Network


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

Hotspot


Dive into the research topics where David L. Aylor is active.

Publication


Featured researches published by David L. Aylor.


Genome Research | 2011

Genetic analysis of complex traits in the emerging Collaborative Cross

David L. Aylor; William Valdar; Wendy Foulds-Mathes; Ryan J. Buus; Ricardo A. Verdugo; Ralph S. Baric; Martin T. Ferris; Jeffrey A. Frelinger; Mark T. Heise; Matt Frieman; Lisa E. Gralinski; Timothy A. Bell; John D. Didion; Kunjie Hua; Derrick L. Nehrenberg; Christine L. Powell; Jill Steigerwalt; Yuying Xie; Samir N. Kelada; Francis S. Collins; Ivana V. Yang; David A. Schwartz; Lisa A. Branstetter; Elissa J. Chesler; Darla R. Miller; Jason S. Spence; Eric Yi Liu; Leonard McMillan; Abhishek Sarkar; Jeremy Wang

The Collaborative Cross (CC) is a mouse recombinant inbred strain panel that is being developed as a resource for mammalian systems genetics. Here we describe an experiment that uses partially inbred CC lines to evaluate the genetic properties and utility of this emerging resource. Genome-wide analysis of the incipient strains reveals high genetic diversity, balanced allele frequencies, and dense, evenly distributed recombination sites-all ideal qualities for a systems genetics resource. We map discrete, complex, and biomolecular traits and contrast two quantitative trait locus (QTL) mapping approaches. Analysis based on inferred haplotypes improves power, reduces false discovery, and provides information to identify and prioritize candidate genes that is unique to multifounder crosses like the CC. The number of expression QTLs discovered here exceeds all previous efforts at eQTL mapping in mice, and we map local eQTL at 1-Mb resolution. We demonstrate that the genetic diversity of the CC, which derives from random mixing of eight founder strains, results in high phenotypic diversity and enhances our ability to map causative loci underlying complex disease-related traits.


Bioinformatics | 2006

SNAP: Combine and Map modules for multilocus population genetic analysis

David L. Aylor; Eric W. Price; Ignazio Carbone

We have added two software tools to our Suite of Nucleotide Analysis Programs (SNAP) for working with DNA sequences sampled from populations. SNAP Map collapses DNA sequence data into unique haplotypes, extracts variable sites and manipulates output into multiple formats for input into existing software packages for evolutionary analyses. Map collapses DNA sequence data into unique haplotypes, extracts variable sites and manipulates output into multiple formats for input into existing software packages for evolutionary analyses. Map includes novel features such as recoding insertions or deletions, including or excluding variable sites that violate an infinite-sites model and the option of collapsing sequences with corresponding phenotypic information, important in testing for significant haplotype-phenotype associations. SNAP Combine merges multiple DNA sequence alignments into a single multiple alignment file. The resulting file can be the union or intersection of the input files. SNAP Combine currently reads from and writes to several sequence alignment file formats including both sequential and interleaved formats. Combine also keeps track of the start and end positions of each separate alignment file allowing the user to exclude variable sites or taxa, important in creating input files for multilocus analyses.


PLOS Pathogens | 2013

Modeling Host Genetic Regulation of Influenza Pathogenesis in the Collaborative Cross

Martin T. Ferris; David L. Aylor; Daniel Bottomly; Alan C. Whitmore; Lauri D. Aicher; Timothy A. Bell; Birgit G. Bradel-Tretheway; Janine T. Bryan; Ryan J. Buus; Lisa E. Gralinski; Bart L. Haagmans; Leonard McMillan; Darla R. Miller; Elizabeth Rosenzweig; William Valdar; Jeremy Wang; Gary A. Churchill; David W. Threadgill; Shannon McWeeney; Michael G. Katze; Fernando Pardo-Manuel de Villena; Ralph S. Baric; Mark T. Heise

Genetic variation contributes to host responses and outcomes following infection by influenza A virus or other viral infections. Yet narrow windows of disease symptoms and confounding environmental factors have made it difficult to identify polymorphic genes that contribute to differential disease outcomes in human populations. Therefore, to control for these confounding environmental variables in a system that models the levels of genetic diversity found in outbred populations such as humans, we used incipient lines of the highly genetically diverse Collaborative Cross (CC) recombinant inbred (RI) panel (the pre-CC population) to study how genetic variation impacts influenza associated disease across a genetically diverse population. A wide range of variation in influenza disease related phenotypes including virus replication, virus-induced inflammation, and weight loss was observed. Many of the disease associated phenotypes were correlated, with viral replication and virus-induced inflammation being predictors of virus-induced weight loss. Despite these correlations, pre-CC mice with unique and novel disease phenotype combinations were observed. We also identified sets of transcripts (modules) that were correlated with aspects of disease. In order to identify how host genetic polymorphisms contribute to the observed variation in disease, we conducted quantitative trait loci (QTL) mapping. We identified several QTL contributing to specific aspects of the host response including virus-induced weight loss, titer, pulmonary edema, neutrophil recruitment to the airways, and transcriptional expression. Existing whole-genome sequence data was applied to identify high priority candidate genes within QTL regions. A key host response QTL was located at the site of the known anti-influenza Mx1 gene. We sequenced the coding regions of Mx1 in the eight CC founder strains, and identified a novel Mx1 allele that showed reduced ability to inhibit viral replication, while maintaining protection from weight loss.


Nature Genetics | 2015

Analyses of allele-specific gene expression in highly divergent mouse crosses identifies pervasive allelic imbalance

James J. Crowley; Vasyl Zhabotynsky; Wei Sun; Shunping Huang; Isa Kemal Pakatci; Yunjung Kim; Jeremy R. Wang; Andrew P. Morgan; John D. Calaway; David L. Aylor; Zaining Yun; Timothy A. Bell; Ryan J. Buus; Mark Calaway; John P. Didion; Terry J. Gooch; Stephanie D. Hansen; Nashiya N. Robinson; Ginger D. Shaw; Jason S. Spence; Corey R. Quackenbush; Cordelia J. Barrick; Randal J. Nonneman; Kyungsu Kim; James Xenakis; Yuying Xie; William Valdar; Alan B. Lenarcic; Wei Wang; Catherine E. Welsh

Complex human traits are influenced by variation in regulatory DNA through mechanisms that are not fully understood. Because regulatory elements are conserved between humans and mice, a thorough annotation of cis regulatory variants in mice could aid in further characterizing these mechanisms. Here we provide a detailed portrait of mouse gene expression across multiple tissues in a three-way diallel. Greater than 80% of mouse genes have cis regulatory variation. Effects from these variants influence complex traits and usually extend to the human ortholog. Further, we estimate that at least one in every thousand SNPs creates a cis regulatory effect. We also observe two types of parent-of-origin effects, including classical imprinting and a new global allelic imbalance in expression favoring the paternal allele. We conclude that, as with humans, pervasive regulatory variation influences complex genetic traits in mice and provide a new resource toward understanding the genetic control of transcription in mammals.


G3: Genes, Genomes, Genetics | 2012

Expression quantitative trait loci for extreme host response to influenza A in pre-collaborative cross mice

Daniel Bottomly; Martin T. Ferris; Lauri D. Aicher; Elizabeth Rosenzweig; Alan C. Whitmore; David L. Aylor; Bart L. Haagmans; Lisa E. Gralinski; Birgit G. Bradel-Tretheway; Janine T. Bryan; David W. Threadgill; Fernando Pardo-Manuel de Villena; Ralph S. Baric; Michael G. Katze; Mark T. Heise; Shannon McWeeney

Outbreaks of influenza occur on a yearly basis, causing a wide range of symptoms across the human population. Although evidence exists that the host response to influenza infection is influenced by genetic differences in the host, this has not been studied in a system with genetic diversity mirroring that of the human population. Here we used mice from 44 influenza-infected pre-Collaborative Cross lines determined to have extreme phenotypes with regard to the host response to influenza A virus infection. Global transcriptome profiling identified 2671 transcripts that were significantly differentially expressed between mice that showed a severe (“high”) and mild (“low”) response to infection. Expression quantitative trait loci mapping was performed on those transcripts that were differentially expressed because of differences in host response phenotype to identify putative regulatory regions potentially controlling their expression. Twenty-one significant expression quantitative trait loci were identified, which allowed direct examination of genes associated with regulation of host response to infection. To perform initial validation of our findings, quantitative polymerase chain reaction was performed in the infected founder strains, and we were able to confirm or partially confirm more than 70% of those tested. In addition, we explored putative causal and reactive (downstream) relationships between the significantly regulated genes and others in the high or low response groups using structural equation modeling. By using systems approaches and a genetically diverse population, we were able to develop a novel framework for identifying the underlying biological subnetworks under host genetic control during influenza virus infection.


Genes & Development | 2009

Elucidation of the transcription network governing mammalian sex determination by exploiting strain-specific susceptibility to sex reversal

Steven C. Munger; David L. Aylor; Haider Ali Syed; Paul M. Magwene; David W. Threadgill; Blanche Capel

Despite the identification of some key genes that regulate sex determination, most cases of disorders of sexual development remain unexplained. Evidence suggests that the sexual fate decision in the developing gonad depends on a complex network of interacting factors that converge on a critical threshold. To elucidate the transcriptional network underlying sex determination, we took the first expression quantitative trait loci (eQTL) approach in a developing organ. We identified reproducible differences in the transcriptome of the embryonic day 11.5 (E11.5) XY gonad between C57BL/6J (B6) and 129S1/SvImJ (129S1), indicating that the reported sensitivity of B6 to sex reversal is consistent with a higher expression of a female-like transcriptome in B6. Gene expression is highly variable in F2 XY gonads from B6 and 129S1 intercrosses, yet strong correlations emerged. We estimated the F2 coexpression network and predicted roles for genes of unknown function based on their connectivity and position within the network. A genetic analysis of the F2 population detected autosomal regions that control the expression of many sex-related genes, including Sry (sex-determining region of the Y chromosome) and Sox9 (Sry-box containing gene 9), the key regulators of male sex determination. Our results reveal the complex transcription architecture underlying sex determination, and provide a mechanism by which individuals may be sensitized for sex reversal.


G3: Genes, Genomes, Genetics | 2012

Genetic Analysis of Hematological Parameters in Incipient Lines of the Collaborative Cross

Samir N. Kelada; David L. Aylor; Bailey C. Peck; Joseph F. Ryan; Urraca Tavarez; Ryan J. Buus; Darla R. Miller; Elissa J. Chesler; David W. Threadgill; Gary A. Churchill; Fernando Pardo-Manuel de Villena; Francis S. Collins

Hematological parameters, including red and white blood cell counts and hemoglobin concentration, are widely used clinical indicators of health and disease. These traits are tightly regulated in healthy individuals and are under genetic control. Mutations in key genes that affect hematological parameters have important phenotypic consequences, including multiple variants that affect susceptibility to malarial disease. However, most variation in hematological traits is continuous and is presumably influenced by multiple loci and variants with small phenotypic effects. We used a newly developed mouse resource population, the Collaborative Cross (CC), to identify genetic determinants of hematological parameters. We surveyed the eight founder strains of the CC and performed a mapping study using 131 incipient lines of the CC. Genome scans identified quantitative trait loci for several hematological parameters, including mean red cell volume (Chr 7 and Chr 14), white blood cell count (Chr 18), percent neutrophils/lymphocytes (Chr 11), and monocyte number (Chr 1). We used evolutionary principles and unique bioinformatics resources to reduce the size of candidate intervals and to view functional variation in the context of phylogeny. Many quantitative trait loci regions could be narrowed sufficiently to identify a small number of promising candidate genes. This approach not only expands our knowledge about hematological traits but also demonstrates the unique ability of the CC to elucidate the genetic architecture of complex traits.


American Journal of Physiology-endocrinology and Metabolism | 2011

Architecture of energy balance traits in emerging lines of the Collaborative Cross

Wendy Foulds Mathes; David L. Aylor; Darla R. Miller; Gary A. Churchill; Elissa J. Chesler; Fernando Pardo-Manuel de Villena; David W. Threadgill; Daniel Pomp

The potential utility of the Collaborative Cross (CC) mouse resource was evaluated to better understand complex traits related to energy balance. A primary focus was to examine if genetic diversity in emerging CC lines (pre-CC) would translate into equivalent phenotypic diversity. Second, we mapped quantitative trait loci (QTL) for 15 metabolism- and exercise-related phenotypes in this population. We evaluated metabolic and voluntary exercise traits in 176 pre-CC lines, revealing phenotypic variation often exceeding that seen across the eight founder strains from which the pre-CC was derived. Many phenotypic correlations existing within the founder strains were no longer significant in the pre-CC population, potentially representing reduced linkage disequilibrium (LD) of regions harboring multiple genes with effects on energy balance or disruption of genetic structure of extant inbred strains with substantial shared ancestry. QTL mapping revealed five significant and eight suggestive QTL for body weight (Chr 4, 7.54 Mb; CI 3.32-10.34 Mb; Bwq14), body composition, wheel running (Chr 16, 33.2 Mb; CI 32.5-38.3 Mb), body weight change in response to exercise (1: Chr 6, 77.7Mb; CI 72.2-83.4 Mb and 2: Chr 6, 42.8 Mb; CI 39.4-48.1 Mb), and food intake during exercise (Chr 12, 85.1 Mb; CI 82.9-89.0 Mb). Some QTL overlapped with previously mapped QTL for similar traits, whereas other QTL appear to represent novel loci. These results suggest that the CC will be a powerful, high-precision tool for examining the genetic architecture of complex traits such as those involved in regulation of energy balance.


PLOS Genetics | 2015

Genome Wide Identification of SARS-CoV Susceptibility Loci Using the Collaborative Cross.

Lisa E. Gralinski; Martin T. Ferris; David L. Aylor; Alan C. Whitmore; Richard Green; Matthew B. Frieman; Damon Deming; Vineet D. Menachery; Darla R. Miller; Ryan J. Buus; Timothy A. Bell; Gary A. Churchill; David W. Threadgill; Michael G. Katze; Leonard McMillan; William Valdar; Mark T. Heise; Fernando Pardo-Manuel de Villena; Ralph S. Baric

New systems genetics approaches are needed to rapidly identify host genes and genetic networks that regulate complex disease outcomes. Using genetically diverse animals from incipient lines of the Collaborative Cross mouse panel, we demonstrate a greatly expanded range of phenotypes relative to classical mouse models of SARS-CoV infection including lung pathology, weight loss and viral titer. Genetic mapping revealed several loci contributing to differential disease responses, including an 8.5Mb locus associated with vascular cuffing on chromosome 3 that contained 23 genes and 13 noncoding RNAs. Integrating phenotypic and genetic data narrowed this region to a single gene, Trim55, an E3 ubiquitin ligase with a role in muscle fiber maintenance. Lung pathology and transcriptomic data from mice genetically deficient in Trim55 were used to validate its role in SARS-CoV-induced vascular cuffing and inflammation. These data establish the Collaborative Cross platform as a powerful genetic resource for uncovering genetic contributions of complex traits in microbial disease severity, inflammation and virus replication in models of outbred populations.


Genes and Immunity | 2014

Using the emerging Collaborative Cross to probe the immune system

J. Phillippi; Yuying Xie; Darla R. Miller; Timothy A. Bell; Zhaojun Zhang; Alan B. Lenarcic; David L. Aylor; S. H. Krovi; David W. Threadgill; F. Pardo-Manuel De Villena; Wei Wang; William Valdar; Jeffrey A. Frelinger

The Collaborative Cross (CC) is an emerging panel of recombinant inbred (RI) mouse strains. Each strain is genetically distinct but all descended from the same eight inbred founders. In 66 strains from incipient lines of the CC (pre-CC), as well as the 8 CC founders and some of their F1 offspring, we examined subsets of lymphocytes and antigen-presenting cells. We found significant variation among the founders, with even greater diversity in the pre-CC. Genome-wide association using inferred haplotypes detected highly significant loci controlling B-to-T cell ratio, CD8 T-cell numbers, CD11c and CD23 expression. Comparison of overall strain effects in the CC founders with strain effects at QTL in the pre-CC revealed sharp contrasts in the genetic architecture of two traits with significant loci: variation in CD23 can be explained largely by additive genetics at one locus, whereas variation in B-to-T ratio has a more complex etiology. For CD23, we found a strong QTL whose confidence interval contained the CD23 structural gene Fcer2a. Our data on the pre-CC demonstrate the utility of the CC for studying immunophenotypes and the value of integrating founder, CC and F1 data. The extreme immunophenotypes observed could have pleiotropic effects in other CC experiments.

Collaboration


Dive into the David L. Aylor's collaboration.

Top Co-Authors

Avatar

Elissa J. Chesler

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Darla R. Miller

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Fernando Pardo-Manuel de Villena

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Timothy A. Bell

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leonard McMillan

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew P. Morgan

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Francis S. Collins

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge