Network


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

Hotspot


Dive into the research topics where Ryan P. Welch is active.

Publication


Featured researches published by Ryan P. Welch.


Bioinformatics | 2010

LocusZoom: regional visualization of genome-wide association scan results

Randall Pruim; Ryan P. Welch; Serena Sanna; Tanya M. Teslovich; Peter S. Chines; Terry P. Gliedt; Michael Boehnke; Gonçalo R. Abecasis; Cristen J. Willer

Summary: Genome-wide association studies (GWAS) have revealed hundreds of loci associated with common human genetic diseases and traits. We have developed a web-based plotting tool that provides fast visual display of GWAS results in a publication-ready format. LocusZoom visually displays regional information such as the strength and extent of the association signal relative to genomic position, local linkage disequilibrium (LD) and recombination patterns and the positions of genes in the region. Availability: LocusZoom can be accessed from a web interface at http://csg.sph.umich.edu/locuszoom. Users may generate a single plot using a web form, or many plots using batch mode. The software utilizes LD information from HapMap Phase II (CEU, YRI and JPT+CHB) or 1000 Genomes (CEU) and gene information from the UCSC browser, and will accept SNP identifiers in dbSNP or 1000 Genomes format. Single plots are generated in ∼20 s. Source code and associated databases are available for download and local installation, and full documentation is available online. Contact: [email protected]


Cell Metabolism | 2010

Global Epigenomic Analysis of Primary Human Pancreatic Islets Provides Insights into Type 2 Diabetes Susceptibility Loci

Michael L. Stitzel; Praveen Sethupathy; Daniel Pearson; Peter S. Chines; Lingyun Song; Michael R. Erdos; Ryan P. Welch; Stephen C. J. Parker; Alan P. Boyle; Laura J. Scott; Elliott H. Margulies; Michael Boehnke; Terrence S. Furey; Gregory E. Crawford; Francis S. Collins

Identifying cis-regulatory elements is important to understanding how human pancreatic islets modulate gene expression in physiologic or pathophysiologic (e.g., diabetic) conditions. We conducted genome-wide analysis of DNase I hypersensitive sites, histone H3 lysine methylation modifications (K4me1, K4me3, K79me2), and CCCTC factor (CTCF) binding in human islets. This identified ∼18,000 putative promoters (several hundred unannotated and islet-active). Surprisingly, active promoter modifications were absent at genes encoding islet-specific hormones, suggesting a distinct regulatory mechanism. Of 34,039 distal (nonpromoter) regulatory elements, 47% are islet unique and 22% are CTCF bound. In the 18 type 2 diabetes (T2D)-associated loci, we identified 118 putative regulatory elements and confirmed enhancer activity for 12 of 33 tested. Among six regulatory elements harboring T2D-associated variants, two exhibit significant allele-specific differences in activity. These findings present a global snapshot of the human islet epigenome and should provide functional context for noncoding variants emerging from genetic studies of T2D and other islet disorders.


PLOS Genetics | 2014

Re-sequencing Expands Our Understanding of the Phenotypic Impact of Variants at GWAS Loci

Tanya M. Teslovich; Christian Fuchsberger; Vasily Ramensky; Pranav Yajnik; Daniel C. Koboldt; David E. Larson; Qunyuan Zhang; Ling Lin; Ryan P. Welch; Li Ding; Michael D. McLellan; Michele O'Laughlin; Catrina C. Fronick; Lucinda Fulton; Vincent Magrini; Amy J. Swift; Paul Elliott; Marjo-Riitta Järvelin; Marika Kaakinen; Mark I. McCarthy; Leena Peltonen; Anneli Pouta; Lori L. Bonnycastle; Francis S. Collins; Heather M. Stringham; Jaakko Tuomilehto; Samuli Ripatti; Robert S. Fulton; Chiara Sabatti; Richard K. Wilson

Genome-wide association studies (GWAS) have identified >500 common variants associated with quantitative metabolic traits, but in aggregate such variants explain at most 20–30% of the heritable component of population variation in these traits. To further investigate the impact of genotypic variation on metabolic traits, we conducted re-sequencing studies in >6,000 members of a Finnish population cohort (The Northern Finland Birth Cohort of 1966 [NFBC]) and a type 2 diabetes case-control sample (The Finland-United States Investigation of NIDDM Genetics [FUSION] study). By sequencing the coding sequence and 5′ and 3′ untranslated regions of 78 genes at 17 GWAS loci associated with one or more of six metabolic traits (serum levels of fasting HDL-C, LDL-C, total cholesterol, triglycerides, plasma glucose, and insulin), and conducting both single-variant and gene-level association tests, we obtained a more complete understanding of phenotype-genotype associations at eight of these loci. At all eight of these loci, the identification of new associations provides significant evidence for multiple genetic signals to one or more phenotypes, and at two loci, in the genes ABCA1 and CETP, we found significant gene-level evidence of association to non-synonymous variants with MAF<1%. Additionally, two potentially deleterious variants that demonstrated significant associations (rs138726309, a missense variant in G6PC2, and rs28933094, a missense variant in LIPC) were considerably more common in these Finnish samples than in European reference populations, supporting our prior hypothesis that deleterious variants could attain high frequencies in this isolated population, likely due to the effects of population bottlenecks. Our results highlight the value of large, well-phenotyped samples for rare-variant association analysis, and the challenge of evaluating the phenotypic impact of such variants.


Nature Communications | 2016

The genetic regulatory signature of type 2 diabetes in human skeletal muscle

Laura J. Scott; Michael R. Erdos; Jeroen R. Huyghe; Ryan P. Welch; Andrew T. Beck; Brooke N. Wolford; Peter S. Chines; John P. Didion; Heather M. Stringham; D. Leland Taylor; Anne U. Jackson; Swarooparani Vadlamudi; Lori L. Bonnycastle; Leena Kinnunen; Jouko Saramies; Jouko Sundvall; Ricardo D'Oliveira Albanus; Anna Kiseleva; John Hensley; Gregory E. Crawford; Hui Jiang; Xiaoquan Wen; Richard M. Watanabe; Timo A. Lakka; Karen L. Mohlke; Markku Laakso; Jaakko Tuomilehto; Heikki A. Koistinen; Michael Boehnke; Francis S. Collins

Type 2 diabetes (T2D) results from the combined effects of genetic and environmental factors on multiple tissues over time. Of the >100 variants associated with T2D and related traits in genome-wide association studies (GWAS), >90% occur in non-coding regions, suggesting a strong regulatory component to T2D risk. Here to understand how T2D status, metabolic traits and genetic variation influence gene expression, we analyse skeletal muscle biopsies from 271 well-phenotyped Finnish participants with glucose tolerance ranging from normal to newly diagnosed T2D. We perform high-depth strand-specific mRNA-sequencing and dense genotyping. Computational integration of these data with epigenome data, including ATAC-seq on skeletal muscle, and transcriptome data across diverse tissues reveals that the tissue-specific genetic regulatory architecture of skeletal muscle is highly enriched in muscle stretch/super enhancers, including some that overlap T2D GWAS variants. In one such example, T2D risk alleles residing in a muscle stretch/super enhancer are linked to increased expression and alternative splicing of muscle-specific isoforms of ANK1.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Genetic regulatory signatures underlying islet gene expression and type 2 diabetes.

Arushi Varshney; Laura J. Scott; Ryan P. Welch; Michael R. Erdos; Peter S. Chines; Ricardo D'Oliveira Albanus; Peter Orchard; Brooke N. Wolford; Romy Kursawe; Swarooparani Vadlamudi; Maren E. Cannon; John P. Didion; John Hensley; Anthony Kirilusha; Lori L. Bonnycastle; D. Leland Taylor; Richard M. Watanabe; Karen L. Mohlke; Michael Boehnke; Francis S. Collins; Stephen C. J. Parker; Michael L. Stitzel

Significance The majority of genetic variants associated with type 2 diabetes (T2D) are located outside of genes in noncoding regions that may regulate gene expression in disease-relevant tissues, like pancreatic islets. Here, we present the largest integrated analysis to date of high-resolution, high-throughput human islet molecular profiling data to characterize the genome (DNA), epigenome (DNA packaging), and transcriptome (gene expression). We find that T2D genetic variants are enriched in regions of the genome where transcription Regulatory Factor X (RFX) is predicted to bind in an islet-specific manner. Genetic variants that increase T2D risk are predicted to disrupt RFX binding, providing a molecular mechanism to explain how the genome can influence the epigenome, modulating gene expression and ultimately T2D risk. Genome-wide association studies (GWAS) have identified >100 independent SNPs that modulate the risk of type 2 diabetes (T2D) and related traits. However, the pathogenic mechanisms of most of these SNPs remain elusive. Here, we examined genomic, epigenomic, and transcriptomic profiles in human pancreatic islets to understand the links between genetic variation, chromatin landscape, and gene expression in the context of T2D. We first integrated genome and transcriptome variation across 112 islet samples to produce dense cis-expression quantitative trait loci (cis-eQTL) maps. Additional integration with chromatin-state maps for islets and other diverse tissue types revealed that cis-eQTLs for islet-specific genes are specifically and significantly enriched in islet stretch enhancers. High-resolution chromatin accessibility profiling using assay for transposase-accessible chromatin sequencing (ATAC-seq) in two islet samples enabled us to identify specific transcription factor (TF) footprints embedded in active regulatory elements, which are highly enriched for islet cis-eQTL. Aggregate allelic bias signatures in TF footprints enabled us de novo to reconstruct TF binding affinities genetically, which support the high-quality nature of the TF footprint predictions. Interestingly, we found that T2D GWAS loci were strikingly and specifically enriched in islet Regulatory Factor X (RFX) footprints. Remarkably, within and across independent loci, T2D risk alleles that overlap with RFX footprints uniformly disrupt the RFX motifs at high-information content positions. Together, these results suggest that common regulatory variations have shaped islet TF footprints and the transcriptome and that a confluent RFX regulatory grammar plays a significant role in the genetic component of T2D predisposition.


Nucleic Acids Research | 2014

ChIP-Enrich: gene set enrichment testing for ChIP-seq data

Ryan P. Welch; Chee Lee; Paul M. Imbriano; Snehal Patil; Terry E. Weymouth; R. Alex Smith; Laura J. Scott; Maureen A. Sartor

Gene set enrichment testing can enhance the biological interpretation of ChIP-seq data. Here, we develop a method, ChIP-Enrich, for this analysis which empirically adjusts for gene locus length (the length of the gene body and its surrounding non-coding sequence). Adjustment for gene locus length is necessary because it is often positively associated with the presence of one or more peaks and because many biologically defined gene sets have an excess of genes with longer or shorter gene locus lengths. Unlike alternative methods, ChIP-Enrich can account for the wide range of gene locus length-to-peak presence relationships (observed in ENCODE ChIP-seq data sets). We show that ChIP-Enrich has a well-calibrated type I error rate using permuted ENCODE ChIP-seq data sets; in contrast, two commonly used gene set enrichment methods, Fishers exact test and the binomial test implemented in Genomic Regions Enrichment of Annotations Tool (GREAT), can have highly inflated type I error rates and biases in ranking. We identify DNA-binding proteins, including CTCF, JunD and glucocorticoid receptor α (GRα), that show different enrichment patterns for peaks closer to versus further from transcription start sites. We also identify known and potential new biological functions of GRα. ChIP-Enrich is available as a web interface (http://chip-enrich.med.umich.edu) and Bioconductor package.


PLOS Genetics | 2017

Common, low-frequency, and rare genetic variants associated with lipoprotein subclasses and triglyceride measures in Finnish men from the METSIM study

James P. Davis; Jeroen R. Huyghe; Adam E. Locke; Anne U. Jackson; Xueling Sim; Heather M. Stringham; Tanya M. Teslovich; Ryan P. Welch; Christian Fuchsberger; Peter S. Chines; Antti J. Kangas; Pasi Soininen; Mika Ala-Korpela; Johanna Kuusisto; Francis S. Collins; Markku Laakso; Michael Boehnke; Karen L. Mohlke

Lipid and lipoprotein subclasses are associated with metabolic and cardiovascular diseases, yet the genetic contributions to variability in subclass traits are not fully understood. We conducted single-variant and gene-based association tests between 15.1M variants from genome-wide and exome array and imputed genotypes and 72 lipid and lipoprotein traits in 8,372 Finns. After accounting for 885 variants at 157 previously identified lipid loci, we identified five novel signals near established loci at HIF3A, ADAMTS3, PLTP, LCAT, and LIPG. Four of the signals were identified with a low-frequency (0.005<minor allele frequency [MAF]<0.05) or rare (MAF<0.005) variant, including Arg123His in LCAT. Gene-based associations (P<10−10) support a role for coding variants in LIPC and LIPG with lipoprotein subclass traits. 30 established lipid-associated loci had a stronger association for a subclass trait than any conventional trait. These novel association signals provide further insight into the molecular basis of dyslipidemia and the etiology of metabolic disorders.


Diabetes | 2017

A Type 2 Diabetes–Associated Functional Regulatory Variant in a Pancreatic Islet Enhancer at the ADCY5 Locus

Tamara S. Roman; Maren E. Cannon; Swarooparani Vadlamudi; Martin L. Buchkovich; Brooke N. Wolford; Ryan P. Welch; Mario A. Morken; Grace J. Kwon; Arushi Varshney; Romy Kursawe; Ying Wu; Anne U. Jackson; Michael R. Erdos; Johanna Kuusisto; Markku Laakso; Laura J. Scott; Michael Boehnke; Francis S. Collins; Stephen C. J. Parker; Michael L. Stitzel; Karen L. Mohlke

Molecular mechanisms remain unknown for most type 2 diabetes genome-wide association study identified loci. Variants associated with type 2 diabetes and fasting glucose levels reside in introns of ADCY5, a gene that encodes adenylate cyclase 5. Adenylate cyclase 5 catalyzes the production of cyclic AMP, which is a second messenger molecule involved in cell signaling and pancreatic β-cell insulin secretion. We demonstrated that type 2 diabetes risk alleles are associated with decreased ADCY5 expression in human islets and examined candidate variants for regulatory function. rs11708067 overlaps a predicted enhancer region in pancreatic islets. The type 2 diabetes risk rs11708067-A allele showed fewer H3K27ac ChIP-seq reads in human islets, lower transcriptional activity in reporter assays in rodent β-cells (rat 832/13 and mouse MIN6), and increased nuclear protein binding compared with the rs11708067-G allele. Homozygous deletion of the orthologous enhancer region in 832/13 cells resulted in a 64% reduction in expression level of Adcy5, but not adjacent gene Sec22a, and a 39% reduction in insulin secretion. Together, these data suggest that rs11708067-A risk allele contributes to type 2 diabetes by disrupting an islet enhancer, which results in reduced ADCY5 expression and impaired insulin secretion.


Human Molecular Genetics | 2018

Identification of seven novel loci associated with amino acid levels using single-variant and gene-based tests in 8545 Finnish men from the METSIM study

Tanya M. Teslovich; Daniel Seung Kim; Xianyong Yin; Alena Stančáková; Anne U. Jackson; Matthias Wielscher; Adam C. Naj; John Perry; Jeroen R. Huyghe; Heather M. Stringham; James P. Davis; Chelsea K. Raulerson; Ryan P. Welch; Christian Fuchsberger; Adam E. Locke; Xueling Sim; Peter S. Chines; Antti J. Kangas; Pasi Soininen; Mika Ala-Korpela; Vilmundur Gudnason; Solomon K. Musani; Marjo-Riitta Järvelin; Gerard D. Schellenberg; Elizabeth K. Speliotes; Johanna Kuusisto; Francis S. Collins; Michael Boehnke; Markku Laakso; Karen L. Mohlke

Comprehensive metabolite profiling captures many highly heritable traits, including amino acid levels, which are potentially sensitive biomarkers for disease pathogenesis. To better understand the contribution of genetic variation to amino acid levels, we performed single variant and gene-based tests of association between nine serum amino acids (alanine, glutamine, glycine, histidine, isoleucine, leucine, phenylalanine, tyrosine, and valine) and 16.6 million genotyped and imputed variants in 8545 non-diabetic Finnish men from the METabolic Syndrome In Men (METSIM) study with replication in Northern Finland Birth Cohort (NFBC1966). We identified five novel loci associated with amino acid levels (P = < 5×10-8): LOC157273/PPP1R3B with glycine (rs9987289, P = 2.3×10-26); ZFHX3 (chr16:73326579, minor allele frequency (MAF) = 0.42%, P = 3.6×10-9), LIPC (rs10468017, P = 1.5×10-8), and WWOX (rs9937914, P = 3.8×10-8) with alanine; and TRIB1 with tyrosine (rs28601761, P = 8×10-9). Gene-based tests identified two novel genes harboring missense variants of MAF <1% that show aggregate association with amino acid levels: PYCR1 with glycine (Pgene = 1.5×10-6) and BCAT2 with valine (Pgene = 7.4×10-7); neither gene was implicated by single variant association tests. These findings are among the first applications of gene-based tests to identify new loci for amino acid levels. In addition to the seven novel gene associations, we identified five independent signals at established amino acid loci, including two rare variant signals at GLDC (rs138640017, MAF=0.95%, Pconditional = 5.8×10-40) with glycine levels and HAL (rs141635447, MAF = 0.46%, Pconditional = 9.4×10-11) with histidine levels. Examination of all single variant association results in our data revealed a strong inverse relationship between effect size and MAF (Ptrend<0.001). These novel signals provide further insight into the molecular mechanisms of amino acid metabolism and potentially, their perturbations in disease.


bioRxiv | 2017

Imputation aware tag SNP selection to improve power for multi-ethnic association studies

Genevieve L Wojcik; Christian Fuchsberger; Ryan P. Welch; Alicia R. Martin; Suyash Shringarpure; Christopher S. Carlson; Gonçalo R. Abecasis; Hyun Min Kang; Michael Boehnke; Carlos Bustamante; Christopher R. Gignoux; Eimear E. Kenny

The emergence of very large cohorts in genomic research has facilitated a focus on genotype-imputation strategies to power rare variant association. Consequently, a new generation of genotyping arrays are being developed designed with tag single nucleotide polymorphisms (SNPs) to improve rare variant imputation. Selection of these tag SNPs poses several challenges as rare variants tend to be continentally-or even population-specific and reflect fine-scale linkage disequilibrium (LD) structure impacted by recent demographic events. To explore the landscape of tag-able variation and guide design considerations for large-cohort and biobank arrays, we developed a novel pipeline to select tag SNPs using the 26 population reference panel from Phase of the 1000 Genomes Project. We evaluate our approach using leave-one-out internal validation via standard imputation methods that allows the direct comparison of tag SNP performance by estimating the correlation of the imputed and real genotypes for each iteration of potential array sites. We show how this approach allows for an assessment of array design and performance that can take advantage of the development of deeper and more diverse sequenced reference panels. We quantify the impact of demography on tag SNP performance across populations and provide population-specific guidelines for tag SNP selection. We also examine array design strategies that target single populations versus multi-ethnic cohorts, and demonstrate a boost in performance for the latter can be obtained by prioritizing tag SNPs that contribute information across multiple populations simultaneously. Finally, we demonstrate the utility of improved array design to provide meaningful improvements in power, particularly in trans-ethnic studies. The unified framework presented will enable investigators to make informed decisions for the design of new arrays, and help empower the next phase of rare variant association for global health.

Collaboration


Dive into the Ryan P. Welch's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Francis S. Collins

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter S. Chines

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karen L. Mohlke

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Markku Laakso

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Michael R. Erdos

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge