Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Amanda Dobbyn is active.

Publication


Featured researches published by Amanda Dobbyn.


PLOS Genetics | 2015

Disproportionate Contributions of Select Genomic Compartments and Cell Types to Genetic Risk for Coronary Artery Disease

Hong-Hee Won; Pradeep Natarajan; Amanda Dobbyn; Daniel M. Jordan; Panos Roussos; Kasper Lage; Soumya Raychaudhuri; Eli A. Stahl; Ron Do

Large genome-wide association studies (GWAS) have identified many genetic loci associated with risk for myocardial infarction (MI) and coronary artery disease (CAD). Concurrently, efforts such as the National Institutes of Health (NIH) Roadmap Epigenomics Project and the Encyclopedia of DNA Elements (ENCODE) Consortium have provided unprecedented data on functional elements of the human genome. In the present study, we systematically investigate the biological link between genetic variants associated with this complex disease and their impacts on gene function. First, we examined the heritability of MI/CAD according to genomic compartments. We observed that single nucleotide polymorphisms (SNPs) residing within nearby regulatory regions show significant polygenicity and contribute between 59–71% of the heritability for MI/CAD. Second, we showed that the polygenicity and heritability explained by these SNPs are enriched in histone modification marks in specific cell types. Third, we found that a statistically higher number of 45 MI/CAD-associated SNPs that have been identified from large-scale GWAS studies reside within certain functional elements of the genome, particularly in active enhancer and promoter regions. Finally, we observed significant heterogeneity of this signal across cell types, with strong signals observed within adipose nuclei, as well as brain and spleen cell types. These results suggest that the genetic etiology of MI/CAD is largely explained by tissue-specific regulatory perturbation within the human genome.


Genome Medicine | 2017

Integrated Bayesian analysis of rare exonic variants to identify risk genes for schizophrenia and neurodevelopmental disorders

Hoang T. Nguyen; April Kim; Amanda Dobbyn; Laura M. Huckins; Ana B. Muñoz-Manchado; Douglas Ruderfer; Giulio Genovese; Menachem Fromer; Xinyi Xu; Dalila Pinto; Sten Linnarsson; Matthijs Verhage; August B. Smit; Jens Hjerling-Leffler; Joseph D. Buxbaum; Christina M. Hultman; Pamela Sklar; S Purcell; Kasper Lage; Xin He; Patrick F. Sullivan; Eli A. Stahl

BackgroundIntegrating rare variation from trio family and case–control studies has successfully implicated specific genes contributing to risk of neurodevelopmental disorders (NDDs) including autism spectrum disorders (ASD), intellectual disability (ID), developmental disorders (DDs), and epilepsy (EPI). For schizophrenia (SCZ), however, while sets of genes have been implicated through the study of rare variation, only two risk genes have been identified.MethodsWe used hierarchical Bayesian modeling of rare-variant genetic architecture to estimate mean effect sizes and risk-gene proportions, analyzing the largest available collection of whole exome sequence data for SCZ (1,077 trios, 6,699 cases, and 13,028 controls), and data for four NDDs (ASD, ID, DD, and EPI; total 10,792 trios, and 4,058 cases and controls).ResultsFor SCZ, we estimate there are 1,551 risk genes. There are more risk genes and they have weaker effects than for NDDs. We provide power analyses to predict the number of risk-gene discoveries as more data become available. We confirm and augment prior risk gene and gene set enrichment results for SCZ and NDDs. In particular, we detected 98 new DD risk genes at FDR < 0.05. Correlations of risk-gene posterior probabilities are high across four NDDs (ρ>0.55), but low between SCZ and the NDDs (ρ<0.3). An in-depth analysis of 288 NDD genes shows there is highly significant protein–protein interaction (PPI) network connectivity, and functionally distinct PPI subnetworks based on pathway enrichment, single-cell RNA-seq cell types, and multi-region developmental brain RNA-seq.ConclusionsWe have extended a pipeline used in ASD studies and applied it to infer rare genetic parameters for SCZ and four NDDs (https://github.com/hoangtn/extTADA). We find many new DD risk genes, supported by gene set enrichment and PPI network connectivity analyses. We find greater similarity among NDDs than between NDDs and SCZ. NDD gene subnetworks are implicated in postnatally expressed presynaptic and postsynaptic genes, and for transcriptional and post-transcriptional gene regulation in prenatal neural progenitor and stem cells.


Arthritis & Rheumatism | 2017

Population‐Specific Resequencing Associates the ATP‐Binding Cassette Subfamily C Member 4 Gene With Gout in New Zealand Māori and Pacific Men

Callum Tanner; James Boocock; Eli A. Stahl; Amanda Dobbyn; Asim K. Mandal; Murray Cadzow; Amanda Phipps-Green; Ruth Topless; Jennie Harré Hindmarsh; Lisa K. Stamp; Nicola Dalbeth; Hyon K. Choi; David B. Mount; Tony R. Merriman

There is no evidence for a genetic association between organic anion transporters 1–3 (SLC22A6, SLC22A7, and SLC22A8) and multidrug resistance protein 4 (MRP4; encoded by ABCC4) with the levels of serum urate or gout. The Māori and Pacific (Polynesian) population of New Zealand has the highest prevalence of gout worldwide. The aim of this study was to determine whether any Polynesian population–specific genetic variants in SLC22A6–8 and ABCC4 are associated with gout.


bioRxiv | 2017

Co-localization of Conditional eQTL and GWAS Signatures in Schizophrenia

Amanda Dobbyn; Laura M. Huckins; James Boocock; Laura G. Sloofman; Benjamin S. Glicksberg; Claudia Giambartolomei; Gabriel E. Hoffman; Thanneer M. Perumal; Kiran Girdhar; Yan Jiang; Douglas Ruderfer; Robin Kramer; Dalila Pinto; Schahram Akbarian; Panos Roussos; Enrico Domenici; Bernie Devlin; Pamela Sklar; Eli A. Stahl; Solveig K. Sieberts

Causal genes and variants within genome-wide association study (GWAS) loci can be identified by integrating GWAS statistics with expression quantitative trait loci (eQTL) and determining which SNPs underlie both GWAS and eQTL signals. Most analyses, however, consider only the marginal eQTL signal, rather than dissecting this signal into multiple independent eQTL for each gene. Here we show that analyzing conditional eQTL signatures, which could be important under specific cellular or temporal contexts, leads to improved fine mapping of GWAS associations. Using genotypes and gene expression levels from post-mortem human brain samples (N=467) reported by the CommonMind Consortium (CMC), we find that conditional eQTL are widespread; 63% of genes with primary eQTL also have conditional eQTL. In addition, genomic features associated with conditional eQTL are consistent with context specific (i.e. tissue, cell type, or developmental time point specific) regulation of gene expression. Integrating the Psychiatric Genomics Consortium schizophrenia (SCZ) GWAS and CMC conditional eQTL data reveals forty loci with strong evidence for co-localization (posterior probability >0.8), including six loci with co-localization of conditional eQTL. Our co-localization analyses support previously reported genes and identify novel genes for schizophrenia risk, and provide specific hypotheses for their functional follow-up.


bioRxiv | 2018

Gene expression imputation across multiple brain regions reveals schizophrenia risk throughout development

Laura M. Huckins; Amanda Dobbyn; Douglas Ruderfer; Gabriel E. Hoffman; Weiqing Wang; Antonio F. Pardiñas; Veera M. Rajagopal; Thomas Damm Als; Hoang Tan Hoang; Kiran Girdhar; James Boocock; Panagiotis Roussos; Menachem Fromer; Robin Kramer; Enrico Domenici; Eric R. Gamazon; Shaun Purcell; Ditte Demontis; Anders D. Børglum; James Tynan Rhys Walters; Michael Conlon O'Donovan; Patrick F. Sullivan; Michael John Owen; Bernie Devlin; Solveig K. Sieberts; Nancy J. Cox; Hae Kyung Im; Pamela Sklar; Eli A. Stahl

Transcriptomic imputation approaches offer an opportunity to test associations between disease and gene expression in otherwise inaccessible tissues, such as brain, by combining eQTL reference panels with large-scale genotype data. These genic associations could elucidate signals in complex GWAS loci and may disentangle the role of different tissues in disease development. Here, we use the largest eQTL reference panel for the dorso-lateral pre-frontal cortex (DLPFC), collected by the CommonMind Consortium, to create a set of gene expression predictors and demonstrate their utility. We applied these predictors to 40,299 schizophrenia cases and 65,264 matched controls, constituting the largest transcriptomic imputation study of schizophrenia to date. We also computed predicted gene expression levels for 12 additional brain regions, using publicly available predictor models from GTEx. We identified 413 genic associations across 13 brain regions. Stepwise conditioning across the genes and tissues identified 71 associated genes (67 outside the MHC), with the majority of associations found in the DLPFC, and of which 14/67 genes did not fall within previously genome-wide significant loci. We identified 36 significantly enriched pathways, including hexosaminidase-A deficiency, and multiple pathways associated with porphyric disorders. We investigated developmental expression patterns for all 67 non-MHC associated genes using BRAINSPAN, and identified groups of genes expressed specifically pre-natally or post-natally.


bioRxiv | 2018

Prioritizing risk genes for neurodevelopmental disorders using pathway information

Hoang T. Nguyen; Amanda Dobbyn; Alex Charney; Nathan Skene; Laura M. Huckins; Weiqing Wang; Douglas Ruderfer; Xinyi Xu; Menachem Fromer; S Purcell; Matthijs Verhage; August B. Smit; Jens Hjerling-Leffler; Joseph D. Buxbaum; Dalila Pinto; Xin He; Patrick F. Sullivan; Eli A. Stahl

Trio family and case-control studies of next-generation sequencing data have proven integral to understanding the contribution of rare inherited and de novo single-nucleotide variants to the genetic architecture of complex disease. Ideally, such studies should identify individual risk genes of moderate to large effect size to generate novel treatment hypotheses for further follow-up. However, due to insufficient power, gene set enrichment analyses have come to be relied upon for detecting differences between cases and controls, implicating sets of hundreds of genes rather than specific targets for further investigation. Here, we present a Bayesian statistical framework, termed gTADA, that integrates gene-set membership information with gene-level de novo and rare inherited case-control counts, to prioritize risk genes with excess rare variant burden within enriched gene sets. Applying gTADA to available whole-exome sequencing datasets for several neuropsychiatric conditions, we replicated previously reported gene set enrichments and identified novel risk genes. For epilepsy, gTADA prioritized 40 risk genes (posterior probabilities > 0.95), 6 of which replicate in an independent whole-genome sequencing study. In addition, 30/40 genes are novel genes. We found that epilepsy genes had high protein-protein interaction (PPI) network connectivity, and show specific expression during human brain development. Some of the top prioritized EPI genes were connected to a PPI subnetwork of immune genes and show specific expression in prenatal microglia. We also identified multiple enriched drug-target gene sets for EPI which included immunostimulants as well as known antiepileptics. Immune biology was supported specifically by case-control variants from familial epilepsies rather than do novo mutations in generalized encephalitic epilepsy.


bioRxiv | 2018

mTADA: a framework for analyzing de novo mutations in multiple traits

Hoang T. Nguyen; Amanda Dobbyn; Joseph D. Buxbaum; Dalila Pinto; S Purcell; Patrick F. Sullivan; Xin He; Eli A. Stahl

Joint analysis of multiple traits can result in the identification of associations not found through the analysis of each trait in isolation. In addition, approaches that consider multiple traits can aid in the characterization of shared genetic etiology among those traits. In recent years, parent-offspring trio studies have reported an enrichment of de novo mutations (DNMs) in neuropsychiatric disorders. The analysis of DNM data in the context of neuropsychiatric disorders has implicated multiple putatively causal genes, and a number of reported genes are shared across disorders. However, a joint analysis method designed to integrate de novo mutation data from multiple studies has yet to be implemented. We here introduce multi pi e-trait TAD A (mTADA) which jointly analyzes two traits using DNMs from non-overlapping family samples. mTADA uses two single-trait analysis data sets to estimate the proportion of overlapping risk genes, and reports genes shared between and specific to the relevant disorders. We applied mTADA to >13,000 trios for six disorders: schizophrenia (SCZ), autism spectrum disorder (ASD), developmental disorders (DD), intellectual disability (ID), epilepsy (EPI), and congenital heart disease (CHD). We report the proportion of overlapping risk genes and the specific risk genes shared for each pair of disorders. A total of 153 genes were found to be shared in at least one pair of disorders. The largest percentages of shared risk genes were observed for pairs of DD, ID, ASD, and CHD (>20%) whereas SCZ, CHD, and EPI did not show strong overlaps In risk gene set between them. Furthermore, mTADA identified additional SCZ, EPI and CHD risk genes through integration with DD de novo mutation data. For CHD, using DD information, 31 risk genes with posterior probabilities > 0.8 were identified, and 20 of these 31 genes were not in the list of known CHD genes. We find evidence that most significant CHD risk genes are strongly expressed in prenatal stages of the human genes. Finally, we validated our findings for CHD and EPI in independent cohorts comprising 1241 CHD trios, 226 CHD singletons and 197 EPI trios. Multiple novel risk genes identified by mTADA also had de novo mutations in these independent data sets. The joint analysis method introduced here, mTADA, is able to identify risk genes shared by two traits as well as additional risk genes not found through single-trait analysis only. A number of risk genes reported by mTADA are identified only through joint analysis, specifically when ASD, DD, or ID are one of the two traits examined. This suggests that novel genes for the trait or a new trait might converge to a core gene list of the three traits.


bioRxiv | 2018

Identifying tissues implicated in Anorexia Nervosa using Transcriptomic Imputation

Laura M. Huckins; Amanda Dobbyn; Whitney McFadden; Douglas Ruderfer; Weiqing Wang; Eric R. Gamazon; Virpi M. Leppä; Bernie Devlin; Solveig K. Sieberts; Nancy J. Cox; Hae Kyung Im; Gerome Breen; Pamela Sklar; Cynthia M. Bulik; Eli A. Stahl

Anorexia nervosa (AN) is a complex and serious eating disorder, occurring in ~1% of individuals. Despite having the highest mortality rate of any psychiatric disorder, little is known about the aetiology of AN, and few effective treatments exist. Global efforts to collect large sample sizes of individuals with AN have been highly successful, and a recent study consequently identified the first genome-wide significant locus involved in AN. This result, coupled with other recent studies and epidemiological evidence, suggest that previous characterizations of AN as a purely psychiatric disorder are over-simplified. Rather, both neurological and metabolic pathways may also be involved. In order to elucidate more of the system-specific aetiology of AN, we applied transcriptomic imputation methods to 3,495 cases and 10,982 controls, collected by the Eating Disorders Working Group of the Psychiatric Genomics Consortium (PGC-ED). Transcriptomic Imputation (TI) methods approaches use machine-learning methods to impute tissue-specific gene expression from large genotype data using curated eQTL reference panels. These offer an exciting opportunity to compare gene associations across neurological and metabolic tissues. Here, we applied CommonMind Consortium (CMC) and GTEx-derived gene expression prediction models for 13 brain tissues and 12 tissues with potential metabolic involvement (adipose, adrenal gland, 2 colon, 3 esophagus, liver, pancreas, small intestine, spleen, stomach). We identified 35 significant gene-tissue associations within the large chromosome 12 region described in the recent PGC-ED GWAS. We applied forward stepwise conditional analyses and FINEMAP to associations within this locus to identify putatively causal signals. We identified four independently associated genes; RPS26, C12orf49, SUOX, and RDH16. We also identified two further genome-wide significant gene-tissue associations, both in brain tissues; REEP5, in the dorso-lateral pre-frontal cortex (DLPFC; p=8.52×10−07), and CUL3, in the caudate basal ganglia (p=1.8×10−06). These genes are significantly enriched for associations with anthropometric phenotypes in the UK BioBank, as well as multiple psychiatric, addiction, and appetite/satiety pathways. Our results support a model of AN risk influenced by both metabolic and psychiatric factors.


American Journal of Human Genetics | 2018

Landscape of Conditional eQTL in Dorsolateral Prefrontal Cortex and Co-localization with Schizophrenia GWAS

Amanda Dobbyn; Laura M. Huckins; James Boocock; Laura G. Sloofman; Benjamin S. Glicksberg; Claudia Giambartolomei; Gabriel E. Hoffman; Thanneer M. Perumal; Kiran Girdhar; Yan Jiang; Towfique Raj; Douglas Ruderfer; Robin Kramer; Dalila Pinto; Pamela Sklar; Joseph D. Buxbaum; Bernie Devlin; David A. Lewis; Raquel E. Gur; Chang-Gyu Hahn; Keisuke Hirai; Hiroyoshi Toyoshiba; Enrico Domenici; Laurent Essioux; Lara M. Mangravite; Mette A. Peters; Thomas Lehner; Barbara K. Lipska; A. Ercument Cicek; Cong Lu

Causal genes and variants within genome-wide association study (GWAS) loci can be identified by integrating GWAS statistics with expression quantitative trait loci (eQTL) and determining which variants underlie both GWAS and eQTL signals. Most analyses, however, consider only the marginal eQTL signal, rather than dissect this signal into multiple conditionally independent signals for each gene. Here we show that analyzing conditional eQTL signatures, which could be important under specific cellular or temporal contexts, leads to improved fine mapping of GWAS associations. Using genotypes and gene expression levels from post-mortem human brain samples (n = 467) reported by the CommonMind Consortium (CMC), we find that conditional eQTL are widespread; 63% of genes with primary eQTL also have conditional eQTL. In addition, genomic features associated with conditional eQTL are consistent with context-specific (e.g., tissue-, cell type-, or developmental time point-specific) regulation of gene expression. Integrating the 2014 Psychiatric Genomics Consortium schizophrenia (SCZ) GWAS and CMC primary and conditional eQTL data reveals 40 loci with strong evidence for co-localization (posterior probability > 0.8), including six loci with co-localization of conditional eQTL. Our co-localization analyses support previously reported genes, identify novel genes associated with schizophrenia risk, and provide specific hypotheses for their functional follow-up.


bioRxiv | 2017

Transcriptomic Imputation of Bipolar Disorder and Bipolar subtypes reveals 29 novel associated genes

Laura M. Huckins; Amanda Dobbyn; Whitney McFadden; Weiqing Wang; Douglas Ruderfer; Gabriel E. Hoffman; Veera M. Rajagopal; Hoang T. Nguyen; Panagiotis Roussos; Menachem Fromer; Robin Kramer; Enrico Domenici; Eric R. Gamazon; Ditte Demontis; Anders D. Børglum; Bernie Devlin; Solveig K. Sieberts; Nancy J. Cox; Hae Kyung Im; Pamela Sklar; Eli A. Stahl

Bipolar disorder is a complex neuropsychiatric disorder presenting with episodic mood disturbances. In this study we use a transcriptomic imputation approach to identify novel genes and pathways associated with bipolar disorder, as well as three diagnostically and genetically distinct subtypes. Transcriptomic imputation approaches leverage well-curated and publicly available eQTL reference panels to create gene-expression prediction models, which may then be applied to “impute” genetically regulated gene expression (GREX) in large GWAS datasets. By testing for association between phenotype and GREX, rather than genotype, we hope to identify more biologically interpretable associations, and thus elucidate more of the genetic architecture of bipolar disorder. We applied GREX prediction models for 13 brain regions (derived from CommonMind Consortium and GTEx eQTL reference panels) to 21,488 bipolar cases and 54,303 matched controls, constituting the largest transcriptomic imputation study of bipolar disorder (BPD) to date. Additionally, we analyzed three specific BPD subtypes, including 14,938 individuals with subtype 1 (BD-I), 3,543 individuals with subtype 2 (BD-II), and 1,500 individuals with schizoaffective subtype (SAB). We identified 125 gene-tissue associations with BPD, of which 53 represent independent associations after FINEMAP analysis. 29/53 associations were novel; i.e., did not lie within 1Mb of a locus identified in the recent PGC-BD GWAS. We identified 37 independent BD-I gene-tissue associations (10 novel), 2 BD-II associations, and 2 SAB associations. Our BPD, BD-I and BD-II associations were significantly more likely to be differentially expressed in post-mortem brain tissue of BPD, BD-I and BD-II cases than we might expect by chance. Together with our pathway analysis, our results support long-standing hypotheses about bipolar disorder risk, including a role for oxidative stress and mitochondrial dysfunction, the post-synaptic density, and an enrichment of circadian rhythm and clock genes within our results.

Collaboration


Dive into the Amanda Dobbyn's collaboration.

Top Co-Authors

Avatar

Eli A. Stahl

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Laura M. Huckins

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Douglas Ruderfer

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Pamela Sklar

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Bernie Devlin

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Dalila Pinto

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Menachem Fromer

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Hoang T. Nguyen

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Joseph D. Buxbaum

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Patrick F. Sullivan

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge