566 results on '"John B Harley"'
Search Results
2. 701 A million veteran program (MVP) genome-wide association study (GWAS) of systemic and cutaneous lupus erythematosus (SLE & CLE)
- Author
-
John B Harley, Viktoryia Laurynenka, Kenneth M Kaufman, Celi Sun, R Hal Scofield, Dennis H Clark, Iouri Chepelev, and Isaac TW Harley
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2024
- Full Text
- View/download PDF
3. 702 Comparative genome wide association studies (GWASs) of systemic lupus erythematosus (SLE) and multiple sclerosis (MS) from the million veteran program (MVP)
- Author
-
John B Harley, Viktoryia Laurynenka, Kenneth M Kaufman, Celi Sun, R Hal Scofield, Dennis H Clark, Iouri Chepelev, and Isaac TW Harley
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2024
- Full Text
- View/download PDF
4. LP-114 Lupus and multiple sclerosis may share an initiating pathogenesis: the anti- EBNA1 heuristic conjecture
- Author
-
John B Harley, Viktoryia Laurynenka, Kenneth M Kaufman, Dennis H Clark, Iouri Chepelev, and Isaac TW Harley
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2023
- Full Text
- View/download PDF
5. 909 Haplotype-specific chromatin looping reveals genetic interactions of regulatory regions modulating gene expression at the SLE susceptibility locus FAM167A-BLK
- Author
-
John B Harley, Bahram Namjou, Iouri Chepelev, Mariana Saint-Just Ribeiro, and Pulak Tripathi
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2022
- Full Text
- View/download PDF
6. 101 Anti-EBNA1 molecular mimicry initiates cross-reacting autoantibodies in Systemic Lupus Erythematosus (SLE) & Multiple Sclerosis (MS)
- Author
-
John B Harley, Dennis H Clark, and Iouri Chepelev
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2022
- Full Text
- View/download PDF
7. 1502 Genetic predisposition to lupus across ancestries has >300 separable genetic contributions: what we know today
- Author
-
John B Harley, Viktoryia Laurynenka, Leah C Kottyan, Matthew T Weirauch, and Kenneth M Kaufman
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2021
- Full Text
- View/download PDF
8. Novel EDGE encoding method enhances ability to identify genetic interactions.
- Author
-
Molly A Hall, John Wallace, Anastasia M Lucas, Yuki Bradford, Shefali S Verma, Bertram Müller-Myhsok, Kristin Passero, Jiayan Zhou, John McGuigan, Beibei Jiang, Sarah A Pendergrass, Yanfei Zhang, Peggy Peissig, Murray Brilliant, Patrick Sleiman, Hakon Hakonarson, John B Harley, Krzysztof Kiryluk, Kristel Van Steen, Jason H Moore, and Marylyn D Ritchie
- Subjects
Genetics ,QH426-470 - Abstract
Assumptions are made about the genetic model of single nucleotide polymorphisms (SNPs) when choosing a traditional genetic encoding: additive, dominant, and recessive. Furthermore, SNPs across the genome are unlikely to demonstrate identical genetic models. However, running SNP-SNP interaction analyses with every combination of encodings raises the multiple testing burden. Here, we present a novel and flexible encoding for genetic interactions, the elastic data-driven genetic encoding (EDGE), in which SNPs are assigned a heterozygous value based on the genetic model they demonstrate in a dataset prior to interaction testing. We assessed the power of EDGE to detect genetic interactions using 29 combinations of simulated genetic models and found it outperformed the traditional encoding methods across 10%, 30%, and 50% minor allele frequencies (MAFs). Further, EDGE maintained a low false-positive rate, while additive and dominant encodings demonstrated inflation. We evaluated EDGE and the traditional encodings with genetic data from the Electronic Medical Records and Genomics (eMERGE) Network for five phenotypes: age-related macular degeneration (AMD), age-related cataract, glaucoma, type 2 diabetes (T2D), and resistant hypertension. A multi-encoding genome-wide association study (GWAS) for each phenotype was performed using the traditional encodings, and the top results of the multi-encoding GWAS were considered for SNP-SNP interaction using the traditional encodings and EDGE. EDGE identified a novel SNP-SNP interaction for age-related cataract that no other method identified: rs7787286 (MAF: 0.041; intergenic region of chromosome 7)-rs4695885 (MAF: 0.34; intergenic region of chromosome 4) with a Bonferroni LRT p of 0.018. A SNP-SNP interaction was found in data from the UK Biobank within 25 kb of these SNPs using the recessive encoding: rs60374751 (MAF: 0.030) and rs6843594 (MAF: 0.34) (Bonferroni LRT p: 0.026). We recommend using EDGE to flexibly detect interactions between SNPs exhibiting diverse action.
- Published
- 2021
- Full Text
- View/download PDF
9. Amino acid signatures of HLA Class-I and II molecules are strongly associated with SLE susceptibility and autoantibody production in Eastern Asians.
- Author
-
Julio E Molineros, Loren L Looger, Kwangwoo Kim, Yukinori Okada, Chikashi Terao, Celi Sun, Xu-Jie Zhou, Prithvi Raj, Yuta Kochi, Akari Suzuki, Shuji Akizuki, Shuichiro Nakabo, So-Young Bang, Hye-Soon Lee, Young Mo Kang, Chang-Hee Suh, Won Tae Chung, Yong-Beom Park, Jung-Yoon Choe, Seung-Cheol Shim, Shin-Seok Lee, Xiaoxia Zuo, Kazuhiko Yamamoto, Quan-Zhen Li, Nan Shen, Lauren L Porter, John B Harley, Kek Heng Chua, Hong Zhang, Edward K Wakeland, Betty P Tsao, Sang-Cheol Bae, and Swapan K Nath
- Subjects
Genetics ,QH426-470 - Abstract
Human leukocyte antigen (HLA) is a key genetic factor conferring risk of systemic lupus erythematosus (SLE), but precise independent localization of HLA effects is extremely challenging. As a result, the contribution of specific HLA alleles and amino-acid residues to the overall risk of SLE and to risk of specific autoantibodies are far from completely understood. Here, we dissected (a) overall SLE association signals across HLA, (b) HLA-peptide interaction, and (c) residue-autoantibody association. Classical alleles, SNPs, and amino-acid residues of eight HLA genes were imputed across 4,915 SLE cases and 13,513 controls from Eastern Asia. We performed association followed by conditional analysis across HLA, assessing both overall SLE risk and risk of autoantibody production. DR15 alleles HLA-DRB1*15:01 (P = 1.4x10-27, odds ratio (OR) = 1.57) and HLA-DQB1*06:02 (P = 7.4x10-23, OR = 1.55) formed the most significant haplotype (OR = 2.33). Conditioned protein-residue signals were stronger than allele signals and mapped predominantly to HLA-DRB1 residue 13 (P = 2.2x10-75) and its proxy position 11 (P = 1.1x10-67), followed by HLA-DRB1-37 (P = 4.5x10-24). After conditioning on HLA-DRB1, novel associations at HLA-A-70 (P = 1.4x10-8), HLA-DPB1-35 (P = 9.0x10-16), HLA-DQB1-37 (P = 2.7x10-14), and HLA-B-9 (P = 6.5x10-15) emerged. Together, these seven residues increased the proportion of explained heritability due to HLA to 2.6%. Risk residues for both overall disease and hallmark autoantibodies (i.e., nRNP: DRB1-11, P = 2.0x10-14; DRB1-13, P = 2.9x10-13; DRB1-30, P = 3.9x10-14) localized to the peptide-binding groove of HLA-DRB1. Enrichment for specific amino-acid characteristics in the peptide-binding groove correlated with overall SLE risk and with autoantibody presence. Risk residues were in primarily negatively charged side-chains, in contrast with rheumatoid arthritis. We identified novel SLE signals in HLA Class I loci (HLA-A, HLA-B), and localized primary Class II signals to five residues in HLA-DRB1, HLA-DPB1, and HLA-DQB1. These findings provide insights about the mechanisms by which the risk residues interact with each other to produce autoantibodies and are involved in SLE pathophysiology.
- Published
- 2019
- Full Text
- View/download PDF
10. Identification of a Sjögren's syndrome susceptibility locus at OAS1 that influences isoform switching, protein expression, and responsiveness to type I interferons.
- Author
-
He Li, Tove Ragna Reksten, John A Ice, Jennifer A Kelly, Indra Adrianto, Astrid Rasmussen, Shaofeng Wang, Bo He, Kiely M Grundahl, Stuart B Glenn, Corinne Miceli-Richard, Simon Bowman, Sue Lester, Per Eriksson, Maija-Leena Eloranta, Johan G Brun, Lasse G Gøransson, Erna Harboe, Joel M Guthridge, Kenneth M Kaufman, Marika Kvarnström, Deborah S Cunninghame Graham, Ketan Patel, Adam J Adler, A Darise Farris, Michael T Brennan, James Chodosh, Rajaram Gopalakrishnan, Michael H Weisman, Swamy Venuturupalli, Daniel J Wallace, Kimberly S Hefner, Glen D Houston, Andrew J W Huang, Pamela J Hughes, David M Lewis, Lida Radfar, Evan S Vista, Contessa E Edgar, Michael D Rohrer, Donald U Stone, Timothy J Vyse, John B Harley, Patrick M Gaffney, Judith A James, Sean Turner, Ilias Alevizos, Juan-Manuel Anaya, Nelson L Rhodus, Barbara M Segal, Courtney G Montgomery, R Hal Scofield, Susan Kovats, Xavier Mariette, Lars Rönnblom, Torsten Witte, Maureen Rischmueller, Marie Wahren-Herlenius, Roald Omdal, Roland Jonsson, Wan-Fai Ng, for UK Primary Sjögren's Syndrome Registry, Gunnel Nordmark, Christopher J Lessard, and Kathy L Sivils
- Subjects
Genetics ,QH426-470 - Abstract
Sjögren's syndrome (SS) is a common, autoimmune exocrinopathy distinguished by keratoconjunctivitis sicca and xerostomia. Patients frequently develop serious complications including lymphoma, pulmonary dysfunction, neuropathy, vasculitis, and debilitating fatigue. Dysregulation of type I interferon (IFN) pathway is a prominent feature of SS and is correlated with increased autoantibody titers and disease severity. To identify genetic determinants of IFN pathway dysregulation in SS, we performed cis-expression quantitative trait locus (eQTL) analyses focusing on differentially expressed type I IFN-inducible transcripts identified through a transcriptome profiling study. Multiple cis-eQTLs were associated with transcript levels of 2'-5'-oligoadenylate synthetase 1 (OAS1) peaking at rs10774671 (PeQTL = 6.05 × 10-14). Association of rs10774671 with SS susceptibility was identified and confirmed through meta-analysis of two independent cohorts (Pmeta = 2.59 × 10-9; odds ratio = 0.75; 95% confidence interval = 0.66-0.86). The risk allele of rs10774671 shifts splicing of OAS1 from production of the p46 isoform to multiple alternative transcripts, including p42, p48, and p44. We found that the isoforms were differentially expressed within each genotype in controls and patients with and without autoantibodies. Furthermore, our results showed that the three alternatively spliced isoforms lacked translational response to type I IFN stimulation. The p48 and p44 isoforms also had impaired protein expression governed by the 3' end of the transcripts. The SS risk allele of rs10774671 has been shown by others to be associated with reduced OAS1 enzymatic activity and ability to clear viral infections, as well as reduced responsiveness to IFN treatment. Our results establish OAS1 as a risk locus for SS and support a potential role for defective viral clearance due to altered IFN response as a genetic pathophysiological basis of this complex autoimmune disease.
- Published
- 2017
- Full Text
- View/download PDF
11. Characterization and classification of lupus patients based on plasma thermograms.
- Author
-
Nichola C Garbett, Guy N Brock, Jonathan B Chaires, Chongkham S Mekmaysy, Lynn DeLeeuw, Kathy L Sivils, John B Harley, Brad H Rovin, K B Kulasekera, and Wael N Jarjour
- Subjects
Medicine ,Science - Abstract
OBJECTIVE:Plasma thermograms (thermal stability profiles of blood plasma) are being utilized as a new diagnostic approach for clinical assessment. In this study, we investigated the ability of plasma thermograms to classify systemic lupus erythematosus (SLE) patients versus non SLE controls using a sample of 300 SLE and 300 control subjects from the Lupus Family Registry and Repository. Additionally, we evaluated the heterogeneity of thermograms along age, sex, ethnicity, concurrent health conditions and SLE diagnostic criteria. METHODS:Thermograms were visualized graphically for important differences between covariates and summarized using various measures. A modified linear discriminant analysis was used to segregate SLE versus control subjects on the basis of the thermograms. Classification accuracy was measured based on multiple training/test splits of the data and compared to classification based on SLE serological markers. RESULTS:Median sensitivity, specificity, and overall accuracy based on classification using plasma thermograms was 86%, 83%, and 84% compared to 78%, 95%, and 86% based on a combination of five antibody tests. Combining thermogram and serology information together improved sensitivity from 78% to 86% and overall accuracy from 86% to 89% relative to serology alone. Predictive accuracy of thermograms for distinguishing SLE and osteoarthritis / rheumatoid arthritis patients was comparable. Both gender and anemia significantly interacted with disease status for plasma thermograms (p
- Published
- 2017
- Full Text
- View/download PDF
12. Regulatory polymorphisms modulate the expression of HLA class II molecules and promote autoimmunity
- Author
-
Prithvi Raj, Ekta Rai, Ran Song, Shaheen Khan, Benjamin E Wakeland, Kasthuribai Viswanathan, Carlos Arana, Chaoying Liang, Bo Zhang, Igor Dozmorov, Ferdicia Carr-Johnson, Mitja Mitrovic, Graham B Wiley, Jennifer A Kelly, Bernard R Lauwerys, Nancy J Olsen, Chris Cotsapas, Christine K Garcia, Carol A Wise, John B Harley, Swapan K Nath, Judith A James, Chaim O Jacob, Betty P Tsao, Chandrashekhar Pasare, David R Karp, Quan Zhen Li, Patrick M Gaffney, and Edward K Wakeland
- Subjects
targeted sequencing ,HLA ,SLE risk ,haplotype ,risk allele ,LD ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Targeted sequencing of sixteen SLE risk loci among 1349 Caucasian cases and controls produced a comprehensive dataset of the variations causing susceptibility to systemic lupus erythematosus (SLE). Two independent disease association signals in the HLA-D region identified two regulatory regions containing 3562 polymorphisms that modified thirty-seven transcription factor binding sites. These extensive functional variations are a new and potent facet of HLA polymorphism. Variations modifying the consensus binding motifs of IRF4 and CTCF in the XL9 regulatory complex modified the transcription of HLA-DRB1, HLA-DQA1 and HLA-DQB1 in a chromosome-specific manner, resulting in a 2.5-fold increase in the surface expression of HLA-DR and DQ molecules on dendritic cells with SLE risk genotypes, which increases to over 4-fold after stimulation. Similar analyses of fifteen other SLE risk loci identified 1206 functional variants tightly linked with disease-associated SNPs and demonstrated that common disease alleles contain multiple causal variants modulating multiple immune system genes.
- Published
- 2016
- Full Text
- View/download PDF
13. A GWAS Study on Liver Function Test Using eMERGE Network Participants.
- Author
-
Bahram Namjou, Keith Marsolo, Todd Lingren, Marylyn D Ritchie, Shefali S Verma, Beth L Cobb, Cassandra Perry, Terrie E Kitchner, Murray H Brilliant, Peggy L Peissig, Kenneth M Borthwick, Marc S Williams, Jane Grafton, Gail P Jarvik, Ingrid A Holm, and John B Harley
- Subjects
Medicine ,Science - Abstract
Liver enzyme levels and total serum bilirubin are under genetic control and in recent years genome-wide population-based association studies have identified different susceptibility loci for these traits. We conducted a genome-wide association study in European ancestry participants from the Electronic Medical Records and Genomics (eMERGE) Network dataset of patient medical records with available genotyping data in order to identify genetic contributors to variability in serum bilirubin levels and other liver function tests and to compare the effects between adult and pediatric populations.The process of whole genome imputation of eMERGE samples with standard quality control measures have been described previously. After removing missing data and outliers based on principal components (PC) analyses, 3294 samples from European ancestry were used for the GWAS study. The association between each single nucleotide polymorphism (SNP) and total serum bilirubin and other liver function tests was tested using linear regression, adjusting for age, gender, site, platform and ancestry principal components (PC).Consistent with previous results, a strong association signal has been detected for UGT1A gene cluster (best SNP rs887829, beta = 0.15, p = 1.30x10-118) for total serum bilirubin level. Indeed, in this region more than 176 SNPs (or indels) had p
- Published
- 2015
- Full Text
- View/download PDF
14. The effect of inversion at 8p23 on BLK association with lupus in Caucasian population.
- Author
-
Bahram Namjou, Yizhao Ni, Isaac T W Harley, Iouri Chepelev, Beth Cobb, Leah C Kottyan, Patrick M Gaffney, Joel M Guthridge, Kenneth Kaufman, and John B Harley
- Subjects
Medicine ,Science - Abstract
UNLABELLED:To explore the potential influence of the polymorphic 8p23.1 inversion on known autoimmune susceptibility risk at or near BLK locus, we validated a new bioinformatics method that utilizes SNP data to enable accurate, high-throughput genotyping of the 8p23.1 inversion in a Caucasian population. METHODS:Principal components analysis (PCA) was performed using markers inside the inversion territory followed by k-means cluster analyses on 7416 European derived and 267 HapMaP CEU and TSI samples. A logistic regression conditional analysis was performed. RESULTS:Three subgroups have been identified; inversion homozygous, heterozygous and non-inversion homozygous. The status of inversion was further validated using HapMap samples that had previously undergone Fluorescence in situ hybridization (FISH) assays with a concordance rate of above 98%. Conditional analyses based on the status of inversion were performed. We found that overall association signals in the BLK region remain significant after controlling for inversion status. The proportion of lupus cases and controls (cases/controls) in each subgroup was determined to be 0.97 for the inverted homozygous group (1067 cases and 1095 controls), 1.12 for the inverted heterozygous group (1935 cases 1717 controls) and 1.36 for non-inverted subgroups (924 cases and 678 controls). After calculating the linkage disequilibrium between inversion status and lupus risk haplotype we found that the lupus risk haplotype tends to reside on non-inversion background. As a result, a new association effect between non-inversion status and lupus phenotype has been identified ((p = 8.18×10(-7), OR = 1.18, 95%CI = 1.10-1.26). CONCLUSION:Our results demonstrate that both known lupus risk haplotype and inversion status act additively in the pathogenesis of lupus. Since inversion regulates expression of many genes in its territory, altered expression of other genes might also be involved in the development of lupus.
- Published
- 2014
- Full Text
- View/download PDF
15. EMR-linked GWAS study: Investigation of variation landscape of loci for Body Mass Index in children
- Author
-
Bahram eNamjou, Mehdi eKeddache, keith emarsolo, michael ewagner, todd elingren, Beth eCobb, cassandra eperry, stephanie ekennebeck, Ingrid A Holm, Rongling eLi, Nancy A Crimmins, Lisa eMartin, Imre eSolti, Isaac eKohane, and John B Harley
- Subjects
Genetics ,Obesity ,Pediatrics ,GWAS ,BMI ,FTO ,QH426-470 - Abstract
Common variation at the loci harboring the fat mass and obesity gene (FTO), MC4R and TMEM18 are consistently reported as being associated with obesity and body mass index especially in adult population. In order to confirm this effect in pediatric population five European ancestry cohorts from pediatric eMERGE-II network (CCHMC-BCH) were evaluated.Method:Data on 5049 samples of European ancestry were obtained from the Electronic Medical Records (EMRs) of two large academic centers in five different genotyped cohorts. For all available samples, gender, age, height and weight were collected and Body Mass Index (BMI) was calculated. To account for age and sex differences in BMI, BMI z-scores were generated using 2000 Centers of Disease Control and Prevention (CDC) growth charts. A Genome-wide association study (GWAS) was performed with BMI z-score. After removing missing data and outliers based on principal components (PC) analyses, 2860 samples were used for the GWAS study. The association between each SNP and BMI was tested using linear regression adjusting for age, gender, and PC by cohort. The effects of SNPs were modeled assuming additive, recessive and dominant effects of the minor allele. Meta-analysis was conducted using a weighted z-score approach. Results:The mean age of subjects was 9.8 years (range 2-19). The proportion of male subjects was 56%. In these cohorts, 14% of samples had a BMI≥95% and 28%≥85%. Meta analyses produced a signal at 16q12 genomic region with the best result of p=1.43x10E-07 (p (rec)=7.34E-08) for the single nucleotide polymorphism (SNP) rs8050136 at the first intron of FTO gene (z=5.26) and with no heterogeneity between cohorts (p=0.77). Imputation in this region using dense 1000-Genome and Hapmap CEU samples revealed 71 SNPs with p
- Published
- 2013
- Full Text
- View/download PDF
16. PTPN22 association in systemic lupus erythematosus (SLE) with respect to individual ancestry and clinical sub-phenotypes.
- Author
-
Bahram Namjou, Xana Kim-Howard, Celi Sun, Adam Adler, Sharon A Chung, Kenneth M Kaufman, Jennifer A Kelly, Stuart B Glenn, Joel M Guthridge, Robert H Scofield, Robert P Kimberly, Elizabeth E Brown, Graciela S Alarcón, Jeffrey C Edberg, Jae-Hoon Kim, Jiyoung Choi, Rosalind Ramsey-Goldman, Michelle A Petri, John D Reveille, Luis M Vilá, Susan A Boackle, Barry I Freedman, Betty P Tsao, Carl D Langefeld, Timothy J Vyse, Chaim O Jacob, Bernardo Pons-Estel, Argentine Collaborative Group, Timothy B Niewold, Kathy L Moser Sivils, Joan T Merrill, Juan-Manuel Anaya, Gary S Gilkeson, Patrick M Gaffney, Sang-Cheol Bae, Marta E Alarcón-Riquelme, BIOLUPUS and GENLES Networks, John B Harley, Lindsey A Criswell, Judith A James, and Swapan K Nath
- Subjects
Medicine ,Science - Abstract
Protein tyrosine phosphatase non-receptor type 22 (PTPN22) is a negative regulator of T-cell activation associated with several autoimmune diseases, including systemic lupus erythematosus (SLE). Missense rs2476601 is associated with SLE in individuals with European ancestry. Since the rs2476601 risk allele frequency differs dramatically across ethnicities, we assessed robustness of PTPN22 association with SLE and its clinical sub-phenotypes across four ethnically diverse populations. Ten SNPs were genotyped in 8220 SLE cases and 7369 controls from in European-Americans (EA), African-Americans (AA), Asians (AS), and Hispanics (HS). We performed imputation-based association followed by conditional analysis to identify independent associations. Significantly associated SNPs were tested for association with SLE clinical sub-phenotypes, including autoantibody profiles. Multiple testing was accounted for by using false discovery rate. We successfully imputed and tested allelic association for 107 SNPs within the PTPN22 region and detected evidence of ethnic-specific associations from EA and HS. In EA, the strongest association was at rs2476601 (P = 4.7 × 10(-9), OR = 1.40 (95% CI = 1.25-1.56)). Independent association with rs1217414 was also observed in EA, and both SNPs are correlated with increased European ancestry. For HS imputed intronic SNP, rs3765598, predicted to be a cis-eQTL, was associated (P = 0.007, OR = 0.79 and 95% CI = 0.67-0.94). No significant associations were observed in AA or AS. Case-only analysis using lupus-related clinical criteria revealed differences between EA SLE patients positive for moderate to high titers of IgG anti-cardiolipin (aCL IgG >20) versus negative aCL IgG at rs2476601 (P = 0.012, OR = 1.65). Association was reinforced when these cases were compared to controls (P = 2.7 × 10(-5), OR = 2.11). Our results validate that rs2476601 is the most significantly associated SNP in individuals with European ancestry. Additionally, rs1217414 and rs3765598 may be associated with SLE. Further studies are required to confirm the involvement of rs2476601 with aCL IgG.
- Published
- 2013
- Full Text
- View/download PDF
17. Admixture mapping in lupus identifies multiple functional variants within IFIH1 associated with apoptosis, inflammation, and autoantibody production.
- Author
-
Julio E Molineros, Amit K Maiti, Celi Sun, Loren L Looger, Shizhong Han, Xana Kim-Howard, Stuart Glenn, Adam Adler, Jennifer A Kelly, Timothy B Niewold, Gary S Gilkeson, Elizabeth E Brown, Graciela S Alarcón, Jeffrey C Edberg, Michelle Petri, Rosalind Ramsey-Goldman, John D Reveille, Luis M Vilá, Barry I Freedman, Betty P Tsao, Lindsey A Criswell, Chaim O Jacob, Jason H Moore, Timothy J Vyse, Carl L Langefeld, Joel M Guthridge, Patrick M Gaffney, Kathy L Moser, R Hal Scofield, Marta E Alarcón-Riquelme, BIOLUPUS Network, Scott M Williams, Joan T Merrill, Judith A James, Kenneth M Kaufman, Robert P Kimberly, John B Harley, and Swapan K Nath
- Subjects
Genetics ,QH426-470 - Abstract
Systemic lupus erythematosus (SLE) is an inflammatory autoimmune disease with a strong genetic component. African-Americans (AA) are at increased risk of SLE, but the genetic basis of this risk is largely unknown. To identify causal variants in SLE loci in AA, we performed admixture mapping followed by fine mapping in AA and European-Americans (EA). Through genome-wide admixture mapping in AA, we identified a strong SLE susceptibility locus at 2q22-24 (LOD=6.28), and the admixture signal is associated with the European ancestry (ancestry risk ratio ~1.5). Large-scale genotypic analysis on 19,726 individuals of African and European ancestry revealed three independently associated variants in the IFIH1 gene: an intronic variant, rs13023380 [P(meta) = 5.20×10(-14); odds ratio, 95% confidence interval = 0.82 (0.78-0.87)], and two missense variants, rs1990760 (Ala946Thr) [P(meta) = 3.08×10(-7); 0.88 (0.84-0.93)] and rs10930046 (Arg460His) [P(dom) = 1.16×10(-8); 0.70 (0.62-0.79)]. Both missense variants produced dramatic phenotypic changes in apoptosis and inflammation-related gene expression. We experimentally validated function of the intronic SNP by DNA electrophoresis, protein identification, and in vitro protein binding assays. DNA carrying the intronic risk allele rs13023380 showed reduced binding efficiency to a cellular protein complex including nucleolin and lupus autoantigen Ku70/80, and showed reduced transcriptional activity in vivo. Thus, in SLE patients, genetic susceptibility could create a biochemical imbalance that dysregulates nucleolin, Ku70/80, or other nucleic acid regulatory proteins. This could promote antibody hypermutation and auto-antibody generation, further destabilizing the cellular network. Together with molecular modeling, our results establish a distinct role for IFIH1 in apoptosis, inflammation, and autoantibody production, and explain the molecular basis of these three risk alleles for SLE pathogenesis.
- Published
- 2013
- Full Text
- View/download PDF
18. MicroRNA-3148 modulates allelic expression of toll-like receptor 7 variant associated with systemic lupus erythematosus.
- Author
-
Yun Deng, Jian Zhao, Daisuke Sakurai, Kenneth M Kaufman, Jeffrey C Edberg, Robert P Kimberly, Diane L Kamen, Gary S Gilkeson, Chaim O Jacob, R Hal Scofield, Carl D Langefeld, Jennifer A Kelly, Rosalind Ramsey-Goldman, Michelle A Petri, John D Reveille, Luis M Vilá, Graciela S Alarcón, Timothy J Vyse, Bernardo A Pons-Estel, Argentine Collaborative Group, Barry I Freedman, Patrick M Gaffney, Kathy Moser Sivils, Judith A James, Peter K Gregersen, Juan-Manuel Anaya, Timothy B Niewold, Joan T Merrill, Lindsey A Criswell, Anne M Stevens, Susan A Boackle, Rita M Cantor, Weiling Chen, Jeniffer M Grossman, Bevra H Hahn, John B Harley, Marta E Alarcόn-Riquelme, BIOLUPUS and GENLES networks, Elizabeth E Brown, and Betty P Tsao
- Subjects
Genetics ,QH426-470 - Abstract
We previously reported that the G allele of rs3853839 at 3'untranslated region (UTR) of Toll-like receptor 7 (TLR7) was associated with elevated transcript expression and increased risk for systemic lupus erythematosus (SLE) in 9,274 Eastern Asians [P = 6.5×10(-10), odds ratio (OR) (95%CI) = 1.27 (1.17-1.36)]. Here, we conducted trans-ancestral fine-mapping in 13,339 subjects including European Americans, African Americans, and Amerindian/Hispanics and confirmed rs3853839 as the only variant within the TLR7-TLR8 region exhibiting consistent and independent association with SLE (Pmeta = 7.5×10(-11), OR = 1.24 [1.18-1.34]). The risk G allele was associated with significantly increased levels of TLR7 mRNA and protein in peripheral blood mononuclear cells (PBMCs) and elevated luciferase activity of reporter gene in transfected cells. TLR7 3'UTR sequence bearing the non-risk C allele of rs3853839 matches a predicted binding site of microRNA-3148 (miR-3148), suggesting that this microRNA may regulate TLR7 expression. Indeed, miR-3148 levels were inversely correlated with TLR7 transcript levels in PBMCs from SLE patients and controls (R(2) = 0.255, P = 0.001). Overexpression of miR-3148 in HEK-293 cells led to significant dose-dependent decrease in luciferase activity for construct driven by TLR7 3'UTR segment bearing the C allele (P = 0.0003). Compared with the G-allele construct, the C-allele construct showed greater than two-fold reduction of luciferase activity in the presence of miR-3148. Reduced modulation by miR-3148 conferred slower degradation of the risk G-allele containing TLR7 transcripts, resulting in elevated levels of gene products. These data establish rs3853839 of TLR7 as a shared risk variant of SLE in 22,613 subjects of Asian, EA, AA, and Amerindian/Hispanic ancestries (Pmeta = 2.0×10(-19), OR = 1.25 [1.20-1.32]), which confers allelic effect on transcript turnover via differential binding to the epigenetic factor miR-3148.
- Published
- 2013
- Full Text
- View/download PDF
19. Trans-ancestral studies fine map the SLE-susceptibility locus TNFSF4.
- Author
-
Harinder Manku, Carl D Langefeld, Sandra G Guerra, Talat H Malik, Marta Alarcon-Riquelme, Juan-Manuel Anaya, Sang-Cheol Bae, Susan A Boackle, Elizabeth E Brown, Lindsey A Criswell, Barry I Freedman, Patrick M Gaffney, Peter A Gregersen, Joel M Guthridge, Sang-Hoon Han, John B Harley, Chaim O Jacob, Judith A James, Diane L Kamen, Kenneth M Kaufman, Jennifer A Kelly, Javier Martin, Joan T Merrill, Kathy L Moser, Timothy B Niewold, So-Yeon Park, Bernardo A Pons-Estel, Amr H Sawalha, R Hal Scofield, Nan Shen, Anne M Stevens, Celi Sun, Gary S Gilkeson, Jeff C Edberg, Robert P Kimberly, Swapan K Nath, Betty P Tsao, and Tim J Vyse
- Subjects
Genetics ,QH426-470 - Abstract
We previously established an 80 kb haplotype upstream of TNFSF4 as a susceptibility locus in the autoimmune disease SLE. SLE-associated alleles at this locus are associated with inflammatory disorders, including atherosclerosis and ischaemic stroke. In Europeans, the TNFSF4 causal variants have remained elusive due to strong linkage disequilibrium exhibited by alleles spanning the region. Using a trans-ancestral approach to fine-map the locus, utilising 17,900 SLE and control subjects including Amerindian/Hispanics (1348 cases, 717 controls), African-Americans (AA) (1529, 2048) and better powered cohorts of Europeans and East Asians, we find strong association of risk alleles in all ethnicities; the AA association replicates in African-American Gullah (152,122). The best evidence of association comes from two adjacent markers: rs2205960-T (P=1.71 × 10(-34) , OR=1.43[1.26-1.60]) and rs1234317-T (P=1.16 × 10(-28) , OR=1.38[1.24-1.54]). Inference of fine-scale recombination rates for all populations tested finds the 80 kb risk and non-risk haplotypes in all except African-Americans. In this population the decay of recombination equates to an 11 kb risk haplotype, anchored in the 5' region proximal to TNFSF4 and tagged by rs2205960-T after 1000 Genomes phase 1 (v3) imputation. Conditional regression analyses delineate the 5' risk signal to rs2205960-T and the independent non-risk signal to rs1234314-C. Our case-only and SLE-control cohorts demonstrate robust association of rs2205960-T with autoantibody production. The rs2205960-T is predicted to form part of a decameric motif which binds NF-κBp65 with increased affinity compared to rs2205960-G. ChIP-seq data also indicate NF-κB interaction with the DNA sequence at this position in LCL cells. Our research suggests association of rs2205960-T with SLE across multiple groups and an independent non-risk signal at rs1234314-C. rs2205960-T is associated with autoantibody production and lymphopenia. Our data confirm a global signal at TNFSF4 and a role for the expressed product at multiple stages of lymphocyte dysregulation during SLE pathogenesis. We confirm the validity of trans-ancestral mapping in a complex trait.
- Published
- 2013
- Full Text
- View/download PDF
20. Evidence of dynamically dysregulated gene expression pathways in hyperresponsive B cells from African American lupus patients.
- Author
-
Igor Dozmorov, Nicolas Dominguez, Andrea L Sestak, Julie M Robertson, John B Harley, Judith A James, and Joel M Guthridge
- Subjects
Medicine ,Science - Abstract
Recent application of gene expression profiling to the immune system has shown a great potential for characterization of complex regulatory processes. It is becoming increasingly important to characterize functional systems through multigene interactions to provide valuable insights into differences between healthy controls and autoimmune patients. Here we apply an original systematic approach to the analysis of changes in regulatory gene interconnections between in Epstein-Barr virus transformed hyperresponsive B cells from SLE patients and normal control B cells. Both traditional analysis of differential gene expression and analysis of the dynamics of gene expression variations were performed in combination to establish model networks of functional gene expression. This Pathway Dysregulation Analysis identified known transcription factors and transcriptional regulators activated uniquely in stimulated B cells from SLE patients.
- Published
- 2013
- Full Text
- View/download PDF
21. A genome-wide integrative genomic study localizes genetic factors influencing antibodies against Epstein-Barr virus nuclear antigen 1 (EBNA-1).
- Author
-
Rohina Rubicz, Robert Yolken, Eugene Drigalenko, Melanie A Carless, Thomas D Dyer, Lara Bauman, Phillip E Melton, Jack W Kent, John B Harley, Joanne E Curran, Matthew P Johnson, Shelley A Cole, Laura Almasy, Eric K Moses, Nikhil V Dhurandhar, Ellen Kraig, John Blangero, Charles T Leach, and Harald H H Göring
- Subjects
Genetics ,QH426-470 - Abstract
Infection with Epstein-Barr virus (EBV) is highly prevalent worldwide, and it has been associated with infectious mononucleosis and severe diseases including Burkitt lymphoma, Hodgkin lymphoma, nasopharyngeal lymphoma, and lymphoproliferative disorders. Although EBV has been the focus of extensive research, much still remains unknown concerning what makes some individuals more sensitive to infection and to adverse outcomes as a result of infection. Here we use an integrative genomics approach in order to localize genetic factors influencing levels of Epstein Barr virus (EBV) nuclear antigen-1 (EBNA-1) IgG antibodies, as a measure of history of infection with this pathogen, in large Mexican American families. Genome-wide evidence of both significant linkage and association was obtained on chromosome 6 in the human leukocyte antigen (HLA) region and replicated in an independent Mexican American sample of large families (minimum p-value in combined analysis of both datasets is 1.4×10(-15) for SNPs rs477515 and rs2516049). Conditional association analyses indicate the presence of at least two separate loci within MHC class II, and along with lymphocyte expression data suggest genes HLA-DRB1 and HLA-DQB1 as the best candidates. The association signals are specific to EBV and are not found with IgG antibodies to 12 other pathogens examined, and therefore do not simply reveal a general HLA effect. We investigated whether SNPs significantly associated with diseases in which EBV is known or suspected to play a role (namely nasopharyngeal lymphoma, Hodgkin lymphoma, systemic lupus erythematosus, and multiple sclerosis) also show evidence of associated with EBNA-1 antibody levels, finding an overlap only for the HLA locus, but none elsewhere in the genome. The significance of this work is that a major locus related to EBV infection has been identified, which may ultimately reveal the underlying mechanisms by which the immune system regulates infection with this pathogen.
- Published
- 2013
- Full Text
- View/download PDF
22. Preferential binding to Elk-1 by SLE-associated IL10 risk allele upregulates IL10 expression.
- Author
-
Daisuke Sakurai, Jian Zhao, Yun Deng, Jennifer A Kelly, Elizabeth E Brown, John B Harley, Sang-Cheol Bae, Marta E Alarcόn-Riquelme, BIOLUPUS and GENLES networks, Jeffrey C Edberg, Robert P Kimberly, Rosalind Ramsey-Goldman, Michelle A Petri, John D Reveille, Luis M Vilá, Graciela S Alarcón, Kenneth M Kaufman, Timothy J Vyse, Chaim O Jacob, Patrick M Gaffney, Kathy Moser Sivils, Judith A James, Diane L Kamen, Gary S Gilkeson, Timothy B Niewold, Joan T Merrill, R Hal Scofield, Lindsey A Criswell, Anne M Stevens, Susan A Boackle, Jae-Hoon Kim, Jiyoung Choi, Bernardo A Pons-Estel, Argentine Collaborative Group, Barry I Freedman, Juan-Manuel Anaya, Javier Martin, C Yung Yu, Deh-Ming Chang, Yeong Wook Song, Carl D Langefeld, Weiling Chen, Jennifer M Grossman, Rita M Cantor, Bevra H Hahn, and Betty P Tsao
- Subjects
Genetics ,QH426-470 - Abstract
Immunoregulatory cytokine interleukin-10 (IL-10) is elevated in sera from patients with systemic lupus erythematosus (SLE) correlating with disease activity. The established association of IL10 with SLE and other autoimmune diseases led us to fine map causal variant(s) and to explore underlying mechanisms. We assessed 19 tag SNPs, covering the IL10 gene cluster including IL19, IL20 and IL24, for association with SLE in 15,533 case and control subjects from four ancestries. The previously reported IL10 variant, rs3024505 located at 1 kb downstream of IL10, exhibited the strongest association signal and was confirmed for association with SLE in European American (EA) (P = 2.7×10⁻⁸, OR = 1.30), but not in non-EA ancestries. SNP imputation conducted in EA dataset identified three additional SLE-associated SNPs tagged by rs3024505 (rs3122605, rs3024493 and rs3024495 located at 9.2 kb upstream, intron 3 and 4 of IL10, respectively), and SLE-risk alleles of these SNPs were dose-dependently associated with elevated levels of IL10 mRNA in PBMCs and circulating IL-10 protein in SLE patients and controls. Using nuclear extracts of peripheral blood cells from SLE patients for electrophoretic mobility shift assays, we identified specific binding of transcription factor Elk-1 to oligodeoxynucleotides containing the risk (G) allele of rs3122605, suggesting rs3122605 as the most likely causal variant regulating IL10 expression. Elk-1 is known to be activated by phosphorylation and nuclear localization to induce transcription. Of interest, phosphorylated Elk-1 (p-Elk-1) detected only in nuclear extracts of SLE PBMCs appeared to increase with disease activity. Co-expression levels of p-Elk-1 and IL-10 were elevated in SLE T, B cells and monocytes, associated with increased disease activity in SLE B cells, and were best downregulated by ERK inhibitor. Taken together, our data suggest that preferential binding of activated Elk-1 to the IL10 rs3122605-G allele upregulates IL10 expression and confers increased risk for SLE in European Americans.
- Published
- 2013
- Full Text
- View/download PDF
23. Genome-wide association study of African and European Americans implicates multiple shared and ethnic specific loci in sarcoidosis susceptibility.
- Author
-
Indra Adrianto, Chee Paul Lin, Jessica J Hale, Albert M Levin, Indrani Datta, Ryan Parker, Adam Adler, Jennifer A Kelly, Kenneth M Kaufman, Christopher J Lessard, Kathy L Moser, Robert P Kimberly, John B Harley, Michael C Iannuzzi, Benjamin A Rybicki, and Courtney G Montgomery
- Subjects
Medicine ,Science - Abstract
Sarcoidosis is a systemic inflammatory disease characterized by the formation of granulomas in affected organs. Genome-wide association studies (GWASs) of this disease have been conducted only in European population. We present the first sarcoidosis GWAS in African Americans (AAs, 818 cases and 1,088 related controls) followed by replication in independent sets of AAs (455 cases and 557 controls) and European Americans (EAs, 442 cases and 2,284 controls). We evaluated >6 million SNPs either genotyped using the Illumina Omni1-Quad array or imputed from the 1000 Genomes Project data. We identified a novel sarcoidosis-associated locus, NOTCH4, that reached genome-wide significance in the combined AA samples (rs715299, P(AA-meta) = 6.51 × 10(-10)) and demonstrated the independence of this locus from others in the MHC region in the same sample. We replicated previous European GWAS associations within HLA-DRA, HLA-DRB5, HLA-DRB1, BTNL2, and ANXA11 in both our AA and EA datasets. We also confirmed significant associations to the previously reported HLA-C and HLA-B regions in the EA but not AA samples. We further identified suggestive associations with several other genes previously reported in lung or inflammatory diseases.
- Published
- 2012
- Full Text
- View/download PDF
24. A comprehensive analysis of shared loci between systemic lupus erythematosus (SLE) and sixteen autoimmune diseases reveals limited genetic overlap.
- Author
-
Paula S Ramos, Lindsey A Criswell, Kathy L Moser, Mary E Comeau, Adrienne H Williams, Nicholas M Pajewski, Sharon A Chung, Robert R Graham, Raphael Zidovetzki, Jennifer A Kelly, Kenneth M Kaufman, Chaim O Jacob, Timothy J Vyse, Betty P Tsao, Robert P Kimberly, Patrick M Gaffney, Marta E Alarcón-Riquelme, John B Harley, Carl D Langefeld, and International Consortium on the Genetics of Systemic Erythematosus
- Subjects
Genetics ,QH426-470 - Abstract
In spite of the well-known clustering of multiple autoimmune disorders in families, analyses of specific shared genes and polymorphisms between systemic lupus erythematosus (SLE) and other autoimmune diseases (ADs) have been limited. Therefore, we comprehensively tested autoimmune variants for association with SLE, aiming to identify pleiotropic genetic associations between these diseases. We compiled a list of 446 non-Major Histocompatibility Complex (MHC) variants identified in genome-wide association studies (GWAS) of populations of European ancestry across 17 ADs. We then tested these variants in our combined Caucasian SLE cohorts of 1,500 cases and 5,706 controls. We tested a subset of these polymorphisms in an independent Caucasian replication cohort of 2,085 SLE cases and 2,854 controls, allowing the computation of a meta-analysis between all cohorts. We have uncovered novel shared SLE loci that passed multiple comparisons adjustment, including the VTCN1 (rs12046117, P = 2.02×10(-06)) region. We observed that the loci shared among the most ADs include IL23R, OLIG3/TNFAIP3, and IL2RA. Given the lack of a universal autoimmune risk locus outside of the MHC and variable specificities for different diseases, our data suggests partial pleiotropy among ADs. Hierarchical clustering of ADs suggested that the most genetically related ADs appear to be type 1 diabetes with rheumatoid arthritis and Crohn's disease with ulcerative colitis. These findings support a relatively distinct genetic susceptibility for SLE. For many of the shared GWAS autoimmune loci, we found no evidence for association with SLE, including IL23R. Also, several established SLE loci are apparently not associated with other ADs, including the ITGAM-ITGAX and TNFSF4 regions. This study represents the most comprehensive evaluation of shared autoimmune loci to date, supports a relatively distinct non-MHC genetic susceptibility for SLE, provides further evidence for previously and newly identified shared genes in SLE, and highlights the value of studies of potentially pleiotropic genes in autoimmune diseases.
- Published
- 2011
- Full Text
- View/download PDF
25. Association of genetic variants in complement factor H and factor H-related genes with systemic lupus erythematosus susceptibility.
- Author
-
Jian Zhao, Hui Wu, Melanie Khosravi, Huijuan Cui, Xiaoxia Qian, Jennifer A Kelly, Kenneth M Kaufman, Carl D Langefeld, Adrienne H Williams, Mary E Comeau, Julie T Ziegler, Miranda C Marion, Adam Adler, Stuart B Glenn, Marta E Alarcón-Riquelme, BIOLUPUS Network, GENLES Network, Bernardo A Pons-Estel, John B Harley, Sang-Cheol Bae, So-Young Bang, Soo-Kyung Cho, Chaim O Jacob, Timothy J Vyse, Timothy B Niewold, Patrick M Gaffney, Kathy L Moser, Robert P Kimberly, Jeffrey C Edberg, Elizabeth E Brown, Graciela S Alarcon, Michelle A Petri, Rosalind Ramsey-Goldman, Luis M Vilá, John D Reveille, Judith A James, Gary S Gilkeson, Diane L Kamen, Barry I Freedman, Juan-Manuel Anaya, Joan T Merrill, Lindsey A Criswell, R Hal Scofield, Anne M Stevens, Joel M Guthridge, Deh-Ming Chang, Yeong Wook Song, Ji Ah Park, Eun Young Lee, Susan A Boackle, Jennifer M Grossman, Bevra H Hahn, Timothy H J Goodship, Rita M Cantor, Chack-Yung Yu, Nan Shen, and Betty P Tsao
- Subjects
Genetics ,QH426-470 - Abstract
Systemic lupus erythematosus (SLE), a complex polygenic autoimmune disease, is associated with increased complement activation. Variants of genes encoding complement regulator factor H (CFH) and five CFH-related proteins (CFHR1-CFHR5) within the chromosome 1q32 locus linked to SLE, have been associated with multiple human diseases and may contribute to dysregulated complement activation predisposing to SLE. We assessed 60 SNPs covering the CFH-CFHRs region for association with SLE in 15,864 case-control subjects derived from four ethnic groups. Significant allelic associations with SLE were detected in European Americans (EA) and African Americans (AA), which could be attributed to an intronic CFH SNP (rs6677604, in intron 11, P(meta) = 6.6×10(-8), OR = 1.18) and an intergenic SNP between CFHR1 and CFHR4 (rs16840639, P(meta) = 2.9×10(-7), OR = 1.17) rather than to previously identified disease-associated CFH exonic SNPs, including I62V, Y402H, A474A, and D936E. In addition, allelic association of rs6677604 with SLE was subsequently confirmed in Asians (AS). Haplotype analysis revealed that the underlying causal variant, tagged by rs6677604 and rs16840639, was localized to a ~146 kb block extending from intron 9 of CFH to downstream of CFHR1. Within this block, the deletion of CFHR3 and CFHR1 (CFHR3-1Δ), a likely causal variant measured using multiplex ligation-dependent probe amplification, was tagged by rs6677604 in EA and AS and rs16840639 in AA, respectively. Deduced from genotypic associations of tag SNPs in EA, AA, and AS, homozygous deletion of CFHR3-1Δ (P(meta) = 3.2×10(-7), OR = 1.47) conferred a higher risk of SLE than heterozygous deletion (P(meta) = 3.5×10(-4), OR = 1.14). These results suggested that the CFHR3-1Δ deletion within the SLE-associated block, but not the previously described exonic SNPs of CFH, might contribute to the development of SLE in EA, AA, and AS, providing new insights into the role of complement regulators in the pathogenesis of SLE.
- Published
- 2011
- Full Text
- View/download PDF
26. Differential genetic associations for systemic lupus erythematosus based on anti-dsDNA autoantibody production.
- Author
-
Sharon A Chung, Kimberly E Taylor, Robert R Graham, Joanne Nititham, Annette T Lee, Ward A Ortmann, Chaim O Jacob, Marta E Alarcón-Riquelme, Betty P Tsao, John B Harley, Patrick M Gaffney, Kathy L Moser, SLEGEN, Michelle Petri, F Yesim Demirci, M Ilyas Kamboh, Susan Manzi, Peter K Gregersen, Carl D Langefeld, Timothy W Behrens, and Lindsey A Criswell
- Subjects
Genetics ,QH426-470 - Abstract
Systemic lupus erythematosus (SLE) is a clinically heterogeneous, systemic autoimmune disease characterized by autoantibody formation. Previously published genome-wide association studies (GWAS) have investigated SLE as a single phenotype. Therefore, we conducted a GWAS to identify genetic factors associated with anti-dsDNA autoantibody production, a SLE-related autoantibody with diagnostic and clinical importance. Using two independent datasets, over 400,000 single nucleotide polymorphisms (SNPs) were studied in a total of 1,717 SLE cases and 4,813 healthy controls. Anti-dsDNA autoantibody positive (anti-dsDNA +, n = 811) and anti-dsDNA autoantibody negative (anti-dsDNA -, n = 906) SLE cases were compared to healthy controls and to each other to identify SNPs associated specifically with these SLE subtypes. SNPs in the previously identified SLE susceptibility loci STAT4, IRF5, ITGAM, and the major histocompatibility complex were strongly associated with anti-dsDNA + SLE. Far fewer and weaker associations were observed for anti-dsDNA - SLE. For example, rs7574865 in STAT4 had an OR for anti-dsDNA + SLE of 1.77 (95% CI 1.57-1.99, p = 2.0E-20) compared to an OR for anti-dsDNA - SLE of 1.26 (95% CI 1.12-1.41, p = 2.4E-04), with p(heterogeneity)
- Published
- 2011
- Full Text
- View/download PDF
27. Risk alleles for systemic lupus erythematosus in a large case-control collection and associations with clinical subphenotypes.
- Author
-
Kimberly E Taylor, Sharon A Chung, Robert R Graham, Ward A Ortmann, Annette T Lee, Carl D Langefeld, Chaim O Jacob, M Ilyas Kamboh, Marta E Alarcón-Riquelme, Betty P Tsao, Kathy L Moser, Patrick M Gaffney, John B Harley, Michelle Petri, Susan Manzi, Peter K Gregersen, Timothy W Behrens, and Lindsey A Criswell
- Subjects
Genetics ,QH426-470 - Abstract
Systemic lupus erythematosus (SLE) is a genetically complex disease with heterogeneous clinical manifestations. Recent studies have greatly expanded the number of established SLE risk alleles, but the distribution of multiple risk alleles in cases versus controls and their relationship to subphenotypes have not been studied. We studied 22 SLE susceptibility polymorphisms with previous genome-wide evidence of association (p < 5 x 10⁻¹²⁸) in 1919 SLE cases from 9 independent Caucasian SLE case series and 4813 independent controls. The mean number of risk alleles in cases was 15.1 (SD 3.1) while the mean in controls was 13.1 (SD 2.8), with trend p = 4 x 10⁻⁸. We defined a genetic risk score (GRS) for SLE as the number of risk alleles with each weighted by the SLE risk odds ratio (OR). The OR for high-low GRS tertiles, adjusted for intra-European ancestry, sex, and parent study, was 4.4 (95% CI 3.8-5.1). We studied associations of individual SNPs and the GRS with clinical manifestations for the cases: age at diagnosis, the 11 American College of Rheumatology classification criteria, and double-stranded DNA antibody (anti-dsDNA) production. Six subphenotypes were significantly associated with the GRS, most notably anti-dsDNA (OR(high-low) = 2.36, p = 9e-9), the immunologic criterion (OR(high-low) = 2.23, p = 3e-7), and age at diagnosis (OR(high-low) = 1.45, p = 0.0060). Finally, we developed a subphenotype-specific GRS (sub-GRS) for each phenotype with more power to detect cumulative genetic associations. The sub-GRS was more strongly associated than any single SNP effect for 5 subphenotypes (the above plus hematologic disorder and oral ulcers), while single loci are more significantly associated with renal disease (HLA-DRB1, OR = 1.37, 95% CI 1.14-1.64) and arthritis (ITGAM, OR = 0.72, 95% CI 0.59-0.88). We did not observe significant associations for other subphenotypes, for individual loci or the sub-GRS. Thus our analysis categorizes SLE subphenotypes into three groups: those having cumulative, single, and no known genetic association with respect to the currently established SLE risk loci.
- Published
- 2011
- Full Text
- View/download PDF
28. Identification of unique microRNA signature associated with lupus nephritis.
- Author
-
Jeannie L Te, Igor M Dozmorov, Joel M Guthridge, Kim L Nguyen, Joshua W Cavett, Jennifer A Kelly, Gail R Bruner, John B Harley, and Joshua O Ojwang
- Subjects
Medicine ,Science - Abstract
MicroRNAs (miRNA) have emerged as an important new class of modulators of gene expression. In this study we investigated miRNA that are differentially expressed in lupus nephritis. Microarray technology was used to investigate differentially expressed miRNA in peripheral blood mononuclear cells (PBMCs) and Epstein-Barr Virus (EBV)-transformed cell lines obtained from lupus nephritis affected patients and unaffected controls. TaqMan-based stem-loop real-time polymerase chain reaction was used for validation. Microarray analysis of miRNA expressed in both African American (AA) and European American (EA) derived lupus nephritis samples revealed 29 and 50 differentially expressed miRNA, respectively, of 850 tested. There were 18 miRNA that were differentially expressed in both racial groups. When samples from both racial groups and different specimen types were considered, there were 5 primary miRNA that were differentially expressed. We have identified 5 miRNA; hsa-miR-371-5P, hsa-miR-423-5P, hsa-miR-638, hsa-miR-1224-3P and hsa-miR-663 that were differentially expressed in lupus nephritis across different racial groups and all specimen types tested. Hsa-miR-371-5P, hsa-miR-1224-3P and hsa-miR-423-5P, are reported here for the first time to be associated with lupus nephritis. Our work establishes EBV-transformed B cell lines as a useful model for the discovery of miRNA as biomarkers for SLE. Based on these findings, we postulate that these differentially expressed miRNA may be potential novel biomarkers for SLE as well as help elucidate pathogenic mechanisms of lupus nephritis. The investigation of miRNA profiles in SLE may lead to the discovery and development of novel methods to diagnosis, treat and prevent SLE.
- Published
- 2010
- Full Text
- View/download PDF
29. 60 kD Ro and nRNP A frequently initiate human lupus autoimmunity.
- Author
-
Latisha D Heinlen, Micah T McClain, Lauren L Ritterhouse, Benjamin F Bruner, Colin C Edgerton, Michael P Keith, Judith A James, and John B Harley
- Subjects
Medicine ,Science - Abstract
Systemic lupus erythematosus (SLE) is a clinically heterogeneous, humoral autoimmune disorder. The unifying feature among SLE patients is the production of large quantities of autoantibodies. Serum samples from 129 patients collected before the onset of SLE and while in the United States military were evaluated for early pre-clinical serologic events. The first available positive serum sample frequently already contained multiple autoantibody specificities (65%). However, in 34 SLE patients the earliest pre-clinical serum sample positive for any detectable common autoantibody bound only a single autoantigen, most commonly 60 kD Ro (29%), nRNP A (24%), anti-phospholipids (18%) or rheumatoid factor (15%). We identified several recurrent patterns of autoantibody onset using these pre-diagnostic samples. In the serum samples available, anti-nRNP A appeared before or simultaneously with anti-nRNP 70 K in 96% of the patients who had both autoantibodies at diagnosis. Anti-60 kD Ro antibodies appeared before or simultaneously with anti-La (98%) or anti-52 kD Ro (95%). The autoantibody response in SLE patients begins simply, often binding a single specific autoantigen years before disease onset, followed by epitope spreading to additional autoantigenic specificities that are accrued in recurring patterns.
- Published
- 2010
- Full Text
- View/download PDF
30. High-density SNP screening of the major histocompatibility complex in systemic lupus erythematosus demonstrates strong evidence for independent susceptibility regions.
- Author
-
Lisa F Barcellos, Suzanne L May, Patricia P Ramsay, Hong L Quach, Julie A Lane, Joanne Nititham, Janelle A Noble, Kimberly E Taylor, Diana L Quach, Sharon A Chung, Jennifer A Kelly, Kathy L Moser, Timothy W Behrens, Michael F Seldin, Glenys Thomson, John B Harley, Patrick M Gaffney, and Lindsey A Criswell
- Subjects
Genetics ,QH426-470 - Abstract
A substantial genetic contribution to systemic lupus erythematosus (SLE) risk is conferred by major histocompatibility complex (MHC) gene(s) on chromosome 6p21. Previous studies in SLE have lacked statistical power and genetic resolution to fully define MHC influences. We characterized 1,610 Caucasian SLE cases and 1,470 parents for 1,974 MHC SNPs, the highly polymorphic HLA-DRB1 locus, and a panel of ancestry informative markers. Single-marker analyses revealed strong signals for SNPs within several MHC regions, as well as with HLA-DRB1 (global p = 9.99 x 10(-16)). The most strongly associated DRB1 alleles were: *0301 (odds ratio, OR = 2.21, p = 2.53 x 10(-12)), *1401 (OR = 0.50, p = 0.0002), and *1501 (OR = 1.39, p = 0.0032). The MHC region SNP demonstrating the strongest evidence of association with SLE was rs3117103, with OR = 2.44 and p = 2.80 x 10(-13). Conditional haplotype and stepwise logistic regression analyses identified strong evidence for association between SLE and the extended class I, class I, class III, class II, and the extended class II MHC regions. Sequential removal of SLE-associated DRB1 haplotypes revealed independent effects due to variation within OR2H2 (extended class I, rs362521, p = 0.006), CREBL1 (class III, rs8283, p = 0.01), and DQB2 (class II, rs7769979, p = 0.003, and rs10947345, p = 0.0004). Further, conditional haplotype analyses demonstrated that variation within MICB (class I, rs3828903, p = 0.006) also contributes to SLE risk independent of HLA-DRB1*0301. Our results for the first time delineate with high resolution several MHC regions with independent contributions to SLE risk. We provide a list of candidate variants based on biologic and functional considerations that may be causally related to SLE risk and warrant further investigation.
- Published
- 2009
- Full Text
- View/download PDF
31. Osteopontin and systemic lupus erythematosus association: a probable gene-gender interaction.
- Author
-
Shizhong Han, Joel M Guthridge, Isaac T W Harley, Andrea L Sestak, Xana Kim-Howard, Kenneth M Kaufman, Bahram Namjou, Harshal Deshmukh, Gail Bruner, Luis R Espinoza, Gary S Gilkeson, John B Harley, Judith A James, and Swapan K Nath
- Subjects
Medicine ,Science - Abstract
Osteopontin (SPP1) is an important bone matrix mediator found to have key roles in inflammation and immunity. SPP1 genetic polymorphisms and increased osteopontin protein levels have been reported to be associated with SLE in small patient collections. The present study evaluates association between SPP1 polymorphisms and SLE in a large cohort of 1141 unrelated SLE patients [707 European-American (EA) and 434 African-American (AA)], and 2009 unrelated controls (1309 EA and 700 AA). Population-based case-control association analyses were performed. To control for potential population stratification, admixture adjusted logistic regression, genomic control (GC), structured association (STRAT), and principal components analysis (PCA) were applied. Combined analysis of 2 ethnic groups, showed the minor allele of 2 SNPs (rs1126616T and rs9138C) significantly associated with higher risk of SLE in males (P = 0.0005, OR = 1.73, 95% CI = 1.28-2.33), but not in females. Indeed, significant gene-gender interactions in the 2 SNPs, rs1126772 and rs9138, were detected (P = 0.001 and P = 0.0006, respectively). Further, haplotype analysis identified rs1126616T-rs1126772A-rs9138C which demonstrated significant association with SLE in general (P = 0.02, OR = 1.30, 95%CI 1.08-1.57), especially in males (P = 0.0003, OR = 2.42, 95%CI 1.51-3.89). Subgroup analysis with single SNPs and haplotypes also identified a similar pattern of gender-specific association in AA and EA. GC, STRAT, and PCA results within each group showed consistent associations. Our data suggest SPP1 is associated with SLE, and this association is especially stronger in males. To our knowledge, this report serves as the first association of a specific autosomal gene with human male lupus.
- Published
- 2008
- Full Text
- View/download PDF
32. Common variants within MECP2 confer risk of systemic lupus erythematosus.
- Author
-
Amr H Sawalha, Ryan Webb, Shizhong Han, Jennifer A Kelly, Kenneth M Kaufman, Robert P Kimberly, Marta E Alarcón-Riquelme, Judith A James, Timothy J Vyse, Gary S Gilkeson, Chan-Bum Choi, R Hal Scofield, Sang-Cheol Bae, Swapan K Nath, and John B Harley
- Subjects
Medicine ,Science - Abstract
Systemic lupus erythematosus (SLE) is a predominantly female autoimmune disease that affects multiple organ systems. Herein, we report on an X-chromosome gene association with SLE. Methyl-CpG-binding protein 2 (MECP2) is located on chromosome Xq28 and encodes for a protein that plays a critical role in epigenetic transcriptional regulation of methylation-sensitive genes. Utilizing a candidate gene association approach, we genotyped 21 SNPs within and around MECP2 in SLE patients and controls. We identify and replicate association between SLE and the genomic element containing MECP2 in two independent SLE cohorts from two ethnically divergent populations. These findings are potentially related to the overexpression of methylation-sensitive genes in SLE.
- Published
- 2008
- Full Text
- View/download PDF
33. Impact of race and ethnicity on family participation in systemic lupus erythematosus genetic studies
- Author
-
R Hal Scofield, Rohan Sharma, Teresa Aberle, Carisa M Cooney, Jennifer A Kelly, John B Harley, and Astrid Rasmussen
- Abstract
ObjectiveSystemic lupus erythematosus (SLE) has a higher prevalence and is more severe in African Americans and Hispanics than in non-Hispanic Whites. To understand the shared and unique genetic risk factors of these populations, an adequate representation of African Americans and Hispanics in clinical and genetic research is indispensable while challenging. The goal of this study was to identify differences in research participation of families of different racial and ethnic backgrounds and the potential causes for the disparities.MethodsFamilies were screened for eligibility to the Lupus Family Registry and Repository (LFRR) after self-referral or physician referral. We recorded the sociodemographic characteristics, self-identified race and ethnicity, ACR-SLE criteria, and the reasons given for not completing study participation for all families.ResultsA total of 1,472 families (950 non-Hispanic White, 405 African American, and 117 Hispanic) were screened but only 366 completed study participation (25%). Participation rates and reasons for non-participation varied between racial and ethnic groups. The main reason for African American families to not participate was that subjects critical to the family structure declined participation (OR = 1.6, p = 0.0001), while for White families, the main cause was that purported SLE patients did not meet ACR SLE criteria (OR = 1.81, p p ConclusionsSuccessful recruitment of patients, families, and specific demographic groups is critical for the study of genetically complex diseases, such as SLE. There are significant disparities in SLE family recruitment across groups of people, likely due to their richly different cultures and environments.
- Published
- 2023
34. Antibody Responses to <scp>Epstein‐Barr</scp> Virus in the Preclinical Period of Rheumatoid Arthritis Suggest the Presence of Increased Viral Reactivation Cycles
- Author
-
William H. Robinson, Jill A Norris, V. Michael Holers, Carla J Guthridge, Kevin D. Deane, Nichole E. Carlson, M. Kristen Demoruelle, Heather M. Berens, Jess D. Edison, Rachel L. Johnson, Elizabeth A. Bemis, Sabrina Fechtner, John B. Harley, and Judith A. James
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,medicine.medical_treatment ,Immunology ,Antibodies, Viral ,medicine.disease_cause ,Anti-Citrullinated Protein Antibodies ,Article ,Virus ,Autoimmunity ,Arthritis, Rheumatoid ,Immune system ,Rheumatology ,Rheumatoid Factor ,Humans ,Immunology and Allergy ,Medicine ,biology ,business.industry ,Autoantibody ,medicine.disease ,Epstein–Barr virus ,Immunoglobulin A ,Cytokine ,Immunoglobulin M ,Immunoglobulin G ,Rheumatoid arthritis ,Antibody Formation ,Cytomegalovirus Infections ,biology.protein ,Antibody ,business - Abstract
Patients with established rheumatoid arthritis (RA) demonstrate altered immune responses to Epstein-Barr virus (EBV), but the presence and roles of EBV have not been fully explored during the pre-clinical disease period. This study was undertaken to determine if EBV infection, as evidenced by an altered anti-EBV antibody response, either plays an important role in driving the development of RA or is a result of expanded RA-related autoimmunity.A total of 83 subjects with RA according to the 1987 American College of Rheumatology (ACR) criteria and 83 age-, sex-, and race-matched control subjects without RA were included in our study. We collected sera from RA subjects and matched controls during the pre-RA and post-RA diagnosis periods and tested the sera for the presence of 5 anti-EBV antibodies (anti-EBV nuclear antigen 1 IgG isotype, anti-viral capsid antigen [anti-VCA] isotypes IgG and IgA, and anti-early antigen [EA] isotypes IgG and IgA), 7 RA-related autoantibodies (rheumatoid factor measured by nephelometry [RF-Neph] as well as isotype-specific IgA-RF, IgM-RF, and IgG-RF, and anti-cyclic citrullinated peptide [anti-CCP] antibodies, including anti-CCP2, anti-CCP3, and anti-CCP3.1), 22 cytokines/chemokines, 36 individual anti-citrullinated protein antibodies, and IgG-cytomegalovirus (CMV) antibodies. Random forest classification, mixed modeling, and joint mixed modeling were used to determine differences in anti-EBV antibody levels between RA subjects and controls.Random forest analysis identified the presence of preclinical EBV antibodies in the serum that differentiated RA subjects from controls without RA. Specifically, IgG-EA antibody levels were higher in RA subjects (mean ± SD 0.82 ± 0.72 international standardized ratio [ISR]) compared to controls (mean ± SD 0.49 ± 0.28 ISR). In subjects with RA, elevated serum IgG-EA levels in the preclinical period before seroconversion were significantly correlated with increased serum IgM-RF levels (P = 0.007), whereas this correlation was not seen in control subjects without RA (P = 0.15). IgG-CMV antibody levels did not differ between groups.Subjects whose serum IgG-EA antibody levels are elevated in the preclinical period will eventually develop RA, which suggests that EBV reactivation cycles are increased during the preclinical period of RA. A combination of RF and EBV reactivation may play an important role in the development of RA.
- Published
- 2022
35. Reanalysis of eMERGE phase III sequence variants in 10,500 participants and infrastructure to support the automated return of knowledge updates
- Author
-
Matthew S. Lebo, Christopher Koch, Lisa Mahanta, Scott T. Weiss, John B. Harley, Gail P. Jarvik, Prathik K. Vijay Kumar, Melanie F. Myers, Kathleen A. Leppig, Elizabeth W. Karlson, Cynthia A. Prows, Marc S. Williams, Andrea M. Oza, Heidi L. Rehm, Hana Zouk, Megan H. Hawley, and Wanfeng Yu
- Subjects
Computer science ,Phase (waves) ,Genetic Variation ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Genomics ,Computational biology ,Article ,Genetics (clinical) ,Sequence (medicine) - Abstract
The clinical genomics knowledgebase is dynamic with variant classifications changing as newly identified cases, additional population data, and other evidence become available. This is a challenge for the clinical laboratory because of limited resource availability for variant reassessment.Throughout the Electronic Medical Records and Genomics phase III program, clinical sites associated with the Mass General Brigham/Broad sequencing center received automated, real-time notifications when reported variants were reclassified. In this study, we summarized the nature of these reclassifications and described the proactive reassessment framework we used for the Electronic Medical Records and Genomics program data set to identify variants most likely to undergo reclassification.Reanalysis of 1855 variants led to the reclassification of 2% (n = 45) of variants, affecting 0.6% (n = 67) of participants. Of these reclassifications, 78% (n = 35) were high-impact changes affecting reportability, with 8 variants downgraded from likely pathogenic/pathogenic to variants of uncertain significance (VUS) and 27 variants upgraded from VUS to likely pathogenic/pathogenic. Most upgraded variants (67%) were initially classified as VUS-Favor Pathogenic, highlighting the benefit of VUS subcategorization. The most common reason for reclassification was new published case data and/or functional evidence.Our results highlight the importance of periodic sequence variant reevaluation and the need for automated approaches to advance routine implementation of variant reevaluations in clinical practice.
- Published
- 2022
36. Author Correction: Genetic regulation of serum IgA levels and susceptibility to common immune, infectious, kidney, and cardio-metabolic traits
- Author
-
Lili Liu, Atlas Khan, Elena Sanchez-Rodriguez, Francesca Zanoni, Yifu Li, Nicholas Steers, Olivia Balderes, Junying Zhang, Priya Krithivasan, Robert A. LeDesma, Clara Fischman, Scott J. Hebbring, John B. Harley, Halima Moncrieffe, Leah C. Kottyan, Bahram Namjou-Khales, Theresa L. Walunas, Rachel Knevel, Soumya Raychaudhuri, Elizabeth W. Karlson, Joshua C. Denny, Ian B. Stanaway, David Crosslin, Thomas Rauen, Jürgen Floege, Frank Eitner, Zina Moldoveanu, Colin Reily, Barbora Knoppova, Stacy Hall, Justin T. Sheff, Bruce A. Julian, Robert J. Wyatt, Hitoshi Suzuki, Jingyuan Xie, Nan Chen, Xujie Zhou, Hong Zhang, Lennart Hammarström, Alexander Viktorin, Patrik K. E. Magnusson, Ning Shang, George Hripcsak, Chunhua Weng, Tatjana Rundek, Mitchell S. V. Elkind, Elizabeth C. Oelsner, R. Graham Barr, Iuliana Ionita-Laza, Jan Novak, Ali G. Gharavi, and Krzysztof Kiryluk
- Subjects
Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Published
- 2023
37. 1401 A Genome Wide Association Scan of SLE genetic risk in a cohort of African-American persons
- Author
-
Isaac TW Harley, Celi Sun, Adrienne H Williams, Julie T Ziegler, Mary E Comeau, Miranda C Marion, Stuart B Glenn, Adam Adler, Summer G Frank-Pearce, Nan Shen, Jennifer A Kelly, Bahram Namjou-Khales, Michelle Petri, Marta Alarcon-Riquelme, W Joseph McCune, Patrick Gaffney, Kathy Sivils, Jane E Salmon, Michael H Weisman, Jeffrey C Edberg, Elizabeth E Brown, Tammy Utset, Lindsey A Criswell, Chaim O Jacob, Betty Tsao, Timothy J Vyse, Judith A James, Gary S Gilkeson, Diane L Kamen, Courtney Montgomery, Joan T Merrill, Swapan K Nath, Viktoryia Laurynenka, Iouri Chepelev, Valerie Harris-Lewis, R Hal Scofield, Robert P Kimberly, Carl D Langefeld, John B Harley, and Kenneth M Kaufman
- Published
- 2022
38. 909 Haplotype-specific chromatin looping reveals genetic interactions of regulatory regions modulating gene expression at the SLE susceptibility locusFAM167A-BLK
- Author
-
Mariana Saint-Just Ribeiro, Pulak Tripathi, Bahram Namjou, John B Harley, and Iouri Chepelev
- Published
- 2022
39. The US Department of Veterans Affairs Science and Health Initiative to Combat Infectious and Emerging Life-Threatening Diseases (VA SHIELD): A Biorepository Addressing National Health Threats
- Author
-
John B Harley, Saiju Pyarajan, Elizabeth S Partan, Lauren Epstein, Jason A Wertheim, Abhinav Diwan, Christopher W Woods, Victoria Davey, Sharlene Blair, Dennis H Clark, Kenneth M Kaufman, Shagufta Khan, Iouri Chepelev, Alexander Devine, Perry Cameron, Monica F McCann, Mary Cloud B Ammons, Devin D Bolz, Jane K Battles, Jeffrey L Curtis, Mark Holodniy, Vincent C Marconi, Charles D Searles, David O Beenhouwer, Sheldon T Brown, Jonathan P Moorman, Zhi Q Yao, Maria C Rodriguez-Barradas, Shyam Mohapatra, Osmara Y Molina De Rodriguez, Emerson B Padiernos, Eric R McIndoo, Emily Price, Hailey M Burgoyne, Ian Robey, Dawn C Schwenke, Carey L Shive, Ronald M Przygodzki, Rachel B Ramoni, Holly K Krull, and Robert A Bonomo
- Subjects
Infectious Diseases ,Oncology - Abstract
Background The coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has demonstrated the need to share data and biospecimens broadly to optimize clinical outcomes for US military Veterans. Methods In response, the Veterans Health Administration established VA SHIELD (Science and Health Initiative to Combat Infectious and Emerging Life-threatening Diseases), a comprehensive biorepository of specimens and clinical data from affected Veterans to advance research and public health surveillance and to improve diagnostic and therapeutic capabilities. Results VA SHIELD now comprises 12 sites collecting de-identified biospecimens from US Veterans affected by SARS-CoV-2. In addition, 2 biorepository sites, a data processing center, and a coordinating center have been established under the direction of the Veterans Affairs Office of Research and Development. Phase 1 of VA SHIELD comprises 34 157 samples. Of these, 83.8% had positive tests for SARS-CoV-2, with the remainder serving as contemporaneous controls. The samples include nasopharyngeal swabs (57.9%), plasma (27.9%), and sera (12.5%). The associated clinical and demographic information available permits the evaluation of biological data in the context of patient demographics, clinical experience and management, vaccinations, and comorbidities. Conclusions VA SHIELD is representative of US national diversity with a significant potential to impact national healthcare. VA SHIELD will support future projects designed to better understand SARS-CoV-2 and other emergent healthcare crises. To the extent possible, VA SHIELD will facilitate the discovery of diagnostics and therapeutics intended to diminish COVID-19 morbidity and mortality and to reduce the impact of new emerging threats to the health of US Veterans and populations worldwide.
- Published
- 2022
40. Lupus Susceptibility Region Containing CDKN1B rs34330 Mechanistically Influences Expression and Function of Multiple Target Genes, Also Linked to Proliferation and Apoptosis
- Author
-
Xu-jie Zhou, Yuta Kochi, Guru P. Maiti, Joel M. Guthridge, Sang Cheol Bae, Mehdi Fazel-Najafabadi, Kek Heng Chua, Judith A. James, Loren L. Looger, Yukinori Okada, Swapan K. Nath, Chikashi Terao, Bhupinder Singh, Celi Sun, Hong Zhang, Matthew T. Weirauch, John B. Harley, and Gaurav K. Varshney
- Subjects
1.1 Normal biological development and functioning ,Immunology ,Lupus ,Apoptosis ,Genome-wide association study ,Biology ,Polymorphism, Single Nucleotide ,Autoimmune Disease ,Article ,Promoter Regions ,Genetic ,Rheumatology ,Underpinning research ,Genetics ,Lupus Erythematosus, Systemic ,Humans ,2.1 Biological and endogenous factors ,Immunology and Allergy ,Genetic Predisposition to Disease ,Epigenetics ,Polymorphism ,Aetiology ,Promoter Regions, Genetic ,Enhancer ,Transcription factor ,Alleles ,Cell Proliferation ,Lupus Erythematosus ,Inflammatory and immune system ,Systemic ,Human Genome ,Computational Biology ,Promoter ,Single Nucleotide ,Chromatin ,CTCF ,H3K4me3 ,Generic health relevance ,Cyclin-Dependent Kinase Inhibitor p27 ,Genome-Wide Association Study ,Biotechnology - Abstract
ObjectiveIn a recent genome-wide association study, a significant genetic association between rs34330 of CDKN1B and risk of systemic lupus erythematosus (SLE) in Han Chinese was identified. This study was undertaken to validate the reported association and elucidate the biochemical mechanisms underlying the effect of the variant.MethodsWe performed an allelic association analysis in patients with SLE, followed by a meta-analysis assessing genome-wide association data across 11 independent cohorts (n = 28,872). In silico bioinformatics analysis and experimental validation in SLE-relevant cell lines were applied to determine the functional consequences of rs34330.ResultsWe replicated a genetic association between SLE and rs34330 (meta-analysis P = 5.29 × 10-22 , odds ratio 0.84 [95% confidence interval 0.81-0.87]). Follow-up bioinformatics and expression quantitative trait locus analysis suggested that rs34330 is located in active chromatin and potentially regulates several target genes. Using luciferase and chromatin immunoprecipitation-real-time quantitative polymerase chain reaction, we demonstrated substantial allele-specific promoter and enhancer activity, and allele-specific binding of 3 histone marks (H3K27ac, H3K4me3, and H3K4me1), RNA polymerase II (Pol II), CCCTC-binding factor, and a critical immune transcription factor (interferon regulatory factor 1 [IRF-1]). Chromosome conformation capture revealed long-range chromatin interactions between rs34330 and the promoters of neighboring genes APOLD1 and DDX47, and effects on CDKN1B and the other target genes were directly validated by clustered regularly interspaced short palindromic repeat (CRISPR)-based genome editing. Finally, CRISPR/dead CRISPR-associated protein 9-based epigenetic activation/silencing confirmed these results. Gene-edited cell lines also showed higher levels of proliferation and apoptosis.ConclusionCollectively, these findings suggest a mechanism whereby the rs34330 risk allele (C) influences the presence of histone marks, RNA Pol II, and IRF-1 transcription factor to regulate expression of several target genes linked to proliferation and apoptosis. This process could potentially underlie the association of rs34330 with SLE.
- Published
- 2021
41. Validation of low‐coverage whole‐genome sequencing for mitochondrial DNA variants suggests mitochondrial DNA as a genetic cause of preterm birth
- Author
-
Kenneth M. Kaufman, Jesse Slone, Yongbo Huang, Louis J. Muglia, Michael W. Pauciulo, Taosheng Huang, Xinjian Wang, Iouri Chepelev, Jack Zhan, Bahram Namjou, John B. Harley, and Zeyu Yang
- Subjects
Genetics ,Whole genome sequencing ,Mitochondrial DNA ,Whole Genome Sequencing ,Mitochondrial disease ,Infant, Newborn ,Infant ,Gestational age ,Biology ,medicine.disease ,DNA, Mitochondrial ,Haplogroup ,Human genetics ,Heteroplasmy ,Mitochondria ,Genome, Mitochondrial ,Cohort ,medicine ,Humans ,Premature Birth ,Genetics (clinical) - Abstract
Preterm birth (PTB), or birth that occurs earlier than 37 weeks of gestational age, is a major contributor to infant mortality and neonatal hospitalization. Mutations in the mitochondrial genome (mtDNA) have been linked to various rare mitochondrial disorders, and may be a contributing factor in PTB given that maternal genetic factors have been strongly linked to PTB. However, to date, no study has found a conclusive connection between a particular mtDNA variant and PTB. Given the high mtDNA copy number per cell, an automated pipeline was developed for detecting mtDNA variants using low-pass whole genome sequencing (lcWGS) data. The pipeline was first validated against samples of known heteroplasmy, and then applied to 929 samples from a PTB cohort from diverse ethnic backgrounds with an average gestational age of 27.18 weeks (range: 21-30). Our new pipeline successfully identified haplogroups and a large number of mtDNA variants in this large PTB cohort, including 8 samples carrying known pathogenic variants and 47 samples carrying rare mtDNA variants. These results confirm that lcWGS can be utilized to reliably identify mtDNA variants. These mtDNA variants may make a contribution toward preterm birth in a small proportion of live births. This article is protected by copyright. All rights reserved.
- Published
- 2021
42. Outcomes of Returning Medically Actionable Genomic Results in Pediatric Research
- Author
-
Amy A. Blumling, Cynthia A. Prows, Margaret H. Harr, Wendy K. Chung, Ellen Wright Clayton, Ingrid A. Holm, Georgia L. Wiesner, John J. Connolly, John B. Harley, Hakon Hakonarson, Michelle L. McGowan, Erin M. Miller, and Melanie F. Myers
- Subjects
genomic screening ,pediatric genomics ,return of genomic results ,healthcare outcomes ,Medicine (miscellaneous) - Abstract
Purpose: The electronic Medical Records and Genomics (eMERGE) Phase III study was undertaken to assess clinical utility of returning medically actionable genomic screening results. We assessed pediatric clinical outcomes following return of pathogenic/likely pathogenic (P/LP) variants in autosomal dominant conditions with available effective interventions. Methods: The two eMERGE III pediatric sites collected outcome data and assessed changes in medical management at 6 and 12 months. Results: We returned P/LP results to 29 participants with outcome data. For 23 of the 29 participants, the P/LP results were previously unknown. Five of the 23 participants were already followed for conditions related to the P/LP variant. Of those receiving novel results and not being followed for the condition related to the P/LP result (n = 18), 14 (77.8%) had a change in healthcare after return of results (RoR). Following RoR, cascade testing of family members occurred for 10 of 23 (43.5%). Conclusions: The most common outcomes post-RoR included imaging/laboratory testing and health behavior recommendations. A change in healthcare was documented in 77.8% of those receiving results by 6 months. Our findings demonstrate how return of genomic screening results impacts healthcare in pediatric populations.
- Published
- 2022
43. Haplotype-specific chromatin looping reveals genetic interactions of regulatory regions modulating gene expression in 8p23.1
- Author
-
Mariana Saint Just Ribeiro, Pulak Tripathi, Bahram Namjou, John B. Harley, and Iouri Chepelev
- Subjects
Genetics ,Molecular Medicine ,Genetics (clinical) - Abstract
A major goal of genetics research is to elucidate mechanisms explaining how genetic variation contributes to phenotypic variation. The genetic variants identified in genome-wide association studies (GWASs) generally explain only a small proportion of heritability of phenotypic traits, the so-called missing heritability problem. Recent evidence suggests that additional common variants beyond lead GWAS variants contribute to phenotypic variation; however, their mechanistic underpinnings generally remain unexplored. Herein, we undertake a study of haplotype-specific mechanisms of gene regulation at 8p23.1 in the human genome, a region associated with a number of complex diseases. The FAM167A-BLK locus in this region has been consistently found in the genome-wide association studies (GWASs) of systemic lupus erythematosus (SLE) in all major ancestries. Our haplotype-specific chromatin interaction (Hi-C) experiments, allele-specific enhancer activity measurements, genetic analyses, and epigenome editing experiments revealed that: 1) haplotype-specific long-range chromatin interactions are prevalent in 8p23.1; 2) BLK promoter and cis-regulatory elements cooperatively interact with haplotype-specificity; 3) genetic variants at distal regulatory elements are allele-specific modifiers of the promoter variants at FAM167A-BLK; 4) the BLK promoter interacts with and, as an enhancer-like promoter, regulates FAM167A expression and 5) local allele-specific enhancer activities are influenced by global haplotype structure due to chromatin looping. Although systemic lupus erythematosus causal variants at the FAM167A-BLK locus are thought to reside in the BLK promoter region, our results reveal that genetic variants at distal regulatory elements modulate promoter activity, changing BLK and FAM167A gene expression and disease risk. Our results suggest that global haplotype-specific 3-dimensional chromatin looping architecture has a strong influence on local allelic BLK and FAM167A gene expression, providing mechanistic details for how regional variants controlling the BLK promoter may influence disease risk.
- Published
- 2022
- Full Text
- View/download PDF
44. Genetic association of primary nonresponse to anti-TNFα therapy in patients with inflammatory bowel disease
- Author
-
Wei-Qi Wei, Erin Smithberger, Jennifer A. Pacheco, Gail P. Jarvik, Madeleine Horvath, James G. Linneman, Tanima De, Loukia Parisiadou, Cristina Alarcon, Adam S. Gordon, Juan Avitia, Scott J. Hebbring, Patrick M. A. Sleiman, Chuyu Chen, Hakon Hakonarson, Sarbani Adhikari, Minoli A. Perera, Sara Kwan, Honghong Zhang, Mary Young, John H. Kwon, John B. Harley, Bianca Lec, and Frank D. Mentch
- Subjects
medicine.medical_specialty ,Necrosis ,Inflammatory bowel disease ,Gastroenterology ,Article ,Pathogenesis ,Cohort Studies ,Internal medicine ,Genotype ,Genetics ,medicine ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,Molecular Biology ,Genetics (clinical) ,Genetic association ,Gene knockdown ,Tumor Necrosis Factor-alpha ,business.industry ,Autophagy ,Odds ratio ,Inflammatory Bowel Diseases ,medicine.disease ,Case-Control Studies ,Immunology ,Expression quantitative trait loci ,Molecular Medicine ,Tumor necrosis factor alpha ,medicine.symptom ,business ,Genome-Wide Association Study - Abstract
OBJECTIVE Primary non-response (PNR) to anti-tumor necrosis factor-α (TNFα) biologics is a serious concern in patients with inflammatory bowel disease (IBD). We aimed to identify the genetic variants associated with PNR. DESIGN Patients were recruited from outpatient GI clinics and PNR was determined using both clinical and endoscopic findings. A case-control genome-wide association study was performed in 589 IBD patients and associations were replicated in an independent cohort of 293 patients. Effect of the associated variant on gene expression and TNFα secretion was assessed by cell-based assays. Pleiotropic effects were investigated by Phenome-wide Association Study (PheWAS). RESULTS We identified rs34767465 as associated with PNR to anti-TNF-α therapy (OR:2.07, 95%CI:1.46-2.94, p=2.43×10−7, [Replication OR:1.8, 95%CI:1.04-3.16, p=0.03]). rs34767465 is a multiple-tissue expression quantitative trait loci for FAM114A2. Using RNA-sequencing and protein quantification from HapMap lymphoblastoid cell lines (LCLs), we found a significant decrease in FAM114A2 mRNA and protein expression in both heterozygous and homozygous genotypes when compared to wild type LCLs. TNF-α secretion was significantly higher in THP-1 cells [differentiated into macrophages] with FAM114A2 knockdown versus controls. Immunoblotting experiments showed that depletion of FAM114A2 impaired autophagy related pathway genes suggesting autophagy mediated TNF-α secretion as a potential mechanism. PheWAS showed rs34767465 was associated with comorbid conditions found in IBD patients (derangement of joints [p=2.3×10−4], pigmentary iris degeneration [p=5.1×10−4], diverticulum of esophagus [p=6.3×10−4]). CONCLUSION We identified a variant rs34767465 associated with PNR to anti-TNFα biologics, which increases TNFα secretion through mechanism related to autophagy. rs34767465 may also explain the comorbidities associated with IBD. Significance of this study What is already known on this subject? Tumor necrosis factor (TNF) α is one of the major pro-inflammatory cytokines in the underlying pathogenesis of mucosal inflammation in inflammatory bowel disease (IBD), and has been the primary therapeutic target for IBD. Anti-TNFα biologic drugs have been shown to induce clinical response in 60-75% of the patients, and long-term maintenance of remission in a large number of patients that respond to the drug. However, approximately 30-40% of the patients fail to respond to anti-TNF α biologics (primary non-responders; PNR). What are the new findings? This is the first genome-wide association study to identify the genetic loci, rs34767465 associated with 2-fold increased risk of PNR to anti-TNFα therapy. rs34767465 is an expressed quantitative trait locus for FAM114A2, and leads to decreased expression of the gene in blood and LCLs. Knockdown of FAM114A2 in THP-1 cells-derived macrophages increase TNFα secretion through mechanism related to autophagy. Phenome-wide association study reveals association of rs34767465 with comorbid conditions like derangement of joints, pigmentary iris degeneration and diverticulum of esophagus, that are observed in IBD patients How might it impact on clinical practice in the foreseeable future? rs34767465 may help guide physicians’ drug choice and therapeutic drug monitoring to prevent PNR to anti-TNFα therapy and IBD associated comorbidities.
- Published
- 2021
45. Meta-analysis of 208370 East Asians identifies 113 susceptibility loci for systemic lupus erythematosus
- Author
-
Yeon-Kyung Lee, Kohei Karino, Dae Jin Park, Xiaoting Chen, Xiaodong Zheng, Dong-Qing Ye, So-Young Bang, Leilei Wen, Tae-Hwan Kim, Takuaki Yamamoto, Yukinori Okada, Young Mo Kang, Sreeja Parameswaran, Xuejun Zhang, Cheng-Xu Li, Eunji Ha, Sang Cheol Bae, Hiroyuki Suetsugu, Koichi Amano, Tsutomu Takeuchi, Takayuki Sumida, Ken Yamaji, Hye-Soon Lee, Yuanjia Tang, Seung-Cheol Shim, Viktoryia Laurynenka, Sen Yang, Wanling Yang, Yujun Sheng, Xianyong Yin, Koichi Matsuda, Keitaro Matsuo, Akari Suzuki, Chang-Hee Suh, Weiran Li, Kwangwoo Kim, Lu Liu, Kyungheon Yoon, Bong-Jo Kim, Mi Yeong Hwang, Takeshi Kuroda, Shruti Eswar, Koichiro Ohmura, Keke Li, Tomoya Miyamura, Shiro Ikegawa, Hanan Salim, Yuta Kochi, Chikashi Terao, Won Tae Chung, Sungsin Jo, Changbing Shen, Kazuhiko Yamamoto, Daisuke Takahashi, Mengwei Wang, Masaya Mukai, Minglong Cai, Matthew T. Weirauch, Nao Otomo, Kazuyoshi Ishigaki, Yuma Sakamoto, Nan Shen, Hiroaki Niiro, Takashi Atsumi, Yong-Fei Wang, Junichi Nakamura, Young-Chang Kwon, Leah C. Kottyan, Yong Cui, John B. Harley, Goro Motomura, Masato Shimizu, Yasushi Kawaguchi, Nobuhiko Sugano, Rui-Xue Leng, Jung-Yoon Choe, Takeshi Miyamoto, Yong Beom Park, Jung-Min Shin, Masaru Koido, G.Y. Ahn, Huihua Ding, Wen-Min Fei, Shin-Seok Lee, Young-Ho Park, He Huang, and Yoshifumi Tada
- Subjects
Male ,0301 basic medicine ,Disease ,polymorphism ,0302 clinical medicine ,Japan ,Polymorphism (computer science) ,Epidemiology ,Prevalence ,Lupus Erythematosus, Systemic ,Immunology and Allergy ,skin and connective tissue diseases ,Genetics ,Asia, Eastern ,Middle Aged ,Meta-analysis ,epidemiology ,Female ,Adult ,China ,medicine.medical_specialty ,Genotype ,Immunology ,Systemic Lupus Erythematosus ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Asian People ,Rheumatology ,Republic of Korea ,medicine ,Humans ,Genetic Predisposition to Disease ,030203 arthritis & rheumatology ,Lupus erythematosus ,business.industry ,Genetic Variation ,Bayes Theorem ,systemic ,Heritability ,medicine.disease ,030104 developmental biology ,Genetic Loci ,Case-Control Studies ,Susceptibility locus ,genetic ,business ,lupus erythematosus ,Genome-Wide Association Study - Abstract
ObjectiveSystemic lupus erythematosus (SLE), an autoimmune disorder, has been associated with nearly 100 susceptibility loci. Nevertheless, these loci only partially explain SLE heritability and their putative causal variants are rarely prioritised, which make challenging to elucidate disease biology. To detect new SLE loci and causal variants, we performed the largest genome-wide meta-analysis for SLE in East Asian populations.MethodsWe newly genotyped 10 029 SLE cases and 180 167 controls and subsequently meta-analysed them jointly with 3348 SLE cases and 14 826 controls from published studies in East Asians. We further applied a Bayesian statistical approach to localise the putative causal variants for SLE associations.ResultsWe identified 113 genetic regions including 46 novel loci at genome-wide significance (p−8). Conditional analysis detected 233 association signals within these loci, which suggest widespread allelic heterogeneity. We detected genome-wide associations at six new missense variants. Bayesian statistical fine-mapping analysis prioritised the putative causal variants to a small set of variants (95% credible set size ≤10) for 28 association signals. We identified 110 putative causal variants with posterior probabilities ≥0.1 for 57 SLE loci, among which we prioritised 10 most likely putative causal variants (posterior probability ≥0.8). Linkage disequilibrium score regression detected genetic correlations for SLE with albumin/globulin ratio (rg=−0.242) and non-albumin protein (rg=0.238).ConclusionThis study reiterates the power of large-scale genome-wide meta-analysis for novel genetic discovery. These findings shed light on genetic and biological understandings of SLE.
- Published
- 2020
46. Lupus enhancer risk variant causes dysregulation of IRF8 through cooperative lncRNA and DNA methylation machinery
- Author
-
Tian Zhou, Xinyi Zhu, Zhizhong Ye, Yong-Fei Wang, Chao Yao, Ning Xu, Mi Zhou, Jianyang Ma, Yuting Qin, Yiwei Shen, Yuanjia Tang, Zhihua Yin, Hong Xu, Yutong Zhang, Xiaoli Zang, Huihua Ding, Wanling Yang, Ya Guo, John B. Harley, Bahram Namjou, Kenneth M. Kaufman, Leah C. Kottyan, Matthew T. Weirauch, Guojun Hou, and Nan Shen
- Subjects
Multidisciplinary ,Proto-Oncogene Proteins ,Interferon Regulatory Factors ,Humans ,General Physics and Astronomy ,RNA, Long Noncoding ,General Chemistry ,DNA Methylation ,Regulatory Sequences, Nucleic Acid ,General Biochemistry, Genetics and Molecular Biology ,Autoimmune Diseases ,Mixed Function Oxygenases - Abstract
Despite strong evidence that human genetic variants affect the expression of many key transcription factors involved in autoimmune diseases, establishing biological links between non-coding risk variants and the gene targets they regulate remains a considerable challenge. Here, we combine genetic, epigenomic, and CRISPR activation approaches to screen for functional variants that regulate IRF8 expression. We demonstrate that the locus containing rs2280381 is a cell-type-specific enhancer for IRF8 that spatially interacts with the IRF8 promoter. Further, rs2280381 mediates IRF8 expression through enhancer RNA AC092723.1, which recruits TET1 to the IRF8 promoter regulating IRF8 expression by affecting methylation levels. The alleles of rs2280381 modulate PU.1 binding and chromatin state to regulate AC092723.1 and IRF8 expression differentially. Our work illustrates an integrative strategy to define functional genetic variants that regulate the expression of critical genes in autoimmune diseases and decipher the mechanisms underlying the dysregulation of IRF8 expression mediated by lupus risk variants.
- Published
- 2022
47. Regulatory Architecture of the RCA Gene Cluster Captures an Intragenic TAD Boundary, CTCF-Mediated Chromatin Looping and a Long-Range Intergenic Enhancer
- Author
-
Joshua S. Clayton, Jessica Cheng, Rhonda L. Taylor, Daniela Ulgiati, José Luis Gómez-Skarmeta, Elizabeth Quail, Rafael D. Acemel, John B. Harley, National Institutes of Health (US), Australian Government, University of Western Australia, and CSIC-JA-UPO - Centro Andaluz de Biología del Desarrollo (CABD)
- Subjects
Complement receptor 2 ,Immunology ,Computational biology ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Gene cluster ,Enhancers ,Immunology and Allergy ,Enhancer ,Gene ,Complement Activation ,030304 developmental biology ,Segmental duplication ,Regulation of gene expression ,0303 health sciences ,B cells ,RCA ,Chromatin ,Complement system ,Enhancer Elements, Genetic ,Gene Expression Regulation ,Multigene Family ,Chromatin looping: TADs ,030215 immunology ,TADs [Chromatin looping] - Abstract
The Regulators of Complement Activation (RCA) gene cluster comprises several tandemly arranged genes with shared functions within the immune system. RCA members, such as complement receptor 2 (CR2), are well-established susceptibility genes in complex autoimmune diseases. Altered expression of RCA genes has been demonstrated at both the functional and genetic level, but the mechanisms underlying their regulation are not fully characterised. We aimed to investigate the structural organisation of the RCA gene cluster to identify key regulatory elements that influence the expression of CR2 and other genes in this immunomodulatory region. Using 4C, we captured extensive CTCF-mediated chromatin looping across the RCA gene cluster in B cells and showed these were organised into two topologically associated domains (TADs). Interestingly, an inter-TAD boundary was located within the CR1 gene at a well-characterised segmental duplication. Additionally, we mapped numerous gene-gene and gene-enhancer interactions across the region, revealing extensive co-regulation. Importantly, we identified an intergenic enhancer and functionally demonstrated this element upregulates two RCA members (CR2 and CD55) in B cells. We have uncovered novel, long-range mechanisms whereby autoimmune disease susceptibility may be influenced by genetic variants, thus highlighting the important contribution of chromatin topology to gene regulation and complex genetic disease., This work was supported by the National Institutes of Health [R01 AI24717 to JH], the Australian Government Research Training Program Scholarship at the University of Western Australia [to JC and JSC], the Spanish Government [BFU2016-74961-P to JG-S] and an institutional grant Unidad de Excelencia María de Maeztu [MDM-206-0687 to the Department of Gene Regulation and Morphogenesis, Centro Andaluz de Biología del Desarrol].
- Published
- 2022
48. Evaluation of the MC4R gene across eMERGE network identifies many unreported obesity-associated variants
- Author
-
John B. Harley, Marc S. Williams, Ozan Dikilitas, Jordan G. Nestor, Joshua C. Denny, Todd Lingren, David J. Carey, Ashley H. Shoemaker, Ian B. Stanaway, Bahram Namjou, Iftikhar J. Kullo, David R. Crosslin, Tooraj Mirshahi, Barbara Benoit, Ning Shang, Xinnan Niu, Rajbir Singh, Frank D. Mentch, Gail P. Jarvik, and Hakon Hakonarson
- Subjects
Adult ,Male ,0301 basic medicine ,Endocrinology, Diabetes and Metabolism ,Medicine (miscellaneous) ,Locus (genetics) ,Genome-wide association study ,Development ,030105 genetics & heredity ,Biology ,Article ,Body Mass Index ,Cohort Studies ,03 medical and health sciences ,Humans ,Obesity ,Allele ,Genotyping ,Aged ,Genetics ,Variant Call Format ,Nutrition and Dietetics ,Haplotype ,Genetic Variation ,Middle Aged ,Penetrance ,030104 developmental biology ,Cohort ,Receptor, Melanocortin, Type 4 ,Female ,Genome-Wide Association Study - Abstract
Background/Objectives Melanocortin-4 receptor (MC4R) plays an essential role in food intake and energy homeostasis. More than 170 MC4R variants have been described over the past two decades, with conflicting reports regarding the prevalence and phenotypic effects of these variants in diverse cohorts. To determine the frequency of MC4R variants in large cohort of different ancestries, we evaluated the MC4R coding region for 20,537 eMERGE participants with sequencing data plus additional 77,454 independent individuals with genome-wide genotyping data at this locus. Subjects/Methods The sequencing data were obtained from the eMERGE phase III study, in which multisample variant call format calls have been generated, curated, and annotated. In addition to penetrance estimation using body mass index (BMI) as a binary outcome, GWAS and PheWAS were performed using median BMI in linear regression analyses. All results were adjusted for principal components, age, sex, and sites of genotyping. Results Targeted sequencing data of MC4R revealed 125 coding variants in 1839 eMERGE participants including 30 unreported coding variants that were predicted to be functionally damaging. Highly penetrant unreported variants included (L325I, E308K, D298N, S270F, F261L, T248A, D111V, and Y80F) in which seven participants had obesity class III defined as BMI ≥ 40 kg/m2. In GWAS analysis, in addition to known risk haplotype upstream of MC4R (best variant rs6567160 (P = 5.36 × 10−25, Beta = 0.37), a novel rare haplotype was detected which was protective against obesity and encompassed the V103I variant with known gain-of-function properties (P = 6.23 × 10−08, Beta = −0.62). PheWAS analyses extended this protective effect of V103I to type 2 diabetes, diabetic nephropathy, and chronic renal failure independent of BMI. Conclusions MC4R screening in a large eMERGE cohort confirmed many previous findings, extend the MC4R pleotropic effects, and discovered additional MC4R rare alleles that probably contribute to obesity.
- Published
- 2020
49. Pleiotropy in the Genetic Predisposition to Rheumatoid Arthritis: A Phenome‐Wide Association Study and Inverse Variance–Weighted Meta‐Analysis
- Author
-
Dan M. Roden, Cecilia P. Chung, Scott J. Hebbring, Mingjian Shi, Nancy J. Cox, Jonathan D. Mosley, Gail P. Jarvik, Eric B. Larson, Ge Liu, Ming T M Lee, C. Michael Stein, eMERGE Investigators, Kenneth M. Kaufman, Vivian K. Kawai, Bahram Namjou, John B. Harley, QiPing Feng, and Adam S. Gordon
- Subjects
Oncology ,medicine.medical_specialty ,Multiple Sclerosis ,Statistics as Topic ,Immunology ,Hyperlipidemias ,Article ,Autoimmune Diseases ,Arthritis, Rheumatoid ,PTPN22 ,Rheumatology ,Pleiotropy ,Internal medicine ,medicine ,Genetic predisposition ,Genetic Pleiotropy ,Humans ,Immunology and Allergy ,Genetic Predisposition to Disease ,Obesity ,Renal Insufficiency, Chronic ,Metabolic Syndrome ,Type 1 diabetes ,Scleroderma, Systemic ,business.industry ,Thyroiditis, Autoimmune ,Mendelian Randomization Analysis ,Odds ratio ,medicine.disease ,Diabetes Mellitus, Type 1 ,Phenotype ,Diabetes Mellitus, Type 2 ,Cardiovascular Diseases ,Meta-analysis ,business - Abstract
Objective This study was undertaken to investigate the hypothesis that a genetic predisposition toward rheumatoid arthritis (RA) increases the risk of 10 cardiometabolic and autoimmune disorders previously associated with RA in epidemiologic studies, and to define new genetic pleiotropy present in RA. Methods Two approaches were used to test our hypothesis. First, we constructed a weighted genetic risk score (wGRS) and then examined its association with 10 prespecified disorders. Additionally, a phenome-wide association study (PheWAS) was carried out to identify potential new associations. Second, inverse variance-weighted regression (IVWR) meta-analysis was used to characterize the association between genetic susceptibility to RA and the prespecified disorders, with the results expressed as odds ratios (ORs) and 95% confidence intervals (95% CIs). Results The wGRS for RA was significantly associated with type 1 diabetes mellitus (DM) (OR 1.10 [95% CI 1.04-1.16]; P = 9.82 × 10-4 ) and multiple sclerosis (OR 0.82 [95% CI 0.77-0.88]; P = 1.73 × 10-8 ), but not with other cardiometabolic phenotypes. In the PheWAS, wGRS was also associated with an increased risk of several autoimmune phenotypes including RA, thyroiditis, and systemic sclerosis, and with a decreased risk of demyelinating disorders. In the IVWR meta-analyses, RA was significantly associated with an increased risk of type 1 DM (P = 1.15 × 10-14 ), with evidence of horizontal pleiotropy (Mendelian Randomization-Egger intercept estimate P = 0.001) likely driven by rs2476601, a PTPN22 variant. The association between type 1 DM and RA remained significant (P = 9.53 × 10-9 ) after excluding rs2476601, with no evidence of horizontal pleiotropy (intercept estimate P = 0.939). RA was also significantly associated with type 2 DM and C-reactive protein levels. These associations were driven by variation in the major histocompatibility complex region. Conclusion This study presents evidence of pleiotropy between the genetic predisposition to RA and associated phenotypes found in other autoimmune and cardiometabolic disorders, including type 1 DM.
- Published
- 2020
50. A Polygenic and Phenotypic Risk Prediction for Polycystic Ovary Syndrome Evaluated by Phenome-Wide Association Studies
- Author
-
Anna O. Basile, Robert J. Carroll, Ky’Era Actkins, Joshua C. Denny, Ian B. Stanaway, Bahram Namjou, Rex L. Chisholm, Yoonjung Yoonie Joo, Kevin Ho, Maureen E. Smith, Jennifer A. Pacheco, Cindy Meun, Tugce Karderi, Margrit Urbanek, Gail P. Jarvik, David R. Crosslin, John B. Harley, Frank D. Mentch, Matthew Jones, Sarah A. Pendergrass, M. Geoffrey Hayes, Abel N. Kho, Felix R. Day, Lea K. Davis, Hakon Hakonarson, Digna R. Velez Edwards, Scott J. Hebbring, Theresa L. Walunas, Marylyn D. Ritchie, Obstetrics & Gynecology, and Internal Medicine
- Subjects
0301 basic medicine ,Multifactorial Inheritance ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Type 2 diabetes ,Bioinformatics ,Biochemistry ,0302 clinical medicine ,Endocrinology ,Risk Factors ,Electronic Health Records ,Phenomics ,Child ,0303 health sciences ,Framingham Risk Score ,Sleep apnea ,Middle Aged ,Prognosis ,Polycystic ovary ,3. Good health ,Phenotype ,Female ,Algorithms ,Polycystic Ovary Syndrome ,medicine.medical_specialty ,Adolescent ,030209 endocrinology & metabolism ,Context (language use) ,Phenome ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,Diabetes mellitus ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Clinical Research Articles ,030304 developmental biology ,Aged ,Genetic association ,business.industry ,Biochemistry (medical) ,medicine.disease ,Comorbidity ,Obesity ,030104 developmental biology ,Case-Control Studies ,business ,Follow-Up Studies ,Genome-Wide Association Study - Abstract
Context As many as 75% of patients with polycystic ovary syndrome (PCOS) are estimated to be unidentified in clinical practice. Objective Utilizing polygenic risk prediction, we aim to identify the phenome-wide comorbidity patterns characteristic of PCOS to improve accurate diagnosis and preventive treatment. Design, Patients, and Methods Leveraging the electronic health records (EHRs) of 124 852 individuals, we developed a PCOS risk prediction algorithm by combining polygenic risk scores (PRS) with PCOS component phenotypes into a polygenic and phenotypic risk score (PPRS). We evaluated its predictive capability across different ancestries and perform a PRS-based phenome-wide association study (PheWAS) to assess the phenomic expression of the heightened risk of PCOS. Results The integrated polygenic prediction improved the average performance (pseudo-R2) for PCOS detection by 0.228 (61.5-fold), 0.224 (58.8-fold), 0.211 (57.0-fold) over the null model across European, African, and multi-ancestry participants respectively. The subsequent PRS-powered PheWAS identified a high level of shared biology between PCOS and a range of metabolic and endocrine outcomes, especially with obesity and diabetes: “morbid obesity”, “type 2 diabetes”, “hypercholesterolemia”, “disorders of lipid metabolism”, “hypertension”, and “sleep apnea” reaching phenome-wide significance. Conclusions Our study has expanded the methodological utility of PRS in patient stratification and risk prediction, especially in a multifactorial condition like PCOS, across different genetic origins. By utilizing the individual genome–phenome data available from the EHR, our approach also demonstrates that polygenic prediction by PRS can provide valuable opportunities to discover the pleiotropic phenomic network associated with PCOS pathogenesis.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.