArticleDOI
25 Feb 2021-Gut
TL;DR: In this article, the authors identify new anatomical subsite-specific risk loci for colorectal cancer and characterised effect heterogeneity at CRC risk locis using multinomial modeling.
Abstract: OBJECTIVE: An understanding of the etiologic heterogeneity of colorectal cancer (CRC) is critical for improving precision prevention, including individualized screening recommendations and the discovery of novel drug targets and repurposable drug candidates for chemoprevention. Known differences in molecular characteristics and environmental risk factors among tumors arising in different locations of the colorectum suggest partly distinct mechanisms of carcinogenesis. The extent to which the contribution of inherited genetic risk factors for CRC differs by anatomical subsite of the primary tumor has not been examined. DESIGN: To identify new anatomical subsite-specific risk loci, we performed genome-wide association study (GWAS) meta-analyses including data of 48 214 CRC cases and 64 159 controls of European ancestry. We characterised effect heterogeneity at CRC risk loci using multinomial modelling. RESULTS: We identified 13 loci that reached genome-wide significance (p<5×10-8) and that were not reported by previous GWASs for overall CRC risk. Multiple lines of evidence support candidate genes at several of these loci. We detected substantial heterogeneity between anatomical subsites. Just over half (61) of 109 known and new risk variants showed no evidence for heterogeneity. In contrast, 22 variants showed association with distal CRC (including rectal cancer), but no evidence for association or an attenuated association with proximal CRC. For two loci, there was strong evidence for effects confined to proximal colon cancer. CONCLUSION: Genetic architectures of proximal and distal CRC are partly distinct. Studies of risk factors and mechanisms of carcinogenesis, and precision prevention strategies should take into consideration the anatomical subsite of the tumour.

Journal ArticleDOI
TL;DR: These findings provide tentative evidence that daytime napping may reduce AD risk, and are the first MR study of multiple self-report and objective sleep traits on AD risk.
Abstract: Background It is established that Alzheimer's disease (AD) patients experience sleep disruption. However, it remains unknown whether disruption in the quantity, quality or timing of sleep is a risk factor for the onset of AD. Methods We used the largest published genome-wide association studies of self-reported and accelerometer-measured sleep traits (chronotype, duration, fragmentation, insomnia, daytime napping and daytime sleepiness), and AD. Mendelian randomization (MR) was used to estimate the causal effect of self-reported and accelerometer-measured sleep parameters on AD risk. Results Overall, there was little evidence to support a causal effect of sleep traits on AD risk. There was some suggestive evidence that self-reported daytime napping was associated with lower AD risk [odds ratio (OR): 0.70, 95% confidence interval (CI): 0.50-0.99). Some other sleep traits (accelerometer-measured 'eveningness' and sleep duration, and self-reported daytime sleepiness) had ORs of a similar magnitude to daytime napping, but were less precisely estimated. Conclusions Overall, we found very limited evidence to support a causal effect of sleep traits on AD risk. Our findings provide tentative evidence that daytime napping may reduce AD risk. Given that this is the first MR study of multiple self-report and objective sleep traits on AD risk, findings should be replicated using independent samples when such data become available.

Journal ArticleDOI
13 Oct 2021
TL;DR: The observed decline in mtDNA CN after 65 years of age may be a key to understanding age-related diseases.
Abstract: Summary Mitochondrial DNA (mtDNA) is present in multiple copies in human cells. We evaluated cross-sectional associations of whole-blood mtDNA copy number (CN) with several cardiometabolic disease traits in 408,361 participants of multiple ancestries in TOPMed and UK Biobank. Age showed a threshold association with mtDNA CN: each additional 10 years of age was associated with a 0.03 SD higher level of mtDNA CN (p = 0.0014) among younger participants (younger than 65 years) versus a 0.14 SD lower level of mtDNA CN (p = 1.82 × 10−13) among older participants (65 years and older). At lower mtDNA CN levels, we found age-independent associations with increased odds of obesity (p = 5.6 × 10−238), hypertension (p = 2.8 × 10−50), diabetes (p = 3.6 × 10−7), and hyperlipidemia (p = 6.3 × 10−56). The observed decline in mtDNA CN after 65 years of age may be a key to understanding age-related diseases.

Journal ArticleDOI
Alexander G. Bick, Joshua S. Weinstock, Satish K. Nandakumar, Charles P. Fulco, Erik L. Bao, Seyedeh M. Zekavat, Mindy D. Szeto, Xiaotian Liao, Matthew Leventhal, Joseph Nasser, Kyle Chang, Cecelia A. Laurie, Bala Bharathi Burugula, Christopher J. Gibson, Abhishek Niroula, Amy E. Lin, Margaret A. Taub, François Aguet, Kristin G. Ardlie, Braxton D. Mitchell, Kathleen C. Barnes, Arden Moscati, Myriam Fornage, Susan Redline, Bruce M. Psaty, Edwin K. Silverman, Scott T. Weiss, Nicholette D. Palmer, Ramachandran S. Vasan, Esteban G. Burchard, Sharon L.R. Kardia, Jiang He, Robert C. Kaplan, Nicholas L. Smith, Donna K. Arnett, David A. Schwartz, Adolfo Correa, Mariza de Andrade, Xiuqing Guo, Barbara A. Konkle, Brian Custer, Juan M. Peralta, Hongsheng Gui, Deborah A. Meyers, Stephen T. McGarvey, Ida Yii-Der Chen, M. Benjamin Shoemaker, Patricia A. Peyser, Jai G. Broome, Stephanie M. Gogarten, Fei Fei Wang, Quenna Wong, May E. Montasser, Michelle Daya, Eimear E. Kenny, Kari E. North, Lenore J. Launer, Brian E. Cade, Joshua C. Bis, Michael H. Cho, Jessica Lasky-Su, Donald W. Bowden, L. Adrienne Cupples, Angel C.Y. Mak, Lewis C. Becker, Jennifer A. Smith, Tanika N. Kelly, Stella Aslibekyan, Susan R. Heckbert, Hemant K. Tiwari, Ivana V. Yang, John A. Heit, Steven A. Lubitz, Jill M. Johnsen, Joanne E. Curran, Sally E. Wenzel, Daniel E. Weeks, Dabeeru C. Rao, Dawood Darbar, Jee-Young Moon, Russell P. Tracy, Erin J Buth, Nicholas Rafaels, Ruth J. F. Loos, Peter Durda, Yongmei Liu, Lifang Hou, Jiwon Lee, Priyadarshini Kachroo, Barry I. Freedman, Daniel Levy, Lawrence F. Bielak, James E. Hixson, James S. Floyd, Eric A. Whitsel, Patrick T. Ellinor, Marguerite R. Irvin, Tasha E. Fingerlin, Laura M. Raffield, Sebastian M. Armasu, Marsha M. Wheeler, Ester Cerdeira Sabino, John Blangero, L. Keoki Williams, Bruce D. Levy, Wayne Huey-Herng Sheu, Dan M. Roden, Eric Boerwinkle, JoAnn E. Manson, Rasika A. Mathias, Pinkal Desai, Kent D. Taylor, Andrew D. Johnson, Paul L. Auer, Charles Kooperberg, Cathy C. Laurie, Thomas W. Blackwell, Albert V. Smith, Hongyu Zhao, Ethan M. Lange, Leslie A. Lange, Stephen S. Rich, Jerome I. Rotter, James G. Wilson, Paul Scheet, Jacob O. Kitzman, Eric S. Lander, Jesse M. Engreitz, Benjamin L. Ebert, Alexander P. Reiner, Siddhartha Jaiswal, Gonçalo R. Abecasis, Vijay G. Sankaran, Sekar Kathiresan, Pradeep Natarajan 
01 Jan 2021-Nature
TL;DR: High-coverage whole genome sequences from 97,691 participants of diverse ancestries in the NHLBI TOPMed program are analyzed and three genetic loci associated with CHIP status are identified, including one locus at TET2 that was African ancestry specific.
Abstract: Age is the dominant risk factor for most chronic human diseases; yet the mechanisms by which aging confers this risk are largely unknown.1 Recently, the age-related acquisition of somatic mutations in regenerating hematopoietic stem cell populations leading to clonal expansion was associated with both hematologic cancer 2–4 and coronary heart disease5, a phenomenon termed ‘Clonal Hematopoiesis of Indeterminate Potential’ (CHIP).6 Simultaneous germline and somatic whole genome sequence analysis now provides the opportunity to identify root causes of CHIP. Here, we analyze high-coverage whole genome sequences from 97,691 participants of diverse ancestries in the NHLBI TOPMed program and identify 4,229 individuals with CHIP. We identify associations with blood cell, lipid, and inflammatory traits specific to different CHIP genes. Association of a genome-wide set of germline genetic variants identified three genetic loci associated with CHIP status, including one locus at TET2 that was African ancestry specific. In silico-informed in vitro evaluation of the TET2 germline locus identified a causal variant that disrupts a TET2 distal enhancer resulting in increased hematopoietic stem cell self-renewal. Overall, we observe that germline genetic variation shapes hematopoietic stem cell function Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use:http://www.nature.com/authors/editorial_policies/license.html#terms Please address correspondence to: Pradeep Natarajan, MD MMSc, 185 Cambridge St, CPZN 3.184, Boston, MA 02114 USA, pradeep@broadinstitute.org, Twitter: @pnatarajanmd, Sekar Kathiresan, MD, 75 Ames St, Cambridge, MA 02139 USA, sekar@broadinstitute.org, Twitter: @skathire. ‡Present Address: Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN. AUTHOR CONTRIBUTIONS A.G.B., P.N. and S.K. conceived the study. A.G.B. and J.S.W. performed the germline and somatic whole genome sequence analyses. C.P.F., E.L.B., S.M.Z, M.D.S., M.J.L., J.N., K.C., C.J.G., A.E.L., B.B.B., P.S., J.K., J.M.E., A.P.R, B.L.E., and S.J. performed additional bioinformatic analyses. S.K.N., X.L. and V.G.S. experimentally characterized the TET2 locus. M.A.T., F.A., K.A., B.D.M, K.C.B., A.M., M.F., S.R., B.M.P., E.K.S., S.T.W., N.D.P., R.S.V., E.G.B., S.L.R.K., J.H., R.C.K., N.L.S., D.K.A., D.A.S., A.C., M.d.A., X.G.,B.A.K., B.C., J.M.P., H.G., D.A.M., S.T.M., I.Y., M.B.S., P.A.P., J.G.B., S.M.G., F.F.W., Q.W., M.E.M., M.D., E.E.K., K.E.N., L.J.L., B.E.C., J.C.B., M.H.C., J.L.S., D.W.B., L.A.C., A.C.M., L.C.B., J.A.S., T.N.K, S.A., S.R.H., H.K.T.,I.V.Y., J.A.H., S.L., J.M.J., J.E.C., S.E.W., D.E.W., D.C.R., D.D., J.Y.M., R.P.T., E.J.B., N.R., R.J.F.L., P.D., Y.L., L.H., J.L., P.K., B.I.F., D.L., L.F.B., J.E.H., J.S.F., E.A.W., P.T.E., M.R.I., T.E.F., L.M.R., S.M.A., M.M.W., E.C.S., J.B., L.K.W., B.D.L., W.H.S., D.M.R., E.B., J.E.M., R.A.M., P.D., K.D.T., A.D.J., P.L.A., C.K., C.C.L., T.W.B., A.V.S., H.Z.,, E.L., L.L., S.S.R., J.I.R., J.G.W., P.S., J.O.K., E.S.L., J.M.E., and G.A. contributed to sample acquisition, DNA sequencing and phenotypic curation for the NHLBI TOPMed constituent cohorts analyzed here. A.G.B., J.S.W., S.K. and P.N. wrote the manuscript with input from all authors. *These individuals contributed equally to this work #These individuals jointly supervised this work †A list of authors and their affiliations appears at the end of the paper DATA AVAILABILITY Individual whole-genome sequence data for TOPMed whole genomes, individual-level harmonized phenotypes, harmonized germline variant call sets, the CHIP somatic variant call sets, RNA-Seq and peripheral blood methylation data used in this analysis are available through restricted access via the dbGaP. Accession numbers for these datasets are provided in Supplementary Table 1. Summary-level genotype data are available through the BRAVO browser (https://bravo.sph.umich.edu/). Full GWAS summary statistics are available for general research use through controlled access at dbGaP accession phs001974: NHLBI TOPMed: Genomic Summary Results for the Trans-Omics for Precision Medicine Program. A subset of the TOPMed cohorts analyzed here are based on sensitive populations, precluding public sharing of full genomic summary results. HHS Public Access Author manuscript Nature. Author manuscript; available in PMC 2021 April 14. Published in final edited form as: Nature. 2020 October ; 586(7831): 763–768. doi:10.1038/s41586-020-2819-2. A uhor M anscript

Journal ArticleDOI
Yao Hu1, Adrienne M. Stilp2, Caitlin P. McHugh2, Shuquan Rao3, Deepti Jain2, Xiuwen Zheng2, John Lane4, Sébastian Méric de Bellefon5, Laura M. Raffield6, Ming-Huei Chen7, Lisa R. Yanek8, Marsha M. Wheeler2, Yao Yao3, Chunyan Ren3, Jai G. Broome2, Jee-Young Moon9, Paul S. de Vries10, Brian D. Hobbs11, Quan Sun6, Praveen Surendran, Jennifer A. Brody2, Thomas W. Blackwell12, Hélène Choquet13, Kathleen A. Ryan14, Ravindranath Duggirala15, Nancy L. Heard-Costa7, Nancy L. Heard-Costa6, Zhe Wang16, Nathalie Chami16, Michael Preuss16, Nancy Min17, Lynette Ekunwe17, Leslie A. Lange18, Mary Cushman19, Nauder Faraday8, Joanne E. Curran15, Laura Almasy20, Kousik Kundu21, Kousik Kundu22, Albert V. Smith12, Stacey Gabriel23, Jerome I. Rotter24, Myriam Fornage10, Donald M. Lloyd-Jones25, Ramachandran S. Vasan7, Nicholas L. Smith2, Nicholas L. Smith13, Nicholas L. Smith26, Kari E. North6, Eric Boerwinkle10, Lewis C. Becker8, Joshua P. Lewis14, Gonçalo R. Abecasis12, Lifang Hou25, Jeffrey R. O'Connell14, Alanna C. Morrison10, Terri H. Beaty27, Robert C. Kaplan9, Adolfo Correa17, John Blangero15, Eric Jorgenson13, Bruce M. Psaty2, Bruce M. Psaty13, Charles Kooperberg1, Russell T. Walton3, Benjamin P. Kleinstiver3, Hua Tang28, Ruth J. F. Loos16, Nicole Soranzo, Adam S. Butterworth, Deborah A. Nickerson2, Stephen S. Rich29, Braxton D. Mitchell14, Andrew D. Johnson7, Paul L. Auer30, Yun Li6, Rasika A. Mathias8, Guillaume Lettre5, Guillaume Lettre31, Nathan Pankratz4, Cathy C. Laurie2, Cecelia A. Laurie2, Daniel E. Bauer3, Matthew P. Conomos2, Alexander P. Reiner2 
TL;DR: In this paper, the authors leveraged whole-genome sequencing (WGS) data in up to 62,653 ethnically diverse participants from the NHLBI Trans-Omics for Precision Medicine (TOPMed) program and assessed statistical association of variants with seven red blood cell (RBC) quantitative traits.
Abstract: Summary Whole-genome sequencing (WGS), a powerful tool for detecting novel coding and non-coding disease-causing variants, has largely been applied to clinical diagnosis of inherited disorders. Here we leveraged WGS data in up to 62,653 ethnically diverse participants from the NHLBI Trans-Omics for Precision Medicine (TOPMed) program and assessed statistical association of variants with seven red blood cell (RBC) quantitative traits. We discovered 14 single variant-RBC trait associations at 12 genomic loci, which have not been reported previously. Several of the RBC trait-variant associations (RPN1, ELL2, MIDN, HBB, HBA1, PIEZO1, and G6PD) were replicated in independent GWAS datasets imputed to the TOPMed reference panel. Most of these discovered variants are rare/low frequency, and several are observed disproportionately among non-European Ancestry (African, Hispanic/Latino, or East Asian) populations. We identified a 3 bp indel p.Lys2169del (g.88717175_88717177TCT[4]) (common only in the Ashkenazi Jewish population) of PIEZO1, a gene responsible for the Mendelian red cell disorder hereditary xerocytosis (MIM: 194380 ), associated with higher mean corpuscular hemoglobin concentration (MCHC). In stepwise conditional analysis and in gene-based rare variant aggregated association analysis, we identified several of the variants in HBB, HBA1, TMPRSS6, and G6PD that represent the carrier state for known coding, promoter, or splice site loss-of-function variants that cause inherited RBC disorders. Finally, we applied base and nuclease editing to demonstrate that the sentinel variant rs112097551 (nearest gene RPN1) acts through a cis-regulatory element that exerts long-range control of the gene RUVBL1 which is essential for hematopoiesis. Together, these results demonstrate the utility of WGS in ethnically diverse population-based samples and gene editing for expanding knowledge of the genetic architecture of quantitative hematologic traits and suggest a continuum between complex trait and Mendelian red cell disorders.

Journal ArticleDOI
TL;DR: In this paper, a 2-sample mendelian randomization was used to assess whether smoking, alcohol consumption, blood pressure, body mass index, and glycemic traits are associated with increased risk of advanced age-related macular degeneration.
Abstract: Importance Advanced age-related macular degeneration (AMD) is a leading cause of blindness in Western countries. Causal, modifiable risk factors need to be identified to develop preventive measures for advanced AMD. Objective To assess whether smoking, alcohol consumption, blood pressure, body mass index, and glycemic traits are associated with increased risk of advanced AMD. Design, Setting, Participants This study used 2-sample mendelian randomization. Genetic instruments composed of variants associated with risk factors at genome-wide significance (P < 5 × 10-8) were obtained from published genome-wide association studies. Summary-level statistics for these instruments were obtained for advanced AMD from the International AMD Genomics Consortium 2016 data set, which consisted of 16 144 individuals with AMD and 17 832 control individuals. Data were analyzed from July 2020 to September 2021. Exposures Smoking initiation, smoking cessation, lifetime smoking, age at smoking initiation, alcoholic drinks per week, body mass index, systolic and diastolic blood pressure, type 2 diabetes, glycated hemoglobin, fasting glucose, and fasting insulin. Main Outcomes and Measures Advanced AMD and its subtypes, geographic atrophy (GA), and neovascular AMD. Results A 1-SD increase in logodds of genetically predicted smoking initiation was associated with higher risk of advanced AMD (odds ratio [OR], 1.26; 95% CI, 1.13-1.40; P < .001), while a 1-SD increase in logodds of genetically predicted smoking cessation (former vs current smoking) was associated with lower risk of advanced AMD (OR, 0.66; 95% CI, 0.50-0.87; P = .003). Genetically predicted increased lifetime smoking was associated with increased risk of advanced AMD (OR per 1-SD increase in lifetime smoking behavior, 1.32; 95% CI, 1.09-1.59; P = .004). Genetically predicted alcohol consumption was associated with higher risk of GA (OR per 1-SD increase of log-transformed alcoholic drinks per week, 2.70; 95% CI, 1.48-4.94; P = .001). There was insufficient evidence to suggest that genetically predicted blood pressure, body mass index, and glycemic traits were associated with advanced AMD. Conclusions and Relevance This study provides genetic evidence that increased alcohol intake may be a causal risk factor for GA. As there are currently no known treatments for GA, this finding has important public health implications. These results also support previous observational studies associating smoking behavior with risk of advanced AMD, thus reinforcing existing public health messages regarding the risk of blindness associated with smoking.

Journal ArticleDOI
TL;DR: The first whole-genome sequence analysis of SDB highlighted associations in genes that modulate lung development, inflammation, respiratory rhythmogenesis and HIF1A-mediated hypoxic response.
Abstract: Author(s): Cade, Brian E; Lee, Jiwon; Sofer, Tamar; Wang, Heming; Zhang, Man; Chen, Han; Gharib, Sina A; Gottlieb, Daniel J; Guo, Xiuqing; Lane, Jacqueline M; Liang, Jingjing; Lin, Xihong; Mei, Hao; Patel, Sanjay R; Purcell, Shaun M; Saxena, Richa; Shah, Neomi A; Evans, Daniel S; Hanis, Craig L; Hillman, David R; Mukherjee, Sutapa; Palmer, Lyle J; Stone, Katie L; Tranah, Gregory J; NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium; Abecasis, Goncalo R; Boerwinkle, Eric A; Correa, Adolfo; Cupples, L Adrienne; Kaplan, Robert C; Nickerson, Deborah A; North, Kari E; Psaty, Bruce M; Rotter, Jerome I; Rich, Stephen S; Tracy, Russell P; Vasan, Ramachandran S; Wilson, James G; Zhu, Xiaofeng; Redline, Susan; TOPMed Sleep Working Group | Abstract: BackgroundSleep-disordered breathing is a common disorder associated with significant morbidity. The genetic architecture of sleep-disordered breathing remains poorly understood. Through the NHLBI Trans-Omics for Precision Medicine (TOPMed) program, we performed the first whole-genome sequence analysis of sleep-disordered breathing.MethodsThe study sample was comprised of 7988 individuals of diverse ancestry. Common-variant and pathway analyses included an additional 13,257 individuals. We examined five complementary traits describing different aspects of sleep-disordered breathing: the apnea-hypopnea index, average oxyhemoglobin desaturation per event, average and minimum oxyhemoglobin saturation across the sleep episode, and the percentage of sleep with oxyhemoglobin saturation l 90%. We adjusted for age, sex, BMI, study, and family structure using MMSKAT and EMMAX mixed linear model approaches. Additional bioinformatics analyses were performed with MetaXcan, GIGSEA, and ReMap.ResultsWe identified a multi-ethnic set-based rare-variant association (p = 3.48 × 10-8) on chromosome X with ARMCX3. Additional rare-variant associations include ARMCX3-AS1, MRPS33, and C16orf90. Novel common-variant loci were identified in the NRG1 and SLC45A2 regions, and previously associated loci in the IL18RAP and ATP2B4 regions were associated with novel phenotypes. Transcription factor binding site enrichment identified associations with genes implicated with respiratory and craniofacial traits. Additional analyses identified significantly associated pathways.ConclusionsWe have identified the first gene-based rare-variant associations with objectively measured sleep-disordered breathing traits. Our results increase the understanding of the genetic architecture of sleep-disordered breathing and highlight associations in genes that modulate lung development, inflammation, respiratory rhythmogenesis, and HIF1A-mediated hypoxic response.

Journal ArticleDOI
TL;DR: A splice acceptor polymorphism appears to be a causal variant for asthma at the 17q12-21.1 locus that appears to have the same magnitude of effect in individuals of African and European descent.
Abstract: Rationale: The 17q12-21.1 locus is one of the most highly replicated genetic associations with asthma. Individuals of African descent have lower LD in this region, which could facilitate identifyin...

Journal ArticleDOI
Xiaoming Jia1, Fernando S. Goes2, Adam E. Locke3, Duncan Palmer4, Weiqing Wang5, Sarah Cohen-Woods6, Sarah Cohen-Woods7, Giulio Genovese4, Anne U. Jackson8, Chen Jiang9, Mark N. Kvale1, Niamh Mullins5, Hoang T. Nguyen5, Mehdi Pirooznia, Margarita Rivera10, Margarita Rivera6, Douglas M. Ruderfer11, Ling Shen9, Khanh K. Thai9, Matthew Zawistowski8, Yongwen Zhuang8, Gonçalo R. Abecasis8, Huda Akil12, Sarah E. Bergen13, Margit Burmeister, Sinéad B. Chapman4, Melissa DelaBastide14, Anders Juréus13, Hyun Min Kang8, Pui-Yan Kwok1, Jun Li8, Shawn Levy, Eric T. Monson15, Jennifer L. Moran16, Janet L. Sobell17, Stanley J. Watson12, Virginia L. Willour15, Sebastian Zöllner8, Rolf Adolfsson18, Douglas Blackwood19, Michael Boehnke8, Gerome Breen6, Aiden Corvin20, Nicholas John Craddock21, Arianna DiFlorio21, Christina M. Hultman13, Mikael Landén22, Mikael Landén13, Cathryn M. Lewis6, Steven A. McCarroll16, W. Richard McCombie14, Peter McGuffin6, Andrew M. McIntosh19, Andrew McQuillin23, Derek W. Morris20, Derek W. Morris24, Richard M. Myers, Michael Conlon O'Donovan21, Roel A. Ophoff25, Marco P. Boks, René S. Kahn5, Willem H. Ouwehand26, Michael John Owen21, Carlos N. Pato17, Carlos N. Pato27, Michele T. Pato27, Michele T. Pato17, Danielle Posthuma28, James B. Potash2, Andreas Reif29, Pamela Sklar5, Jordan W. Smoller4, Jordan W. Smoller16, Patrick F. Sullivan30, John B. Vincent31, John B. Vincent32, James T.R. Walters21, Benjamin M. Neale4, Benjamin M. Neale16, Shaun Purcell33, Shaun Purcell4, Neil Risch1, Catherine Schaefer9, Eli A. Stahl5, Peter P. Zandi2, Laura J. Scott8 
TL;DR: In this article, the authors examined the protein-coding (exonic) sequences of 3,987 unrelated individuals with bipolar disorder and 5,322 controls of predominantly European ancestry across four cohorts from the Bipolar Sequencing Consortium (BSC).
Abstract: Bipolar disorder (BD) is a serious mental illness with substantial common variant heritability. However, the role of rare coding variation in BD is not well established. We examined the protein-coding (exonic) sequences of 3,987 unrelated individuals with BD and 5,322 controls of predominantly European ancestry across four cohorts from the Bipolar Sequencing Consortium (BSC). We assessed the burden of rare, protein-altering, single nucleotide variants classified as pathogenic or likely pathogenic (P-LP) both exome-wide and within several groups of genes with phenotypic or biologic plausibility in BD. While we observed an increased burden of rare coding P-LP variants within 165 genes identified as BD GWAS regions in 3,987 BD cases (meta-analysis OR = 1.9, 95% CI = 1.3-2.8, one-sided p = 6.0 × 10-4), this enrichment did not replicate in an additional 9,929 BD cases and 14,018 controls (OR = 0.9, one-side p = 0.70). Although BD shares common variant heritability with schizophrenia, in the BSC sample we did not observe a significant enrichment of P-LP variants in SCZ GWAS genes, in two classes of neuronal synaptic genes (RBFOX2 and FMRP) associated with SCZ or in loss-of-function intolerant genes. In this study, the largest analysis of exonic variation in BD, individuals with BD do not carry a replicable enrichment of rare P-LP variants across the exome or in any of several groups of genes with biologic plausibility. Moreover, despite a strong shared susceptibility between BD and SCZ through common genetic variation, we do not observe an association between BD risk and rare P-LP coding variants in genes known to modulate risk for SCZ.

Journal ArticleDOI
TL;DR: It is reported that PRF1:p.A91V, is associated with increase of lymphocyte levels, especially within the cytotoxic memory T-cells, at general population level with reduced interleukin 7 receptor expression on these cells.
Abstract: Background:Defective alleles within the PRF1 gene, encoding the pore-forming protein perforin, in combination with environmental factors, cause familial type 2 hemophagocytic lymphohistiocytosis (F...

Journal ArticleDOI
TL;DR: FIVEx, an interactive eQTL/sQTL browser with an intuitive interface tailored to the functional interpretation of associated variants, is developed, which provides important insights for understanding potential tissue-specific regulatory mechanisms underlying trait-associated signals.
Abstract: SUMMARY Expression quantitative trait loci (eQTLs) characterize the associations between genetic variation and gene expression to provide insights into tissue-specific gene regulation. Interactive visualization of tissue-specific eQTLs or splice QTLs (sQTLs) can facilitate our understanding of functional variants relevant to disease-related traits. However, combining the multi-dimensional nature of eQTLs/sQTLs into a concise and informative visualization is challenging. Existing QTL visualization tools provide useful ways to summarize the unprecedented scale of transcriptomic data but are not necessarily tailored to answer questions about the functional interpretations of trait-associated variants or other variants of interest. We developed FIVEx, an interactive eQTL/sQTL browser with an intuitive interface tailored to the functional interpretation of associated variants. It features the ability to navigate seamlessly between different data views while providing relevant tissue- and locus-specific information to offer users a better understanding of population-scale multi-tissue transcriptomic profiles. Our implementation of the FIVEx browser on the EBI eQTL catalogue, encompassing 16 publicly available RNA-seq studies, provides important insights for understanding potential tissue-specific regulatory mechanisms underlying trait-associated signals. AVAILABILITY AND IMPLEMENTATION A FIVEx instance visualizing EBI eQTL catalogue data can be found at https://fivex.sph.umich.edu. Its source code is open source under an MIT license at https://github.com/statgen/fivex. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.

Journal ArticleDOI
Anna V. Mikhaylova1, Caitlin P. McHugh1, Linda M. Polfus2, Laura M. Raffield3, Meher Preethi Boorgula4, Thomas W. Blackwell5, Jennifer A. Brody1, Jai G. Broome1, Nathalie Chami6, Ming-Huei Chen7, Matthew P. Conomos1, Corey Cox4, Joanne E. Curran8, Michelle Daya4, Lynette Ekunwe9, David C. Glahn10, Nancy L. Heard-Costa7, Heather M. Highland3, Brian D. Hobbs11, Yann Ilboudo12, Deepti Jain1, Leslie A. Lange4, Tyne W Miller-Fleming13, Nancy Min9, Jee-Young Moon14, Michael Preuss6, Jonathon Rosen3, Kathleen A. Ryan15, Albert V. Smith5, Quan Sun3, Praveen Surendran16, Paul S. de Vries17, Klaudia Walter18, Zhe Wang6, Marsha M. Wheeler1, Lisa R. Yanek19, Xue Zhong13, Gonçalo R. Abecasis5, Laura Almasy20, Kathleen C. Barnes4, Terri H. Beaty21, Lewis C. Becker19, John Blangero8, Eric Boerwinkle17, Adam S. Butterworth22, Sameer Chavan4, Michael H. Cho11, Hélène Choquet23, Adolfo Correa9, Nancy J. Cox13, Dawn L. DeMeo11, Nauder Faraday19, Myriam Fornage17, Robert E. Gerszten24, Lifang Hou25, Andrew D. Johnson7, Eric Jorgenson, Robert C. Kaplan14, Charles Kooperberg26, Kousik Kundu18, Cecelia A. Laurie1, Guillaume Lettre12, Joshua P. Lewis15, Bingshan Li27, Yun Li3, Donald M. Lloyd-Jones25, Ruth J. F. Loos6, Ani Manichaikul28, Deborah A. Meyers29, Braxton D. Mitchell15, Alanna C. Morrison17, Debby Ngo30, Deborah A. Nickerson1, Suraj S. Nongmaithem18, Kari E. North3, Jeffrey R. O'Connell15, Victor E. Ortega31, Nathan Pankratz32, James A. Perry15, Bruce M. Psaty1, Stephen S. Rich28, Nicole Soranzo22, Jerome I. Rotter33, Edwin K. Silverman11, Nicholas L. Smith1, Hua Tang34, Russell P. Tracy35, Timothy A. Thornton1, Ramachandran S. Vasan7, Joe Zein36, Rasika A. Mathias19, Alexander P. Reiner15, Paul L. Auer37 
TL;DR: This article performed a GWAS of total leukocyte, neutrophil, lymphocyte, monocyte, eosinophil, and basophil counts generated from 109,563,748 variants in the autosomes and the X chromosome in the Trans-Omics for Precision Medicine (TOPMed) program, which included data from 61,802 individuals of diverse ancestry.
Abstract: Many common and rare variants associated with hematologic traits have been discovered through imputation on large-scale reference panels. However, the majority of genome-wide association studies (GWASs) have been conducted in Europeans, and determining causal variants has proved challenging. We performed a GWAS of total leukocyte, neutrophil, lymphocyte, monocyte, eosinophil, and basophil counts generated from 109,563,748 variants in the autosomes and the X chromosome in the Trans-Omics for Precision Medicine (TOPMed) program, which included data from 61,802 individuals of diverse ancestry. We discovered and replicated 7 leukocyte trait associations, including (1) the association between a chromosome X, pseudo-autosomal region (PAR), noncoding variant located between cytokine receptor genes (CSF2RA and CLRF2) and lower eosinophil count; and (2) associations between single variants found predominantly among African Americans at the S1PR3 (9q22.1) and HBB (11p15.4) loci and monocyte and lymphocyte counts, respectively. We further provide evidence indicating that the newly discovered eosinophil-lowering chromosome X PAR variant might be associated with reduced susceptibility to common allergic diseases such as atopic dermatitis and asthma. Additionally, we found a burden of very rare FLT3 (13q12.2) variants associated with monocyte counts. Together, these results emphasize the utility of whole-genome sequencing in diverse samples in identifying associations missed by European-ancestry-driven GWASs.

Journal ArticleDOI
TL;DR: The largest genome-wide association analysis to date of serum ALT and AST levels in over 388k people of European ancestry from UK biobank and DiscovEHR was reported in this article.
Abstract: Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) are biomarkers for liver health. Here we report the largest genome-wide association analysis to date of serum ALT and AST levels in over 388k people of European ancestry from UK biobank and DiscovEHR. Eleven million imputed markers with a minor allele frequency (MAF) ≥ 0.5% were analyzed. Overall, 300 ALT and 336 AST independent genome-wide significant associations were identified. Among them, 81 ALT and 61 AST associations are reported for the first time. Genome-wide interaction study identified 9 ALT and 12 AST independent associations significantly modified by body mass index (BMI), including several previously reported potential liver disease therapeutic targets, for example, PNPLA3, HSD17B13, and MARC1. While further work is necessary to understand the effect of ALT and AST-associated variants on liver disease, the weighted burden of significant BMI-modified signals is significantly associated with liver disease outcomes. In summary, this study identifies genetic associations which offer an important step forward in understanding the genetic architecture of serum ALT and AST levels. Significant interactions between BMI and genetic loci not only highlight the important role of adiposity in liver damage but also shed light on the genetic etiology of liver disease in obese individuals.

Posted ContentDOI
Tetsushi Nakao, Alexander G. Bick1, Alexander G. Bick2, Margaret A. Taub3, Seyedeh M. Zekavat4, Mesbah Uddin5, Mesbah Uddin1, Abhishek Niroula5, Abhishek Niroula1, Cara L. Carty6, John Lane7, Michael C. Honigberg1, Michael C. Honigberg5, Joshua S. Weinstock8, Akhil Pampana5, Akhil Pampana1, Christopher J. Gibson5, Gabriel K. Griffin9, Gabriel K. Griffin1, Gabriel K. Griffin5, Shoa L. Clarke10, Romit Bhattacharya5, Themistocles L. Assimes10, Themistocles L. Assimes11, Leslie S. Emery12, Adrienne M. Stilp12, Quenna Wong12, Jai G. Broome12, Cecelia A. Laurie12, Alyna T. Khan12, Albert V. Smith8, Thomas W. Blackwell8, Zachary T. Yoneda, Juan M. Peralta13, Donald W. Bowden14, Marguerite R. Irvin, Meher Preethi Boorgula15, Wei Zhao8, Lisa R. Yanek16, Kerri L. Wiggins12, James E. Hixson17, C. Charles Gu18, Gina M. Peloso19, Dan M. Roden20, Muagututi’a S. Reupena, Chii-Min Hwu21, Chii-Min Hwu22, Dawn L. DeMeo9, Kari E. North23, Shannon Kelly24, Solomon K. Musani25, Joshua C. Bis12, Donald M. Lloyd-Jones26, Jill M. Johnsen, Michael Preuss27, Russell P. Tracy28, Patricia A. Peyser8, Dandi Qiao9, Pinkal Desai29, Joanne E. Curran13, Barry I. Freedman14, Hemant K. Tiwari30, Sameer Chavan15, Jennifer A. Smith8, Nicholas L. Smith31, Nicholas L. Smith12, Tanika N. Kelly32, Bertha A. Hildalgo30, L. Adrienne Cupples19, Daniel E. Weeks33, Nicola L. Hawley4, Ryan L. Minster33, Ranjan Deka, Take Naseri18, Lisa de las Fuentes23, Laura M. Raffield17, Alanna C. Morrison17, Paul S. Vries34, Christie M. Ballantyne27, Eimear E. Kenny35, Stephen S. Rich23, Eric A. Whitsel9, Michael H. Cho, M. Benjamin Shoemaker36, Betty S. Pace13, John Blangero14, Nicholette D. Palmer37, Nicholette D. Palmer38, Braxton D. Mitchell38, A. R. Shuldiner15, Kathleen C. Barnes5, S Redline8, Sharon L.R. Kardia8, Sharon L.R. Kardia39, Gonçalo R. Abecasis16, Lewis C. Becker12, Lewis C. Becker31, Susan R. Heckbert32, Jiang He3, Wendy Post40, Donna K. Arnett19, Ramachandran S. Vasan41, Dawood Darbar9, Dawood Darbar5, Scott T. Weiss42, Stephen T. McGarvey43, Mariza de Andrade44, Yii-Der Ida Chen45, Yii-Der Ida Chen46, Robert C. Kaplan47, Deborah A. Meyers, Brian Custer25, Adolfo Correa12, Bruce M. Psaty48, Bruce M. Psaty17, Myriam Fornage5, JoAnn E. Manson8, Eric Boerwinkle12, Barbara A. Konkle27, Ruth J.F. Loos44, Jerome I. Rotter9, Edwin K. Silverman, Charles Kooperberg10, Siddhartha Jaiswal9, Siddhartha Jaiswal5, Peter Libby1, Patrick T. Ellinor7, Nathan Pankratz5, Nathan Pankratz1, Nathan Pankratz49, Benjamin L. Ebert, Alexander P. Reiner16, R. Mathias27, Ron Do, Pradeep Natarajan 
Broad Institute1, Vanderbilt University2, Johns Hopkins University3, Yale University4, Harvard University5, Washington State University6, University of Minnesota7, University of Michigan8, Brigham and Women's Hospital9, Stanford University10, VA Palo Alto Healthcare System11, University of Washington12, University of Texas at Austin13, Wake Forest University14, University of Colorado Boulder15, Johns Hopkins University School of Medicine16, University of Texas Health Science Center at Houston17, Washington University in St. Louis18, Boston University19, Vanderbilt University Medical Center20, National Yang-Ming University21, Taipei Veterans General Hospital22, University of North Carolina at Chapel Hill23, Children's Hospital Oakland24, University of Mississippi Medical Center25, Northwestern University26, Icahn School of Medicine at Mount Sinai27, University of Vermont28, Cornell University29, University of Alabama30, Kaiser Permanente31, Tulane University32, University of Pittsburgh33, Baylor College of Medicine34, University of Virginia35, Georgia Regents University36, United States Department of Veterans Affairs37, University of Maryland, Baltimore38, Regeneron39, University of Kentucky40, University of Illinois at Urbana–Champaign41, Brown University42, Mayo Clinic43, Los Angeles Biomedical Research Institute44, Albert Einstein College of Medicine45, Fred Hutchinson Cancer Research Center46, University of Arizona47, University of Texas Health Science Center at San Antonio48, Howard Hughes Medical Institute49
01 Mar 2021-medRxiv
TL;DR: In this article, the authors investigated the relationship between CHIP, LTL, and CAD in Trans-Omics for Precision Medicine (TOPMed) program (N=63,302) and UK Biobank (n=48,658).
Abstract: Human genetic studies support an inverse causal relationship between leukocyte telomere length (LTL) and coronary artery disease (CAD), but directionally mixed effects for LTL and diverse malignancies. Clonal hematopoiesis of indeterminate potential (CHIP), characterized by expansion of hematopoietic cells bearing leukemogenic mutations, predisposes both hematologic malignancy and CAD. TERT (which encodes telomerase reverse transcriptase) is the most significantly associated germline locus for CHIP in genome-wide association studies. Here, we investigated the relationship between CHIP, LTL, and CAD in Trans-Omics for Precision Medicine (TOPMed) program (N=63,302) and UK Biobank (N=48,658). Bidirectional Mendelian randomization studies were consistent with LTL lengthening increasing propensity to develop CHIP, but CHIP then in turn hastening LTL shortening. We also demonstrated evidence of modest mediation between CHIP and CAD by LTL. Our data promote an understanding of potential causal relationships across CHIP and LTL toward prevention of CAD.

Journal ArticleDOI
TL;DR: This article used whole-genome sequencing from NHLBI's Trans-Omics for Precision Medicine Initiative (TOPMed) in a large multi-ethnic sample to further explore common and rare variation contributing to PLT and MPV.
Abstract: Platelets play a key role in thrombosis and hemostasis. Platelet count (PLT) and mean platelet volume (MPV) are highly heritable quantitative traits, with hundreds of genetic signals previously identified, mostly in European ancestry populations. We here utilize whole genome sequencing from NHLBI's Trans-Omics for Precision Medicine Initiative (TOPMed) in a large multi-ethnic sample to further explore common and rare variation contributing to PLT (n = 61 200) and MPV (n = 23 485). We identified and replicated secondary signals at MPL (rs532784633) and PECAM1 (rs73345162), both more common in African ancestry populations. We also observed rare variation in Mendelian platelet related disorder genes influencing variation in platelet traits in TOPMed cohorts (not enriched for blood disorders). For example, association of GP9 with lower PLT and higher MPV was partly driven by a pathogenic Bernard-Soulier syndrome variant (rs5030764, p.Asn61Ser), and the signals at TUBB1 and CD36 were partly driven by loss of function variants not annotated as pathogenic in ClinVar (rs199948010 and rs571975065). However, residual signal remained for these gene-based signals after adjusting for lead variants, suggesting that additional variants in Mendelian genes with impacts in general population cohorts remain to be identified. Gene-based signals were also identified at several GWAS identified loci for genes not annotated for Mendelian platelet disorders (PTPRH, TET2, CHEK2), with somatic variation driving the result at TET2. These results highlight the value of whole genome sequencing in populations of diverse genetic ancestry to identify novel regulatory and coding signals, even for well-studied traits like platelet traits.


Posted ContentDOI
24 Jan 2021-bioRxiv
TL;DR: FIVEx as discussed by the authors is an interactive eQTL browser with an intuitive interface tailored to the functional interpretation of associated variants, which can provide users with a better understanding of population-scale multi-tissue transcriptomic profiles.
Abstract: Expression quantitative trait loci (eQTLs) characterize the associations between genetic variation and gene expression to provide insights into tissue-specific gene regulation.Interactive visualization of tissue-specific eQTLs can facilitateourunderstanding of functional variants relevant to disease-related traits. However, combiningthe multi-dimensional nature of eQTLs into a concise and informative visualization ischallenging. Existing eQTL visualization tools provide useful ways to summarize the unprecedented scale of transcriptomic data but are not necessarily tailored to answer questions aboutthefunctional interpretations of trait-associated variants or other variants of interest. We developed FIVEx, an interactive eQTL browser with an intuitive interface tailored to the functional interpretation of associated variants. It features the ability to navigate seamlessly between different data views while providing relevant tissue-and locus-specific information to offer users a better understanding of population-scale multi-tissue transcriptomic profiles. Our implementation of the FIVEx browser on the Gene-Tissue Expression (GTEx) dataset providesimportant insights for understandingpotential tissue-specific regulatory mechanisms underlying trait-associated signals.