Network


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

Hotspot


Dive into the research topics where Laura M. Yerges-Armstrong is active.

Publication


Featured researches published by Laura M. Yerges-Armstrong.


PLOS Medicine | 2013

Causal Relationship Between Obesity and Vitamin D Status: Bi-Directional Mendelian Randomization Analysis of Multiple Cohorts

Karani Santhanakrishnan Vimaleswaran; Diane J. Berry; Emmi Tikkanen; Stefan Pilz; Linda T. Hiraki; Jason D. Cooper; Zari Dastani; Denise K. Houston; Andrew R. Wood; Liesbeth Vandenput; Lina Zgaga; Laura M. Yerges-Armstrong; Mark I. McCarthy; Marika Kaakinen; Marcus E. Kleber; Kurt Lohman; Luigi Ferrucci; Liisa Byberg; Lars Lind; Mattias Lorentzon; Veikko Salomaa; Harry Campbell; Malcolm G. Dunlop; Braxton D. Mitchell; Karl-Heinz Herzig; Elizabeth A. Streeten; Evropi Theodoratou; Antti Jula; Nicholas J. Wareham; Claes Ohlsson

A mendelian randomization study based on data from multiple cohorts conducted by Karani Santhanakrishnan Vimaleswaran and colleagues re-examines the causal nature of the relationship between vitamin D levels and obesity.


PLOS Genetics | 2012

WNT16 influences bone mineral density, cortical bone thickness, bone strength, and osteoporotic fracture risk.

Hou-Feng Zheng; Jon H Tobias; Emma L. Duncan; David Evans; Joel Eriksson; Lavinia Paternoster; Laura M. Yerges-Armstrong; Terho Lehtimäki; Ulrica Bergström; Mika Kähönen; Paul Leo; Olli T. Raitakari; Marika Laaksonen; Geoffrey C. Nicholson; Jorma Viikari; Martin Ladouceur; Leo-Pekka Lyytikäinen; Carolina Medina-Gomez; Fernando Rivadeneira; Richard L. Prince; Harri Sievänen; William D. Leslie; Dan Mellström; John A. Eisman; Sofia Movérare-Skrtic; David Goltzman; David A. Hanley; Graeme Jones; Beate St Pourcain; Yongjun Xiao

We aimed to identify genetic variants associated with cortical bone thickness (CBT) and bone mineral density (BMD) by performing two separate genome-wide association study (GWAS) meta-analyses for CBT in 3 cohorts comprising 5,878 European subjects and for BMD in 5 cohorts comprising 5,672 individuals. We then assessed selected single-nucleotide polymorphisms (SNPs) for osteoporotic fracture in 2,023 cases and 3,740 controls. Association with CBT and forearm BMD was tested for ∼2.5 million SNPs in each cohort separately, and results were meta-analyzed using fixed effect meta-analysis. We identified a missense SNP (Thr>Ile; rs2707466) located in the WNT16 gene (7q31), associated with CBT (effect size of −0.11 standard deviations [SD] per C allele, P = 6.2×10−9). This SNP, as well as another nonsynonymous SNP rs2908004 (Gly>Arg), also had genome-wide significant association with forearm BMD (−0.14 SD per C allele, P = 2.3×10−12, and −0.16 SD per G allele, P = 1.2×10−15, respectively). Four genome-wide significant SNPs arising from BMD meta-analysis were tested for association with forearm fracture. SNP rs7776725 in FAM3C, a gene adjacent to WNT16, was associated with a genome-wide significant increased risk of forearm fracture (OR = 1.33, P = 7.3×10−9), with genome-wide suggestive signals from the two missense variants in WNT16 (rs2908004: OR = 1.22, P = 4.9×10−6 and rs2707466: OR = 1.22, P = 7.2×10−6). We next generated a homozygous mouse with targeted disruption of Wnt16. Female Wnt16−/− mice had 27% (P<0.001) thinner cortical bones at the femur midshaft, and bone strength measures were reduced between 43%–61% (6.5×10−13<P<5.9×10−4) at both femur and tibia, compared with their wild-type littermates. Natural variation in humans and targeted disruption in mice demonstrate that WNT16 is an important determinant of CBT, BMD, bone strength, and risk of fracture.


Diabetes | 2014

Genetic evidence for a normal-weight “metabolically obese” phenotype linking insulin resistance, hypertension, coronary artery disease and type 2 diabetes

Hanieh Yaghootkar; Robert A. Scott; Charles C. White; Weihua Zhang; Elizabeth K. Speliotes; Patricia B. Munroe; Georg B. Ehret; Joshua C. Bis; Caroline S. Fox; M. Walker; Ingrid B. Borecki; Joshua W. Knowles; Laura M. Yerges-Armstrong; Claes Ohlsson; John Perry; John Chambers; Jaspal S. Kooner; Nora Franceschini; Claudia Langenberg; Marie-France Hivert; Zari Dastani; J. Brent Richards; Robert K. Semple; Timothy M. Frayling

The mechanisms that predispose to hypertension, coronary artery disease (CAD), and type 2 diabetes (T2D) in individuals of normal weight are poorly understood. In contrast, in monogenic primary lipodystrophy—a reduction in subcutaneous adipose tissue—it is clear that it is adipose dysfunction that causes severe insulin resistance (IR), hypertension, CAD, and T2D. We aimed to test the hypothesis that common alleles associated with IR also influence the wider clinical and biochemical profile of monogenic IR. We selected 19 common genetic variants associated with fasting insulin–based measures of IR. We used hierarchical clustering and results from genome-wide association studies of eight nondisease outcomes of monogenic IR to group these variants. We analyzed genetic risk scores against disease outcomes, including 12,171 T2D cases, 40,365 CAD cases, and 69,828 individuals with blood pressure measurements. Hierarchical clustering identified 11 variants associated with a metabolic profile consistent with a common, subtle form of lipodystrophy. A genetic risk score consisting of these 11 IR risk alleles was associated with higher triglycerides (β = 0.018; P = 4 × 10−29), lower HDL cholesterol (β = −0.020; P = 7 × 10−37), greater hepatic steatosis (β = 0.021; P = 3 × 10−4), higher alanine transaminase (β = 0.002; P = 3 × 10−5), lower sex-hormone-binding globulin (β = −0.010; P = 9 × 10−13), and lower adiponectin (β = −0.015; P = 2 × 10−26). The same risk alleles were associated with lower BMI (per-allele β = −0.008; P = 7 × 10−8) and increased visceral-to-subcutaneous adipose tissue ratio (β = −0.015; P = 6 × 10−7). Individuals carrying ≥17 fasting insulin–raising alleles (5.5% population) were slimmer (0.30 kg/m2) but at increased risk of T2D (odds ratio [OR] 1.46; per-allele P = 5 × 10−13), CAD (OR 1.12; per-allele P = 1 × 10−5), and increased blood pressure (systolic and diastolic blood pressure of 1.21 mmHg [per-allele P = 2 × 10−5] and 0.67 mmHg [per-allele P = 2 × 10−4], respectively) compared with individuals carrying ≤9 risk alleles (5.5% population). Our results provide genetic evidence for a link between the three diseases of the “metabolic syndrome” and point to reduced subcutaneous adiposity as a central mechanism.


The New England Journal of Medicine | 2014

Null mutation in hormone-sensitive lipase gene and risk of type 2 diabetes.

Jessica S. Albert; Laura M. Yerges-Armstrong; Richard B. Horenstein; Toni I. Pollin; Urmila Sreenivasan; Sumbul Chai; William S. Blaner; Soren Snitker; Jeffrey R. O'Connell; Da Wei Gong; Richard J. Breyer; Alice S. Ryan; John C. McLenithan; Alan R. Shuldiner; Carole Sztalryd; Coleen M. Damcott

BACKGROUND Lipolysis regulates energy homeostasis through the hydrolysis of intracellular triglycerides and the release of fatty acids for use as energy substrates or lipid mediators in cellular processes. Genes encoding proteins that regulate energy homeostasis through lipolysis are thus likely to play an important role in determining susceptibility to metabolic disorders. METHODS We sequenced 12 lipolytic-pathway genes in Old Order Amish participants whose fasting serum triglyceride levels were at the extremes of the distribution and identified a novel 19-bp frameshift deletion in exon 9 of LIPE, encoding hormone-sensitive lipase (HSL), a key enzyme for lipolysis. We genotyped the deletion in DNA from 2738 Amish participants and performed association analyses to determine the effects of the deletion on metabolic traits. We also obtained biopsy specimens of abdominal subcutaneous adipose tissue from 2 study participants who were homozygous for the deletion (DD genotype), 10 who were heterozygous (ID genotype), and 7 who were noncarriers (II genotype) for assessment of adipose histologic characteristics, lipolysis, enzyme activity, cytokine release, and messenger RNA (mRNA) and protein levels. RESULTS Carriers of the mutation had dyslipidemia, hepatic steatosis, systemic insulin resistance, and diabetes. In adipose tissue from study participants with the DD genotype, the mutation resulted in the absence of HSL protein, small adipocytes, impaired lipolysis, insulin resistance, and inflammation. Transcription factors responsive to peroxisome-proliferator-activated receptor γ (PPAR-γ) and downstream target genes were down-regulated in adipose tissue from participants with the DD genotype, altering the regulation of pathways influencing adipogenesis, insulin sensitivity, and lipid metabolism. CONCLUSIONS These findings indicate the physiological significance of HSL in adipocyte function and the regulation of systemic lipid and glucose homeostasis and underscore the severe metabolic consequences of impaired lipolysis. (Funded by the National Institutes of Health and others).


Journal of Bone and Mineral Research | 2013

META-ANALYSIS OF GENOME-WIDE STUDIES IDENTIFIES WNT16 AND ESR1 SNPS ASSOCIATED WITH BONE MINERAL DENSITY IN PREMENOPAUSAL WOMEN **

Daniel L. Koller; Hou-Feng Zheng; David Karasik; Laura M. Yerges-Armstrong; Ching-Ti Liu; Fiona McGuigan; John P. Kemp; Sylvie Giroux; Dongbing Lai; Howard J. Edenberg; Munro Peacock; Stefan A. Czerwinski; Audrey C. Choh; George McMahon; Beate St Pourcain; Nicholas J. Timpson; Debbie A. Lawlor; David Evans; Bradford Towne; John Blangero; Melanie A. Carless; Candace M. Kammerer; David Goltzman; Christopher S. Kovacs; Jerilynn C. Prior; Tim D. Spector; François Rousseau; Jonathan H Tobias; Kristina Åkesson; Michael J. Econs

Previous genome‐wide association studies (GWAS) have identified common variants in genes associated with variation in bone mineral density (BMD), although most have been carried out in combined samples of older women and men. Meta‐analyses of these results have identified numerous single‐nucleotide polymorphisms (SNPs) of modest effect at genome‐wide significance levels in genes involved in both bone formation and resorption, as well as other pathways. We performed a meta‐analysis restricted to premenopausal white women from four cohorts (n = 4061 women, aged 20 to 45 years) to identify genes influencing peak bone mass at the lumbar spine and femoral neck. After imputation, age‐ and weight‐adjusted bone‐mineral density (BMD) values were tested for association with each SNP. Association of an SNP in the WNT16 gene (rs3801387; p = 1.7 × 10−9) and multiple SNPs in the ESR1/C6orf97 region (rs4870044; p = 1.3 × 10−8) achieved genome‐wide significance levels for lumbar spine BMD. These SNPs, along with others demonstrating suggestive evidence of association, were then tested for association in seven replication cohorts that included premenopausal women of European, Hispanic‐American, and African‐American descent (combined n = 5597 for femoral neck; n = 4744 for lumbar spine). When the data from the discovery and replication cohorts were analyzed jointly, the evidence was more significant (WNT16 joint p = 1.3 × 10−11; ESR1/C6orf97 joint p = 1.4 × 10−10). Multiple independent association signals were observed with spine BMD at the ESR1 region after conditioning on the primary signal. Analyses of femoral neck BMD also supported association with SNPs in WNT16 and ESR1/C6orf97 (p < 1 × 10−5). Our results confirm that several of the genes contributing to BMD variation across a broad age range in both sexes have effects of similar magnitude on BMD of the spine in premenopausal women. These data support the hypothesis that variants in these genes of known skeletal function also affect BMD during the premenopausal period.


Nature Communications | 2013

A rare functional cardioprotective APOC3 variant has risen in frequency in distinct population isolates.

Ioanna Tachmazidou; George V. Dedoussis; Lorraine Southam; Aliki-Eleni Farmaki; Graham R. S. Ritchie; Dionysia K. Xifara; Angela Matchan; Konstantinos Hatzikotoulas; N W Rayner; Yuning Chen; Toni I. Pollin; O'Connell; Laura M. Yerges-Armstrong; Chrysoula Kiagiadaki; Kalliope Panoutsopoulou; Jeremy Schwartzentruber; Loukas Moutsianas; Emmanouil Tsafantakis; Chris Tyler-Smith; Gilean McVean; Yali Xue; Eleftheria Zeggini

Isolated populations can empower the identification of rare variation associated with complex traits through next generation association studies, but the generalizability of such findings remains unknown. Here we genotype 1,267 individuals from a Greek population isolate on the Illumina HumanExome Beadchip, in search of functional coding variants associated with lipids traits. We find genome-wide significant evidence for association between R19X, a functional variant in APOC3, with increased high-density lipoprotein and decreased triglycerides levels. Approximately 3.8% of individuals are heterozygous for this cardioprotective variant, which was previously thought to be private to the Amish founder population. R19X is rare (<0.05% frequency) in outbred European populations. The increased frequency of R19X enables discovery of this lipid traits signal at genome-wide significance in a small sample size. This work exemplifies the value of isolated populations in successfully detecting transferable rare variant associations of high medical relevance.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2010

Association of the Vitamin D Metabolism Gene CYP24A1 With Coronary Artery Calcification

Haiqing Shen; Lawrence F. Bielak; Jane F. Ferguson; Elizabeth A. Streeten; Laura M. Yerges-Armstrong; Jie Liu; Wendy S. Post; Jeffery R. O'Connell; James E. Hixson; Sharon L.R. Kardia; Yan V. Sun; Mina A. Jhun; Xuexia Wang; Nehal N. Mehta; Mingyao Li; Daniel L. Koller; Hakan Hakonarson; Brendan J. Keating; Daniel J. Rader; Alan R. Shuldiner; Patricia A. Peyser; Muredach P. Reilly; Braxton D. Mitchell

Objective—The vitamin D endocrine system is essential for calcium homeostasis, and low levels of vitamin D metabolites have been associated with cardiovascular disease risk. We hypothesized that DNA sequence variation in genes regulating vitamin D metabolism and signaling pathways might influence variation in coronary artery calcification (CAC). Methods and Results—We genotyped single-nucleotide polymorphisms (SNPs) in GC, CYP27B1, CYP24A1, and VDR and tested their association with CAC quantity, as measured by electron beam computed tomography. Initial association studies were carried out in a discovery sample comprising 697 Amish subjects, and SNPs nominally associated with CAC quantity (4 SNPs in CYP24A1, P=0.008 to 0.00003) were then tested for association with CAC quantity in 2 independent cohorts of subjects of white European ancestry (Genetic Epidemiology Network of Arteriopathy study [n=916] and the Penn Coronary Artery Calcification sample [n=2061]). One of the 4 SNPs, rs2762939, was associated with CAC quantity in both the Genetic Epidemiology Network of Arteriopathy (P=0.007) and Penn Coronary Artery Calcification (P=0.01) studies. In all 3 populations, the rs2762939 C allele was associated with lower CAC quantity. Metaanalysis for the association of this SNP with CAC quantity across all 3 studies yielded a P value of 2.9×10−6. Conclusion—A common SNP in the CYP24A1 gene was associated with CAC quantity in 3 independent populations. This result suggests a role for vitamin D metabolism in the development of CAC quantity.


Human Heredity | 2013

Genetic Variation at NCAN Locus Is Associated with Inflammation and Fibrosis in Non-Alcoholic Fatty Liver Disease in Morbid Obesity

Alexis Gorden; Rongze Yang; Laura M. Yerges-Armstrong; Kathy A. Ryan; Elizabeth K. Speliotes; Ingrid B. Borecki; Tamara B. Harris; Xin Chu; G.C. Wood; C.D. Still; Alan R. Shuldiner; Glenn S. Gerhard

Objective: Obesity-associated non-alcoholic fatty liver disease (NAFLD) may cause liver dysfunction and failure. In a previously reported genome-wide association meta-analysis, single nucleotide polymorphisms (SNPs) near PNPLA3, NCAN, GCKR, LYPLAL1 and PPP1R3B were associated with NAFLD and with distinctive serum lipid profiles. The present study examined the relevance of these variants to NAFLD in extreme obesity. Methods: In 1,092 bariatric surgery patients, the candidate SNPs were genotyped and association analyses with liver histology and serum lipids were performed. Results: We replicated the association of hepatosteatosis with PNPLA3 rs738409[G] and with NCAN rs2228603[T]. We also replicated the association of rs2228603[T] with hepatic inflammation and fibrosis. rs2228603[T] was associated with lower serum low-density lipoprotein, total cholesterol and triglycerides. After stratification by the presence or absence of NAFLD, these associations were present predominantly in the subgroup with NAFLD. Conclusion:NCAN rs2228603[T] is a risk factor for liver inflammation and fibrosis, suggesting that this locus is responsible for progression from steatosis to steatohepatitis. In this bariatric cohort, rs2228603[T] was associated with low serum lipids only in patients with NAFLD. This supports a NAFLD model in which the liver may sequester triglycerides as a result of either increased triglyceride uptake and/or decreased lipolysis.


Hepatology | 2013

Characterization of european ancestry nonalcoholic fatty liver disease-associated variants in individuals of african and hispanic descent

Nicholette D. Palmer; Solomon K. Musani; Laura M. Yerges-Armstrong; Mary F. Feitosa; Lawrence F. Bielak; Ruben Hernaez; Bratati Kahali; J. Jeffrey Carr; Tamara B. Harris; Min A. Jhun; Sharon L.R. Kardia; Carl D. Langefeld; Thomas H. Mosley; Jill M. Norris; Albert V. Smith; Herman A. Taylor; Lynne E. Wagenknecht; Jiankang Liu; Ingrid B. Borecki; Patricia A. Peyser; Elizabeth K. Speliotes

Nonalcoholic fatty liver disease (NAFLD) is an obesity‐related condition affecting over 50% of individuals in some populations and is expected to become the number one cause of liver disease worldwide by 2020. Common, robustly associated genetic variants in/near five genes were identified for hepatic steatosis, a quantifiable component of NAFLD, in European ancestry individuals. Here we tested whether these variants were associated with hepatic steatosis in African‐ and/or Hispanic‐Americans and fine‐mapped the observed association signals. We measured hepatic steatosis using computed tomography in five African American (n = 3,124) and one Hispanic American (n = 849) cohorts. All analyses controlled for variation in age, age2, gender, alcoholic drinks, and population substructure. Heritability of hepatic steatosis was estimated in three cohorts. Variants in/near PNPLA3, NCAN, LYPLAL1, GCKR, and PPP1R3B were tested for association with hepatic steatosis using a regression framework in each cohort and meta‐analyzed. Fine‐mapping across African American cohorts was conducted using meta‐analysis. African‐ and Hispanic‐American cohorts were 33.9/37.5% male, with average age of 58.6/42.6 years and body mass index of 31.8/28.9 kg/m2, respectively. Hepatic steatosis was 0.20‐0.34 heritable in African‐ and Hispanic‐American families (P < 0.02 in each cohort). Variants in or near PNPLA3, NCAN, GCKR, PPP1R3B in African Americans and PNPLA3 and PPP1R3B in Hispanic Americans were significantly associated with hepatic steatosis; however, allele frequency and effect size varied across ancestries. Fine‐mapping in African Americans highlighted missense variants at PNPLA3 and GCKR and redefined the association region at LYPLAL1. Conclusion: Multiple genetic variants are associated with hepatic steatosis across ancestries. This explains a substantial proportion of the genetic predisposition in African‐ and Hispanic‐Americans. Missense variants in PNPLA3 and GCKR are likely functional across multiple ancestries. (Hepatology 2013;53:966–975)


Clinical Pharmacology & Therapeutics | 2013

Purine Pathway Implicated in Mechanism of Resistance to Aspirin Therapy: Pharmacometabolomics-Informed Pharmacogenomics

Laura M. Yerges-Armstrong; Sandrine Ellero-Simatos; Anastasia Georgiades; Hongjie Zhu; Joshua P. Lewis; Richard B. Horenstein; Amber L. Beitelshees; Adrie Dane; Theo H. Reijmers; Thomas Hankemeier; Oliver Fiehn; Alan R. Shuldiner; Rima Kaddurah-Daouk

Although aspirin is a well‐established antiplatelet agent, the mechanisms of aspirin resistance remain poorly understood. Metabolomics allows for measurement of hundreds of small molecules in biological samples, enabling detailed mapping of pathways involved in drug response. We defined the metabolic signature of aspirin exposure in subjects from the Heredity and Phenotype Intervention Heart Study. Many metabolites, including known aspirin catabolites, changed on exposure to aspirin, and pathway enrichment analysis identified purine metabolism as significantly affected by drug exposure. Furthermore, purines were associated with aspirin response, and poor responders had higher postaspirin adenosine and inosine levels than did good responders (n = 76; both P < 4 × 10−3). Using our established “pharmacometabolomics‐informed pharmacogenomics” approach, we identified genetic variants in adenosine kinase associated with aspirin response. Combining metabolomics and genomics allowed for more comprehensive interrogation of mechanisms of variation in aspirin response—an important step toward personalized treatment approaches for cardiovascular disease.

Collaboration


Dive into the Laura M. Yerges-Armstrong's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ingrid B. Borecki

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Tamara B. Harris

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mary F. Feitosa

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge