Network


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

Hotspot


Dive into the research topics where Leah C. Kottyan is active.

Publication


Featured researches published by Leah C. Kottyan.


Frontiers in Genetics | 2014

Imputation and quality control steps for combining multiple genome-wide datasets

Shefali S. Verma; Mariza de Andrade; Gerard Tromp; Helena Kuivaniemi; Elizabeth W. Pugh; Bahram Namjou-Khales; Shubhabrata Mukherjee; Gail P. Jarvik; Leah C. Kottyan; Amber A. Burt; Yuki Bradford; Gretta D. Armstrong; Kimberly Derr; Dana C. Crawford; Jonathan L. Haines; Rongling Li; David R. Crosslin; Marylyn D. Ritchie

The electronic MEdical Records and GEnomics (eMERGE) network brings together DNA biobanks linked to electronic health records (EHRs) from multiple institutions. Approximately 51,000 DNA samples from distinct individuals have been genotyped using genome-wide SNP arrays across the nine sites of the network. The eMERGE Coordinating Center and the Genomics Workgroup developed a pipeline to impute and merge genomic data across the different SNP arrays to maximize sample size and power to detect associations with a variety of clinical endpoints. The 1000 Genomes cosmopolitan reference panel was used for imputation. Imputation results were evaluated using the following metrics: accuracy of imputation, allelic R2 (estimated correlation between the imputed and true genotypes), and the relationship between allelic R2 and minor allele frequency. Computation time and memory resources required by two different software packages (BEAGLE and IMPUTE2) were also evaluated. A number of challenges were encountered due to the complexity of using two different imputation software packages, multiple ancestral populations, and many different genotyping platforms. We present lessons learned and describe the pipeline implemented here to impute and merge genomic data sets. The eMERGE imputed dataset will serve as a valuable resource for discovery, leveraging the clinical data that can be mined from the EHR.


Frontiers in Genetics | 2014

Phenome-wide association study (PheWAS) in EMR-linked pediatric cohorts, genetically links PLCL1 to speech language development and IL5-IL13 to Eosinophilic Esophagitis.

Bahram Namjou; Keith Marsolo; Robert J. Caroll; Joshua C. Denny; Marylyn D. Ritchie; Shefali S. Verma; Todd Lingren; Aleksey Porollo; Beth L. Cobb; Cassandra Perry; Leah C. Kottyan; Marc E. Rothenberg; Susan D. Thompson; Ingrid A. Holm; Isaac S. Kohane; John B. Harley

Objective: We report the first pediatric specific Phenome-Wide Association Study (PheWAS) using electronic medical records (EMRs). Given the early success of PheWAS in adult populations, we investigated the feasibility of this approach in pediatric cohorts in which associations between a previously known genetic variant and a wide range of clinical or physiological traits were evaluated. Although computationally intensive, this approach has potential to reveal disease mechanistic relationships between a variant and a network of phenotypes. Method: Data on 5049 samples of European ancestry were obtained from the EMRs of two large academic centers in five different genotyped cohorts. Recently, these samples have undergone whole genome imputation. After standard quality controls, removing missing data and outliers based on principal components analyses (PCA), 4268 samples were used for the PheWAS study. We scanned for associations between 2476 single-nucleotide polymorphisms (SNP) with available genotyping data from previously published GWAS studies and 539 EMR-derived phenotypes. The false discovery rate was calculated and, for any new PheWAS findings, a permutation approach (with up to 1,000,000 trials) was implemented. Results: This PheWAS found a variety of common variants (MAF > 10%) with prior GWAS associations in our pediatric cohorts including Juvenile Rheumatoid Arthritis (JRA), Asthma, Autism and Pervasive Developmental Disorder (PDD) and Type 1 Diabetes with a false discovery rate < 0.05 and power of study above 80%. In addition, several new PheWAS findings were identified including a cluster of association near the NDFIP1 gene for mental retardation (best SNP rs10057309, p = 4.33 × 10−7, OR = 1.70, 95%CI = 1.38 − 2.09); association near PLCL1 gene for developmental delays and speech disorder [best SNP rs1595825, p = 1.13 × 10−8, OR = 0.65(0.57 − 0.76)]; a cluster of associations in the IL5-IL13 region with Eosinophilic Esophagitis (EoE) [best at rs12653750, p = 3.03 × 10−9, OR = 1.73 95%CI = (1.44 − 2.07)], previously implicated in asthma, allergy, and eosinophilia; and association of variants in GCKR and JAZF1 with allergic rhinitis in our pediatric cohorts [best SNP rs780093, p = 2.18 × 10−5, OR = 1.39, 95%CI = (1.19 − 1.61)], previously demonstrated in metabolic disease and diabetes in adults. Conclusion: The PheWAS approach with re-mapping ICD-9 structured codes for our European-origin pediatric cohorts, as with the previous adult studies, finds many previously reported associations as well as presents the discovery of associations with potentially important clinical implications.


Journal of Leukocyte Biology | 2012

Genetic susceptibility to lupus: the biological basis of genetic risk found in B cell signaling pathways

Samuel E. Vaughn; Leah C. Kottyan; Melissa E. Munroe; John B. Harley

Over 50 genetic variants have been statistically associated with the development of SLE (or lupus). Each genetic association is a key component of a pathway to lupus pathogenesis, the majority of which requires further mechanistic studies to understand the functional changes to cellular physiology. Whereas their use in clinical practice has yet to be established, these genes guide efforts to develop more specific therapeutic approaches. The BCR signaling pathways are rich in lupus susceptibility genes and may well provide novel opportunities for the understanding and clinical treatment of this complex disease.


The New England Journal of Medicine | 2017

Genetic Associations with Gestational Duration and Spontaneous Preterm Birth

Ge Zhang; Bjarke Feenstra; Jonas Bacelis; Xueping Liu; Lisa M. Muglia; Julius Juodakis; Daniel Miller; Nadia K. Litterman; Pan-Pan Jiang; Laura Russell; David A. Hinds; Youna Hu; Matthew T. Weirauch; Xiaoting Chen; Arun R. Chavan; Günter P. Wagner; Mihaela Pavlicev; Mauris C. Nnamani; Jamie Maziarz; Minna K. Karjalainen; Mika Rämet; Verena Sengpiel; Frank Geller; Heather A. Boyd; Aarno Palotie; Allison M. Momany; Bruce Bedell; Kelli K. Ryckman; Johanna M. Huusko; Carmy Forney

BACKGROUND Despite evidence that genetic factors contribute to the duration of gestation and the risk of preterm birth, robust associations with genetic variants have not been identified. We used large data sets that included the gestational duration to determine possible genetic associations. METHODS We performed a genomewide association study in a discovery set of samples obtained from 43,568 women of European ancestry using gestational duration as a continuous trait and term or preterm (<37 weeks) birth as a dichotomous outcome. We used samples from three Nordic data sets (involving a total of 8643 women) to test for replication of genomic loci that had significant genomewide association (P<5.0×10‐8) or an association with suggestive significance (P<1.0×10‐6) in the discovery set. RESULTS In the discovery and replication data sets, four loci (EBF1, EEFSEC, AGTR2, and WNT4) were significantly associated with gestational duration. Functional analysis showed that an implicated variant in WNT4 alters the binding of the estrogen receptor. The association between variants in ADCY5 and RAP2C and gestational duration had suggestive significance in the discovery set and significant evidence of association in the replication sets; these variants also showed genomewide significance in a joint analysis. Common variants in EBF1, EEFSEC, and AGTR2 showed association with preterm birth with genomewide significance. An analysis of mother–infant dyads suggested that these variants act at the level of the maternal genome. CONCLUSIONS In this genomewide association study, we found that variants at the EBF1, EEFSEC, AGTR2, WNT4, ADCY5, and RAP2C loci were associated with gestational duration and variants at the EBF1, EEFSEC, and AGTR2 loci with preterm birth. Previously established roles of these genes in uterine development, maternal nutrition, and vascular control support their mechanistic involvement. (Funded by the March of Dimes and others.)


Human Molecular Genetics | 2015

The IRF5–TNPO3 association with systemic lupus erythematosus has two components that other autoimmune disorders variably share

Leah C. Kottyan; Erin E. Zoller; Jessica Bene; Xiaoming Lu; Jennifer A. Kelly; Andrew Rupert; Christopher J. Lessard; Samuel E. Vaughn; Miranda C. Marion; Matthew T. Weirauch; Bahram Namjou; Adam Adler; Astrid Rasmussen; Stuart B. Glenn; Courtney G. Montgomery; Gideon M. Hirschfield; Gang Xie; Catalina Coltescu; Christopher I. Amos; He Li; John A. Ice; Swapan K. Nath; Xavier Mariette; Simon Bowman; Maureen Rischmueller; Susan Lester; Johan G. Brun; Lasse G. Gøransson; Erna Harboe; Roald Omdal

Exploiting genotyping, DNA sequencing, imputation and trans-ancestral mapping, we used Bayesian and frequentist approaches to model the IRF5-TNPO3 locus association, now implicated in two immunotherapies and seven autoimmune diseases. Specifically, in systemic lupus erythematosus (SLE), we resolved separate associations in the IRF5 promoter (all ancestries) and with an extended European haplotype. We captured 3230 IRF5-TNPO3 high-quality, common variants across 5 ethnicities in 8395 SLE cases and 7367 controls. The genetic effect from the IRF5 promoter can be explained by any one of four variants in 5.7 kb (P-valuemeta = 6 × 10(-49); OR = 1.38-1.97). The second genetic effect spanned an 85.5-kb, 24-variant haplotype that included the genes IRF5 and TNPO3 (P-valuesEU = 10(-27)-10(-32), OR = 1.7-1.81). Many variants at the IRF5 locus with previously assigned biological function are not members of either final credible set of potential causal variants identified herein. In addition to the known biologically functional variants, we demonstrated that the risk allele of rs4728142, a variant in the promoter among the lowest frequentist probability and highest Bayesian posterior probability, was correlated with IRF5 expression and differentially binds the transcription factor ZBTB3. Our analytical strategy provides a novel framework for future studies aimed at dissecting etiological genetic effects. Finally, both SLE elements of the statistical model appear to operate in Sjögrens syndrome and systemic sclerosis whereas only the IRF5-TNPO3 gene-spanning haplotype is associated with primary biliary cirrhosis, demonstrating the nuance of similarity and difference in autoimmune disease risk mechanisms at IRF5-TNPO3.


Circulation-cardiovascular Genetics | 2014

Whole exome sequencing for familial bicuspid aortic valve identifies putative variants.

Lisa J. Martin; Valentina Pilipenko; Kenneth M. Kaufman; Linda H. Cripe; Leah C. Kottyan; Mehdi Keddache; Phillip Dexheimer; Matthew T. Weirauch; D. Woodrow Benson

Background—Bicuspid aortic valve (BAV) is the most common congenital cardiovascular malformation. Although highly heritable, few causal variants have been identified. The purpose of this study was to identify genetic variants underlying BAV by whole exome sequencing a multiplex BAV kindred. Methods and Results—Whole exome sequencing was performed on 17 individuals from a single family (BAV=3; other cardiovascular malformation, 3). Postvariant calling error control metrics were established after examining the relationship between Mendelian inheritance error rate and coverage, quality score, and call rate. To determine the most effective approach to identifying susceptibility variants from among 54 674 variants passing error control metrics, we evaluated 3 variant selection strategies frequently used in whole exome sequencing studies plus extended family linkage. No putative rare, high-effect variants were identified in all affected but no unaffected individuals. Eight high-effect variants were identified by ≥2 of the commonly used selection strategies; however, these were either common in the general population (>10%) or present in the majority of the unaffected family members. However, using extended family linkage, 3 synonymous variants were identified; all 3 variants were identified by at least one other strategy. Conclusions—These results suggest that traditional whole exome sequencing approaches, which assume causal variants alter coding sense, may be insufficient for BAV and other complex traits. Identification of disease-associated variants is facilitated by the use of segregation within families.


Journal of Translational Medicine | 2012

IRF5 gene polymorphisms in melanoma

Lorenzo Uccellini; Valeria De Giorgi; Yingdong Zhao; Barbara Tumaini; Narnygerel Erdenebileg; Mark E Dudley; Sara Tomei; Davide Bedognetti; Maria Libera Ascierto; Qiuzhen Liu; Richard Simon; Leah C. Kottyan; Kenneth M. Kaufman; John B. Harley; Ena Wang; Steven A. Rosenberg; Francesco M. Marincola

BackgroundInterferon regulatory factor (IRF)-5 is a transcription factor involved in type I interferon signaling whose germ line variants have been associated with autoimmune pathogenesis. Since relationships have been observed between development of autoimmunity and responsiveness of melanoma to several types of immunotherapy, we tested whether polymorphisms of IRF5 are associated with responsiveness of melanoma to adoptive therapy with tumor infiltrating lymphocytes (TILs).Methods140 TILs were genotyped for four single nucleotide polymorphisms (rs10954213, rs11770589, rs6953165, rs2004640) and one insertion-deletion in the IRF5 gene by sequencing. Gene-expression profile of the TILs, 112 parental melanoma metastases (MM) and 9 cell lines derived from some metastases were assessed by Affymetrix Human Gene ST 1.0 array.ResultsLack of A allele in rs10954213 (G > A) was associated with non-response (p < 0.005). Other polymorphisms in strong linkage disequilibrium with rs10954213 demonstrated similar trends. Genes differentially expressed in vitro between cell lines carrying or not the A allele could be applied to the transcriptional profile of 112 melanoma metastases to predict their responsiveness to therapy, suggesting that IRF5 genotype may influence immune responsiveness by affecting the intrinsic biology of melanoma.ConclusionsThis study is the first to analyze associations between melanoma immune responsiveness and IRF5 polymorphism. The results support a common genetic basis which may underline the development of autoimmunity and melanoma immune responsiveness.


Annals of the Rheumatic Diseases | 2017

Genetic architecture distinguishes systemic juvenile idiopathic arthritis from other forms of juvenile idiopathic arthritis: clinical and therapeutic implications

Michael J. Ombrello; Victoria L. Arthur; Elaine F. Remmers; Anne Hinks; Ioanna Tachmazidou; Alexei A. Grom; Dirk Foell; Alberto Martini; Marco Gattorno; Seza Ozen; Sampath Prahalad; Andrew Zeft; John F. Bohnsack; Norman T. Ilowite; Elizabeth D. Mellins; Ricardo Russo; Claudio Arnaldo Len; Maria Odete Esteves Hilário; Sheila Knupp Feitosa de Oliveira; Rae S. M. Yeung; Alan M. Rosenberg; Lucy R. Wedderburn; Jordi Anton; Johannes-Peter Haas; Angela Rösen-Wolff; K. Minden; Klaus Tenbrock; Erkan Demirkaya; Joanna Cobb; Elizabeth Baskin

Objectives Juvenile idiopathic arthritis (JIA) is a heterogeneous group of conditions unified by the presence of chronic childhood arthritis without an identifiable cause. Systemic JIA (sJIA) is a rare form of JIA characterised by systemic inflammation. sJIA is distinguished from other forms of JIA by unique clinical features and treatment responses that are similar to autoinflammatory diseases. However, approximately half of children with sJIA develop destructive, long-standing arthritis that appears similar to other forms of JIA. Using genomic approaches, we sought to gain novel insights into the pathophysiology of sJIA and its relationship with other forms of JIA. Methods We performed a genome-wide association study of 770 children with sJIA collected in nine countries by the International Childhood Arthritis Genetics Consortium. Single nucleotide polymorphisms were tested for association with sJIA. Weighted genetic risk scores were used to compare the genetic architecture of sJIA with other JIA subtypes. Results The major histocompatibility complex locus and a locus on chromosome 1 each showed association with sJIA exceeding the threshold for genome-wide significance, while 23 other novel loci were suggestive of association with sJIA. Using a combination of genetic and statistical approaches, we found no evidence of shared genetic architecture between sJIA and other common JIA subtypes. Conclusions The lack of shared genetic risk factors between sJIA and other JIA subtypes supports the hypothesis that sJIA is a unique disease process and argues for a different classification framework. Research to improve sJIA therapy should target its unique genetics and specific pathophysiological pathways.


Frontiers in Genetics | 2014

The struggle to find reliable results in exome sequencing data: filtering out Mendelian errors

Zubin Patel; Leah C. Kottyan; Sara Lazaro; Marc S. Williams; David H. Ledbetter; Gerard Tromp; Andrew Rupert; Mojtaba Kohram; Michael Wagner; Ammar Husami; Yaping Qian; C. Alexander Valencia; Kejian Zhang; Margaret K. Hostetter; John B. Harley; Kenneth M. Kaufman

Next Generation Sequencing studies generate a large quantity of genetic data in a relatively cost and time efficient manner and provide an unprecedented opportunity to identify candidate causative variants that lead to disease phenotypes. A challenge to these studies is the generation of sequencing artifacts by current technologies. To identify and characterize the properties that distinguish false positive variants from true variants, we sequenced a child and both parents (one trio) using DNA isolated from three sources (blood, buccal cells, and saliva). The trio strategy allowed us to identify variants in the proband that could not have been inherited from the parents (Mendelian errors) and would most likely indicate sequencing artifacts. Quality control measurements were examined and three measurements were found to identify the greatest number of Mendelian errors. These included read depth, genotype quality score, and alternate allele ratio. Filtering the variants on these measurements removed ~95% of the Mendelian errors while retaining 80% of the called variants. These filters were applied independently. After filtering, the concordance between identical samples isolated from different sources was 99.99% as compared to 87% before filtering. This high concordance suggests that different sources of DNA can be used in trio studies without affecting the ability to identify causative polymorphisms. To facilitate analysis of next generation sequencing data, we developed the Cincinnati Analytical Suite for Sequencing Informatics (CASSI) to store sequencing files, metadata (eg. relatedness information), file versioning, data filtering, variant annotation, and identify candidate causative polymorphisms that follow either de novo, rare recessive homozygous or compound heterozygous inheritance models. We conclude the data cleaning process improves the signal to noise ratio in terms of variants and facilitates the identification of candidate disease causative polymorphisms.


American Journal of Human Genetics | 2015

Lupus Risk Variant Increases pSTAT1 Binding and Decreases ETS1 Expression

Xiaoming Lu; Erin E. Zoller; Matthew T. Weirauch; Zhiguo Wu; Bahram Namjou; Adrienne H. Williams; Julie T. Ziegler; Mary E. Comeau; Miranda C. Marion; Stuart B. Glenn; Adam Adler; Nan Shen; Swapan K. Nath; Anne M. Stevens; Barry I. Freedman; Betty P. Tsao; Chaim O. Jacob; Diane L. Kamen; Elizabeth E. Brown; Gary S. Gilkeson; Graciela S. Alarcón; John D. Reveille; Juan-Manuel Anaya; Judith A. James; Kathy L. Sivils; Lindsey A. Criswell; Luis M. Vilá; Marta E. Alarcón-Riquelme; Michelle Petri; R. Hal Scofield

Genetic variants at chromosomal region 11q23.3, near the gene ETS1, have been associated with systemic lupus erythematosus (SLE), or lupus, in independent cohorts of Asian ancestry. Several recent studies have implicated ETS1 as a critical driver of immune cell function and differentiation, and mice deficient in ETS1 develop an SLE-like autoimmunity. We performed a fine-mapping study of 14,551 subjects from multi-ancestral cohorts by starting with genotyped variants and imputing to all common variants spanning ETS1. By constructing genetic models via frequentist and Bayesian association methods, we identified 16 variants that are statistically likely to be causal. We functionally assessed each of these variants on the basis of their likelihood of affecting transcription factor binding, miRNA binding, or chromatin state. Of the four variants that we experimentally examined, only rs6590330 differentially binds lysate from B cells. Using mass spectrometry, we found more binding of the transcription factor signal transducer and activator of transcription 1 (STAT1) to DNA near the risk allele of rs6590330 than near the non-risk allele. Immunoblot analysis and chromatin immunoprecipitation of pSTAT1 in B cells heterozygous for rs6590330 confirmed that the risk allele increased binding to the active form of STAT1. Analysis with expression quantitative trait loci indicated that the risk allele of rs6590330 is associated with decreased ETS1 expression in Han Chinese, but not other ancestral cohorts. We propose a model in which the risk allele of rs6590330 is associated with decreased ETS1 expression and increases SLE risk by enhancing the binding of pSTAT1.

Collaboration


Dive into the Leah C. Kottyan's collaboration.

Top Co-Authors

Avatar

John B. Harley

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marc E. Rothenberg

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kenneth M. Kaufman

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Matthew T. Weirauch

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lisa J. Martin

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hua He

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Daniel Miller

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Adam Adler

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Bahram Namjou

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Carmy Forney

Cincinnati Children's Hospital Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge