Network


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

Hotspot


Dive into the research topics where Daniel J. Wegner is active.

Publication


Featured researches published by Daniel J. Wegner.


Nature Methods | 2009

Quantification of rare allelic variants from pooled genomic DNA

Todd E. Druley; Francesco Vallania; Daniel J. Wegner; Katherine E. Varley; Olivia L. Knowles; Jacqueline A. Bonds; Sarah W. Robison; Scott W. Doniger; Aaron Hamvas; F. Sessions Cole; Justin C. Fay; Robi D. Mitra

We report a targeted, cost-effective method to quantify rare single-nucleotide polymorphisms from pooled human genomic DNA using second-generation sequencing. We pooled DNA from 1,111 individuals and targeted four genes to identify rare germline variants. Our base-calling algorithm, SNPSeeker, derived from large deviation theory, detected single-nucleotide polymorphisms present at frequencies below the raw error rate of the sequencing platform.


Pediatric Research | 2008

Population and Disease-Based Prevalence of the Common Mutations Associated With Surfactant Deficiency

Tami H. Garmany; Jennifer A. Wambach; Hillary B. Heins; Julie Watkins-Torry; Daniel J. Wegner; Kate W. Bennet; Ping An; Garland Land; Ola Didrik Saugstad; Howard E. Henderson; Lawrence M. Nogee; F. Sessions Cole; Aaron Hamvas

The prevalence of the common mutations in the surfactant protein-B (121ins2), surfactant protein-C (I73T), and ATP-binding cassette member A3 (E292V) genes in population-based or case-control cohorts of newborn respiratory distress syndrome (RDS) is unknown. We determined the frequencies of these mutations in ethnically diverse population and disease-based cohorts using restriction enzyme analysis (121ins2 and E292V) and a 5′ nuclease assay (I73T) in DNA samples from population-based cohorts in Missouri, Norway, South Korea, and South Africa, and from a case–control cohort of newborns with and without RDS (n = 420). We resequenced the ATP-binding cassette member A3 gene (ABCA3) in E292V carriers and computationally inferred ABCA3 haplotypes. The population-based frequencies of 121ins2, E292V, and I73T were rare (<0.4%). E292V was present in 3.8% of newborns with RDS, a 10-fold greater prevalence than in the Missouri cohort (p < 0.001). We did not identify other loss of function mutations in ABCA3 among patients with E292V that would account for their RDS. E292V occurred on a unique haplotype that was derived from a recombination of two common ABCA3 haplotypes. E292V was over-represented in newborns with RDS suggesting that E292V or its unique haplotype impart increased genetic risk for RDS.


Pediatrics | 2012

Single ABCA3 Mutations Increase Risk for Neonatal Respiratory Distress Syndrome

Jennifer A. Wambach; Daniel J. Wegner; Kelcey DePass; Hillary B. Heins; Todd E. Druley; Robi D. Mitra; Ping An; Qunyuan Zhang; Lawrence M. Nogee; F. Sessions Cole; Aaron Hamvas

BACKGROUND AND OBJECTIVE: Neonatal respiratory distress syndrome (RDS) due to pulmonary surfactant deficiency is heritable, but common variants do not fully explain disease heritability. METHODS: Using next-generation, pooled sequencing of race-stratified DNA samples from infants ≥34 weeks’ gestation with and without RDS (n = 513) and from a Missouri population-based cohort (n = 1066), we scanned all exons of 5 surfactant-associated genes and used in silico algorithms to identify functional mutations. We validated each mutation with an independent genotyping platform and compared race-stratified, collapsed frequencies of rare mutations by gene to investigate disease associations and estimate attributable risk. RESULTS: Single ABCA3 mutations were overrepresented among European-descent RDS infants (14.3% of RDS vs 3.7% of non-RDS; P = .002) but were not statistically overrepresented among African-descent RDS infants (4.5% of RDS vs 1.5% of non-RDS; P = .23). In the Missouri population-based cohort, 3.6% of European-descent and 1.5% of African-descent infants carried a single ABCA3 mutation. We found no mutations among the RDS infants and no evidence of contribution to population-based disease burden for SFTPC, CHPT1, LPCAT1, or PCYT1B. CONCLUSIONS: In contrast to lethal neonatal RDS resulting from homozygous or compound heterozygous ABCA3 mutations, single ABCA3 mutations are overrepresented among European-descent infants ≥34 weeks’ gestation with RDS and account for ∼10.9% of the attributable risk among term and late preterm infants. Although ABCA3 mutations are individually rare, they are collectively common among European- and African-descent individuals in the general population.


Pediatric Research | 2012

An intronic ABCA3 mutation that is responsible for respiratory disease

Amit Agrawal; Aaron Hamvas; F. Sessions Cole; Jennifer A. Wambach; Daniel J. Wegner; Carl Coghill; Keith Harrison; Lawrence M. Nogee

Introduction:Member A3 of the ATP-binding cassette family of transporters (ABCA3) is essential for surfactant metabolism. Nonsense, missense, frameshift, and splice-site mutations in the ABCA3 gene (ABCA3) have been reported as causes of neonatal respiratory failure (NRF) and interstitial lung disease. We tested the hypothesis that mutations in noncoding regions of ABCA3 may cause lung disease.Methods:ABCA3-specific cDNA was generated and sequenced from frozen lung tissue from a child with fatal lung disease with only one identified ABCA3 mutation. ABCA3 was sequenced from genomic DNA prepared from blood samples obtained from the proband, parents, and other children with NRF.Results:ABCA3 cDNA from the proband contained sequences derived from intron 25 that would be predicted to alter the structure and function of the ABCA3 protein. Genomic DNA sequencing revealed a heterozygous C>T transition in intron 25 trans to the known mutation, creating a new donor splice site. Seven additional infants with an ABCA3-deficient phenotype and inconclusive genetic findings had this same variant, which was not found in 2,132 control chromosomes.Discussion:These findings support that this variant is a disease-causing mutation that may account for additional cases of ABCA3 deficiency with negative genetic studies.


The Journal of Pediatrics | 2009

Inherited surfactant deficiency caused by uniparental disomy of rare mutations in the surfactant protein-B and ATP binding cassette, subfamily a, member 3 genes.

Aaron Hamvas; Lawrence M. Nogee; Daniel J. Wegner; Kelcey DePass; John Christodoulou; Bruce Bennetts; Leon R. McQuade; Peter H. Gray; Robin R. Deterding; Travis R. Carroll; Anja Kammesheidt; Laura Kasch; Shashikant Kulkarni; F. Sessions Cole

OBJECTIVE To characterize inheritance of homozygous, rare, recessive loss-of-function mutations in surfactant protein-B (SFTPB) or ATP binding cassette, subfamily A, member 3 (ABCA3) genes in newborns with lethal respiratory failure. STUDY DESIGN We resequenced genes from parents whose infants were homozygous for mutations in SFTPB or ABCA3. For infants with only 1 heterozygous parent, we performed microsatellite analysis for chromosomes 2 (SFTPB) and 16 (ABCA3). RESULTS We identified 1 infant homozygous for the g.1549C > GAA mutation (121ins2) in SFTPB for whom only the mother was heterozygous and 3 infants homozygous for mutations in ABCA3 (p.K914R, p.P147L, and c.806_7insGCT) for whom only the fathers were heterozygous. For the SP-B-deficient infant, microsatellite markers confirmed maternal heterodisomy with segmental isodisomy. Microsatellite analysis confirmed paternal isodisomy for the 3 ABCA3-deficient infants. Two ABCA3-deficient infants underwent lung transplantation at 3 and 5 months of age, respectively, and 2 infants died. None exhibited any nonpulmonary phenotype. CONCLUSIONS Uniparental disomy should be suspected in infants with rare homozygous mutations in SFTPB or ABCA3. Confirmation of parental carrier status is important to provide recurrence risk and to monitor expression of other phenotypes that may emerge through reduction to homozygosity of recessive alleles.


BMC Medical Genetics | 2013

Rare recessive loss-of-function methionyl-tRNA synthetase mutations presenting as a multi-organ phenotype

Eline van Meel; Daniel J. Wegner; Paul F. Cliften; Marcia C. Willing; Frances V. White; Stuart Kornfeld; F. Sessions Cole

BackgroundMethionyl-tRNA synthetase (MARS) catalyzes the ligation of methionine to its cognate transfer RNA and therefore plays an essential role in protein biosynthesis.MethodsWe used exome sequencing, aminoacylation assays, homology modeling, and immuno-isolation of transfected MARS to identify and characterize mutations in the methionyl-tRNA synthetase gene (MARS) in an infant with an unexplained multi-organ phenotype.ResultsWe identified compound heterozygous mutations (F370L and I523T) in highly conserved regions of MARS. The parents were each heterozygous for one of the mutations. Aminoacylation assays documented that the F370L and I523T MARS mutants had 18 ± 6% and 16 ± 6%, respectively, of wild-type activity. Homology modeling of the human MARS sequence with the structure of E. coli MARS showed that the F370L and I523T mutations are in close proximity to each other, with residue I523 located in the methionine binding pocket. We found that the F370L and I523T mutations did not affect the association of MARS with the multisynthetase complex.ConclusionThis infant expands the catalogue of inherited human diseases caused by mutations in aminoacyl-tRNA synthetase genes.


Pediatric Research | 2010

Surfactant Protein-C Promoter Variants Associated With Neonatal Respiratory Distress Syndrome Reduce Transcription

Jennifer A. Wambach; Ping Yang; Daniel J. Wegner; Ping An; Brian P. Hackett; F. S. Cole; Aaron Hamvas

Dominant mutations in coding regions of the surfactant protein-C gene, SFTPC, cause respiratory distress syndrome (RDS) in infants. However, the contribution of variants in noncoding regions of SFTPC to pulmonary phenotypes is unknown. By using a case-control group of infants ≥34 weeks gestation (n = 538), we used complete resequencing of SFTPC and its promoter, genotyping, and logistic regression to identify 80 single nucleotide polymorphisms (SNPs). Three promoter SNPs were statistically associated with neonatal RDS among European descent infants. To assess the transcriptional effects of these three promoter SNPs, we selectively mutated the SFTPC promoter and performed transient transfection using MLE-15 cells and a firefly luciferase reporter vector. Each promoter SNP decreased SFTPC transcription. The combination of two variants in high linkage dysequilibrium also decreased SFTPC transcription. In silico evaluation of transcription factor binding demonstrated that the rare allele at g.−1167 disrupts a SOX (SRY-related high mobility group box) consensus motif and introduces a GATA-1 site, at g.−2385 removes a MZF-1 (myeloid zinc finger) binding site, and at g.−1647 removes a potential methylation site. This combined statistical, in vitro, and in silico approach suggests that reduced SFTPC transcription contributes to the genetic risk for neonatal RDS in developmentally susceptible infants.


BMJ Open Respiratory Research | 2014

Sequencing of idiopathic pulmonary fibrosis-related genes reveals independent single gene associations

Meghan A. Coghlan; Adrian Shifren; H.J. Huang; Tonya D. Russell; Robi D. Mitra; Qunyuan Zhang; Daniel J. Wegner; F. Sessions Cole; Aaron Hamvas

Background Previous studies investigating a genetic basis for idiopathic pulmonary fibrosis (IPF) have focused on resequencing single genes in IPF kindreds or cohorts to determine the genetic contributions to IPF. None has investigated interactions among the candidate genes. Objective To compare the frequencies and interactions of mutations in six IPF-associated genes in a cohort of 132 individuals with IPF with those of a disease-control cohort of 192 individuals with chronic obstructive pulmonary disease (COPD) and the population represented in the Exome Variant Server. Methods We resequenced the genes encoding surfactant proteins A2 (SFTPA2), and C (SFTPC), the ATP binding cassette member A3 (ABCA3), telomerase (TERT), thyroid transcription factor (NKX2-1) and mucin 5B (MUC5B) and compared the collapsed frequencies of rare (minor allele frequency <1%), computationally predicted deleterious variants in each cohort. We also genotyped a common MUC5B promoter variant that is over-represented in individuals with IPF. Results We found 15 mutations in 14 individuals (11%) in the IPF cohort: (SFTPA2 (n=1), SFTPC (n=5), ABCA3 (n=4) and TERT (n=5)). No individual with IPF had two different mutations, but one individual with IPF was homozygous for p.E292V, the most common ABCA3 disease-causing variant. We did not detect an interaction between any of the mutations and the MUC5B promoter variant. Conclusions Rare mutations in SFTPA2, SFTPC and TERT are collectively over-represented in individuals with IPF. Genetic analysis and counselling should be considered as part of the IPF evaluation.


Acta Paediatrica | 2007

A major deletion in the surfactant protein-B gene causing lethal respiratory distress

Daniel J. Wegner; T. Hertzberg; Hillary B. Heins; Göran Elmberger; Michael J. MacCoss; Christopher S. Carlson; Lawrence M. Nogee; F. Sessions Cole; Aaron Hamvas

Background: Loss of function mutations in the surfactant protein‐B gene (SFTPB) cause lethal neonatal respiratory distress due to reduced or absent expression of mature surfactant protein B (SP‐B, encoded in exons 6 and 7). No large deletions in SFTPB have been previously identified.


Pediatric Pulmonology | 2008

Recombination as a mechanism for sporadic mutation in the surfactant protein-C gene

Amy D. McBee; Daniel J. Wegner; Christopher S. Carlson; Jennifer A. Wambach; Ping Yang; Hillary B. Heins; Ola Didrik Saugstad; Michelle Trusgnich; Julie Watkins-Torry; Lawrence M. Nogee; Howard E. Henderson; F. Sessions Cole; Aaron Hamvas

To determine haplotype background of common mutations in the genes encoding surfactant proteins B and C (SFTPB and SFTPC) and to assess recombination in SFTPC.

Collaboration


Dive into the Daniel J. Wegner's collaboration.

Top Co-Authors

Avatar

Aaron Hamvas

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

F. Sessions Cole

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jennifer A. Wambach

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Hillary B. Heins

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Lawrence M. Nogee

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Michelle Trusgnich

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Kelcey DePass

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Ping An

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Robi D. Mitra

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Todd E. Druley

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge