62 results on '"Sandling JK"'
Search Results
2. S4A:5 High genetic risk score is associated with organ damage in systemic lupus erythematosus
- Author
-
Reid, S, primary, Alexsson, A, additional, Frodlund, M, additional, Svenungsson, E, additional, Sandling, JK, additional, Jönsen, A, additional, Bengtsson, C, additional, Gunnarsson, I, additional, Bengtsson, A, additional, Rantapää-Dahlqvist, S, additional, Syvänen, A-C, additional, Sjöwal, C, additional, Rönnblom, L, additional, and Leonard, D, additional
- Published
- 2018
- Full Text
- View/download PDF
3. 237 Ischaemic stroke in systemic lupus erythematosus, -distribution of subtypes and a risk genotype in the stat4 gene
- Author
-
Svenungsson, E, primary, Hopia, L, additional, Laveskog, A, additional, Jönsen, A, additional, Leonard, D, additional, Gustafsson, JT, additional, Gunnarsson, I, additional, Zickert, A, additional, Nordmark, G, additional, Bengtsson, AA, additional, Elvin, K, additional, Sandling, JK, additional, Syvänen, AC, additional, Rönnblom, L, additional, and Andersson, M, additional
- Published
- 2017
- Full Text
- View/download PDF
4. Meta-analysis in more than 17,900 cases of ischemic stroke reveals a novel association at 12q24.12
- Author
-
Kilarski, LL, Achterberg, S, Devan, WJ, Traylor, M, Malik, R, Lindgren, A, Pare, G, Sharma, P, Slowik, A, Thijs, V, Walters, M, Worrall, BB, Sale, MM, Algra, A, Kappelle, LJ, Wijmenga, C, Norrving, B, Sandling, JK, Roennblom, L, Goris, A, Franke, A, Sudlow, C, Rothwell, PM, Levi, C, Holliday, EG, Fornage, M, Psaty, B, Gretarsdottir, S, Thorsteinsdottir, U, Seshadri, S, Mitchell, BD, Kittner, S, Clarke, R, Hopewell, JC, Bis, JC, Boncoraglio, GB, Meschia, J, Ikram, MA, Hansen, BM, Montaner, J, Thorleifsson, G, Stefanson, K, Rosand, J, de Bakker, PIW, Farrall, M, Dichgans, M, Markus, HS, Bevan, S, Kilarski, LL, Achterberg, S, Devan, WJ, Traylor, M, Malik, R, Lindgren, A, Pare, G, Sharma, P, Slowik, A, Thijs, V, Walters, M, Worrall, BB, Sale, MM, Algra, A, Kappelle, LJ, Wijmenga, C, Norrving, B, Sandling, JK, Roennblom, L, Goris, A, Franke, A, Sudlow, C, Rothwell, PM, Levi, C, Holliday, EG, Fornage, M, Psaty, B, Gretarsdottir, S, Thorsteinsdottir, U, Seshadri, S, Mitchell, BD, Kittner, S, Clarke, R, Hopewell, JC, Bis, JC, Boncoraglio, GB, Meschia, J, Ikram, MA, Hansen, BM, Montaner, J, Thorleifsson, G, Stefanson, K, Rosand, J, de Bakker, PIW, Farrall, M, Dichgans, M, Markus, HS, and Bevan, S
- Abstract
OBJECTIVES: To perform a genome-wide association study (GWAS) using the Immunochip array in 3,420 cases of ischemic stroke and 6,821 controls, followed by a meta-analysis with data from more than 14,000 additional ischemic stroke cases. METHODS: Using the Immunochip, we genotyped 3,420 ischemic stroke cases and 6,821 controls. After imputation we meta-analyzed the results with imputed GWAS data from 3,548 cases and 5,972 controls recruited from the ischemic stroke WTCCC2 study, and with summary statistics from a further 8,480 cases and 56,032 controls in the METASTROKE consortium. A final in silico "look-up" of 2 single nucleotide polymorphisms in 2,522 cases and 1,899 controls was performed. Associations were also examined in 1,088 cases with intracerebral hemorrhage and 1,102 controls. RESULTS: In an overall analysis of 17,970 cases of ischemic stroke and 70,764 controls, we identified a novel association on chromosome 12q24 (rs10744777, odds ratio [OR] 1.10 [1.07-1.13], p = 7.12 × 10(-11)) with ischemic stroke. The association was with all ischemic stroke rather than an individual stroke subtype, with similar effect sizes seen in different stroke subtypes. There was no association with intracerebral hemorrhage (OR 1.03 [0.90-1.17], p = 0.695). CONCLUSION: Our results show, for the first time, a genetic risk locus associated with ischemic stroke as a whole, rather than in a subtype-specific manner. This finding was not associated with intracerebral hemorrhage.
- Published
- 2014
5. Coronary heart disease in systemic lupus erythematosus is associated with interferon regulatory factor-8 gene variants
- Author
-
Leonard, D, Svenungsson, E, Sandling, JK, Berggren, O, Jonsen, A, Bengtsson, Christine, Wang, C, Jensen-Urstad, K, Granstam, S-O, Bengtsson, AA, Gustafsson, JT, Gunnarsson, I, Rantapää-Dahlqvist, Solbritt, Nordmark, G, Eloranta, M-L, Syvanen, A-C, Rönnblom, L, Leonard, D, Svenungsson, E, Sandling, JK, Berggren, O, Jonsen, A, Bengtsson, Christine, Wang, C, Jensen-Urstad, K, Granstam, S-O, Bengtsson, AA, Gustafsson, JT, Gunnarsson, I, Rantapää-Dahlqvist, Solbritt, Nordmark, G, Eloranta, M-L, Syvanen, A-C, and Rönnblom, L
- Published
- 2013
6. Association between Common Variation at the FTO Locus and Changes in Body Mass Index from Infancy to Late Childhood: The Complex Nature of Genetic Association through Growth and Development
- Author
-
Sovio, U, Mook, Dennis, Warrington, NM, Lawrence, R, Briollais, L, Palmer, CNA, Cecil, J, Sandling, JK, Syvanen, AC, Kaakinen, M, Beilin, LJ, Millwood, IY, Bennett, AJ, Laitinen, J, Pouta, A, Molitor, J, Smith, GD, Ben-Shlomo, Y, Jaddoe, Vincent, Palmer, LJ, Pennell, CE, Cole, TJ, McCarthy, MI, Jarvelin, MR, Timpson, NJ, Sovio, U, Mook, Dennis, Warrington, NM, Lawrence, R, Briollais, L, Palmer, CNA, Cecil, J, Sandling, JK, Syvanen, AC, Kaakinen, M, Beilin, LJ, Millwood, IY, Bennett, AJ, Laitinen, J, Pouta, A, Molitor, J, Smith, GD, Ben-Shlomo, Y, Jaddoe, Vincent, Palmer, LJ, Pennell, CE, Cole, TJ, McCarthy, MI, Jarvelin, MR, and Timpson, NJ
- Abstract
An age-dependent association between variation at the FTO locus and BMI in children has been suggested. We meta-analyzed associations between the FTO locus (rs9939609) and BMI in samples, aged from early infancy to 13 years, from 8 cohorts of European ancestry. We found a positive association between additional minor (A) alleles and BMI from 5.5 years onwards, but an inverse association below age 2.5 years. Modelling median BMI curves for each genotype using the LMS method, we found that carriers of minor alleles showed lower BMI in infancy, earlier adiposity rebound (AR), and higher BMI later in childhood. Differences by allele were consistent with two independent processes: earlier AR equivalent to accelerating developmental age by 2.37% (95% CI 1.87, 2.87, p = 10(-20)) per A allele and a positive age by genotype interaction such that BMI increased faster with age (p = 10 223). We also fitted a linear mixed effects model to relate genotype to the BMI curve inflection points adiposity peak (AP) in infancy and AR. Carriage of two minor alleles at rs9939609 was associated with lower BMI at AP (-0.40% (95% CI: -0.74, -0.06), p = 0.02), higher BMI at AR (0.93% (95% CI: 0.22, 1.64), p = 0.01), and earlier AR (-4.72% (-5.81, -3.63), p = 10 217), supporting cross-sectional results. Overall, we confirm the expected association between variation at rs9939609 and BMI in childhood, but only after an inverse association between the same variant and BMI in infancy. Patterns are consistent with a shift on the developmental scale, which is reflected in association with the timing of AR rather than just a global increase in BMI. Results provide important information about longitudinal gene effects and about the role of FTO in adiposity. The associated shifts in developmental timing have clinical importance with respect to known relationships between AR and both later-life BMI and metabolic disease risk.
- Published
- 2011
7. Immunoseq: the identification of functionally relevant variants through targeted capture and sequencing of active regulatory regions in human immune cells
- Author
-
Morin, A, Kwan, T, Ge, B, Letourneau, L, Ban, M, Tandre, K, Caron, M, Sandling, JK, Carlsson, J, Bourque, G, Laprise, C, Montpetit, A, Syvanen, A-C, Ronnblom, L, Sawcer, SJ, Lathrop, MG, and Pastinen, T
- Subjects
gene expression ,rare variants ,immune disease ,next-generation sequencing ,capture ,3. Good health - Abstract
$\textbf{BACKGROUND}$: The observation that the genetic variants identified in genome-wide association studies (GWAS) frequently lie in non-coding regions of the genome that contain cis-regulatory elements suggests that altered gene expression underlies the development of many complex traits. In order to efficiently make a comprehensive assessment of the impact of non-coding genetic variation in immune related diseases we emulated the whole-exome sequencing paradigm and developed a custom capture panel for the known DNase I hypersensitive site (DHS) in immune cells - "Immunoseq". $\textbf{RESULTS}$: We performed Immunoseq in 30 healthy individuals where we had existing transcriptome data from T cells. We identified a large number of novel non-coding variants in these samples. Relying on allele specific expression measurements, we also showed that our selected capture regions are enriched for functional variants that have an impact on differential allelic gene expression. The results from a replication set with 180 samples confirmed our observations. $\textbf{CONCLUSIONS}$: We show that Immunoseq is a powerful approach to detect novel rare variants in regulatory regions. We also demonstrate that these novel variants have a potential functional role in immune cells.
8. Unraveling the Genetics of Shared Clinical and Serological Manifestations in Patients With Systemic Inflammatory Autoimmune Diseases.
- Author
-
Bianchi M, Kozyrev SV, Notarnicola A, Sandling JK, Pettersson M, Leonard D, Sjöwall C, Gunnarsson I, Rantapää-Dahlqvist S, Bengtsson AA, Jönsen A, Svenungsson E, Enocsson H, Kvarnström M, Forsblad-d'Elia H, Bucher SM, Norheim KB, Baecklund E, Jonsson R, Hammenfors D, Eriksson P, Mandl T, Omdal R, Padyukov L, Andersson H, Molberg Ø, Diederichsen LP, Syvänen AC, Wahren-Herlenius M, Nordmark G, Lundberg IE, Rönnblom L, and Lindblad-Toh K
- Abstract
Objective: Systemic inflammatory autoimmune diseases (SIADs) such as systemic lupus erythematosus (SLE), primary Sjögren disease (pSS), and idiopathic inflammatory myopathies (myositis) are complex conditions characterized by shared circulating autoantibodies and clinical manifestations, including skin rashes, among others. This study was aimed at elucidating the genetics underlying these common features., Methods: We performed targeted DNA sequencing of coding and regulatory regions from approximately 1,900 immune-related genes in a large cohort of 2,292 well-characterized Scandinavian patients with SIADs with SLE, pSS, and myositis as well as 1,252 controls. A gene-based functionally weighted genetic score for aggregate testing of all genetic variants, including rare variants, was complemented by in silico functional analyses and in vitro reporter experiments., Results: Case-control association analysis detected known and potentially novel genetic loci in agreement with previous genetic and transcriptomics findings linked to the SIAD autoimmune background. Intriguingly, case-case comparisons between patient subgroups with and without specific autoantibodies revealed that the subgroups defined by antinuclear antibodies and anti-double-stranded DNA antibodies have unique genetic profiles reflecting their heterogeneity. When focusing on clinical features, we overall showed that dual-specificity phosphatase 1 (DUSP1) protective genetic variants lead to increased gene expression and potentially to anti-inflammatory effects on the SIAD-associated skin phenotype. This is consistent with recent genetic findings on eczema and with the previously reported down-regulation of the MAPK signaling-related gene DUSP1 in other skin disorders., Conclusion: Together, this suggests common molecular mechanisms potentially underlying overlapping clinical manifestations shared among different disorders and informs clinical heterogeneity, which could be translated to improve disease diagnostic and treatment, also in more generalized disease frameworks., (© 2024 The Author(s). Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)
- Published
- 2024
- Full Text
- View/download PDF
9. A survey of ficolin-3 activity in Systemic Lupus Erythematosus reveals a link to hematological disease manifestations and autoantibody profile.
- Author
-
Lindelöf L, Rantapää-Dahlqvist S, Lundtoft C, Sandling JK, Leonard D, Sayadi A, Rönnblom L, Enocsson H, Sjöwall C, Jönsen A, Bengtsson AA, Hong MG, Diaz-Gallo LM, Bianchi M, Kozyrev SV, Lindblad-Toh K, Nilsson Ekdahl K, Nilsson B, Gunnarsson I, Svenungsson E, and Eriksson O
- Subjects
- Humans, Antibodies, Antinuclear, Autoantibodies, Complement System Proteins, Ficolins, Lectins genetics, Hematologic Diseases, Lupus Erythematosus, Systemic diagnosis, Lupus Erythematosus, Systemic epidemiology, Lupus Erythematosus, Systemic genetics, Lymphopenia
- Abstract
The complement system plays a central role in the pathogenesis of Systemic Lupus Erythematosus (SLE), but most studies have focused on the classical pathway. Ficolin-3 is the main initiator of the lectin pathway of complement in humans, but its role in systemic autoimmune disease has not been conclusively determined. Here, we combined biochemical and genetic approaches to assess the contribution of ficolin-3 to SLE risk and disease manifestations. Ficolin-3 activity was measured by a functional assay in serum or plasma samples from Swedish SLE patients (n = 786) and controls matched for age and sex (n = 566). Genetic variants in an extended 300 kb genomic region spanning the FCN3 locus were analyzed for their association with ficolin-3 activity and SLE manifestations in a Swedish multicenter cohort (n = 985). Patients with ficolin-3 activity in the highest tertile showed a strong enrichment in an SLE cluster defined by anti-Sm/DNA/nucleosome antibodies (OR 3.0, p < 0.001) and had increased rates of hematological disease (OR 1.4, p = 0.078) and lymphopenia (OR = 1.6, p = 0.039). Genetic variants associated with low ficolin-3 activity mapped to an extended haplotype in high linkage disequilibrium upstream of the FCN3 gene. Patients carrying the lead genetic variant associated with low ficolin-3 activity had a lower frequency of hematological disease (OR 0.67, p = 0.018) and lymphopenia (OR 0.63, p = 0.031) and fewer autoantibodies (p = 0.0019). Loss-of-function variants in the FCN3 gene were not associated with SLE, but four (0.5 %) SLE patients developed acquired ficolin-3 deficiency where ficolin-3 activity in serum was depleted following diagnosis of SLE. Taken together, our results provide genetic and biochemical evidence that implicate the lectin pathway in hematological SLE manifestations. We also identify lectin pathway activation through ficolin-3 as a factor that contributes to the autoantibody response in SLE., Competing Interests: Declaration of competing interest The authors declare no competing financial interests., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. B cell polygenic risk scores associate with anti-dsDNA antibodies and nephritis in systemic lupus erythematosus.
- Author
-
Hedenstedt A, Reid S, Sayadi A, Eloranta ML, Skoglund E, Bolin K, Frodlund M, Lerang K, Jönsen A, Rantapää-Dahlqvist S, Bengtsson AA, Rudin A, Molberg Ø, Sjöwall C, Sandling JK, and Leonard D
- Subjects
- Humans, Female, Antibodies, Antinuclear, Autoantibodies, DNA, HLA-DRB1 Chains, Risk Factors, Lupus Erythematosus, Systemic complications, Lupus Nephritis diagnosis
- Abstract
Objective: B cell function and autoantibodies are important in SLE pathogenesis. In this work, we aimed to investigate the impact of cumulative SLE B cell genetics on SLE subphenotype and autoantibody profile., Methods: Female patients with SLE (n=1248) and healthy controls (n=400) were genotyped using Illumina's Global Screening Array. Two polygenic risk scores (PRSs), one representing B cell genes and the other B cell activation genes, were calculated for each individual using risk loci for SLE in genes assigned to B cell-related pathways according to the Kyoto Encyclopedia of Genes and Genomes, Gene Ontology and Reactome Databases., Results: Double-stranded DNA (dsDNA) antibodies were more prevalent among patients with a high compared with a low SLE B cell PRS (OR 1.47 (1.07 to 2.01), p=0.018), and effect sizes were augmented in patients with human leucocyte antigen (HLA) risk haplotypes HLA-DRB1*03:01 and HLA-DRB1*15:01 (DRB1*03/15 -/- (OR 0.99 (0.56 to 1.77), p=0.98; DRB1*03/15 +/- or -/+ (OR 1.64 (1.06 to 2.54), p=0.028; and DRB1*03/15 +/+ (OR 4.47 (1.21 to 16.47), p=0.024). Further, a high compared with a low B cell PRS was associated with low complement levels in DRB1*03/15 +/+ patients (OR 3.92 (1.22 to 12.64), p=0.022). The prevalence of lupus nephritis (LN) was higher in patients with a B cell activation PRS above the third quartile compared with patients below (OR 1.32 (1.00 to 1.74), p=0.048)., Conclusions: High genetic burden related to B cell function is associated with dsDNA antibody development and LN. Assessing B cell PRSs may be important in order to determine immunological pathways influencing SLE and to predict clinical phenotype., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
11. Distinct HLA associations with autoantibody-defined subgroups in idiopathic inflammatory myopathies.
- Author
-
Leclair V, Galindo-Feria AS, Rothwell S, Kryštůfková O, Zargar SS, Mann H, Diederichsen LP, Andersson H, Klein M, Tansley S, Rönnblom L, Lindblad-Toh K, Syvänen AC, Wahren-Herlenius M, Sandling JK, McHugh N, Lamb JA, Vencovský J, Chinoy H, Holmqvist M, Bianchi M, Padyukov L, Lundberg IE, and Diaz-Gallo LM
- Subjects
- Humans, Alleles, Genetic Predisposition to Disease, Haplotypes, HLA-DRB1 Chains genetics, Phenotype, Autoantibodies genetics, Myositis genetics, Myositis immunology
- Abstract
Background: In patients with idiopathic inflammatory myopathies (IIM), autoantibodies are associated with specific clinical phenotypes suggesting a pathogenic role of adaptive immunity. We explored if autoantibody profiles are associated with specific HLA genetic variants and clinical manifestations in IIM., Methods: We included 1348 IIM patients and determined the occurrence of 14 myositis-specific or -associated autoantibodies. We used unsupervised cluster analysis to identify autoantibody-defined subgroups and logistic regression to estimate associations with clinical manifestations, HLA-DRB1, HLA-DQA1, HLA-DQB1 alleles, and amino acids imputed from genetic information of HLA class II and I molecules., Findings: We identified eight subgroups with the following dominant autoantibodies: anti-Ro52, -U1RNP, -PM/Scl, -Mi2, -Jo1, -Jo1/Ro52, -TIF1γ or negative for all analysed autoantibodies. Associations with HLA-DRB1∗11, HLA-DRB1∗15, HLA-DQA1∗03, and HLA-DQB1∗03 were present in the anti-U1RNP-dominated subgroup. HLA-DRB1∗03, HLA-DQA1∗05, and HLA-DQB1∗02 alleles were overrepresented in the anti-PM/Scl and anti-Jo1/Ro52-dominated subgroups. HLA-DRB1∗16, HLA-DRB1∗07 alleles were most frequent in anti-Mi2 and HLA-DRB1∗01 and HLA-DRB1∗07 alleles in the anti-TIF1γ subgroup. The HLA-DRB1∗13, HLA-DQA1∗01 and HLA-DQB1∗06 alleles were overrepresented in the negative subgroup. Significant signals from variations in class I molecules were detected in the subgroups dominated by anti-Mi2, anti-Jo1/Ro52, anti-TIF1γ, and the negative subgroup., Interpretation: Distinct HLA class II and I associations were observed for almost all autoantibody-defined subgroups. The associations support autoantibody profiles use for classifying IIM which would likely reflect underlying pathogenic mechanisms better than classifications based on clinical symptoms and/or histopathological features., Funding: See a detailed list of funding bodies in the Acknowledgements section at the end of the manuscript., Competing Interests: Declaration of interests Professor Lundberg has received consulting fees from Corbus Pharmaceuticals, Inc and research grants from Astra Zeneca and has been serving on the advisory board for Astra Zeneca, Bristol Myers Squibb, EMD Serono Research & Development Institute, Argenx, Octapharma, Kezaar, Orphazyme, Pfizer and Janssen and has stock shares in Roche and Novartis. Professor Vencovský has received speaker fees from Abbvie, Biogen, Boehringer, Eli Lilly, Gilead, MSD, Novartis, Pfizer, Roche, Sanofi, UCB, Werfen; and has received consulting fees from Abbvie, Argenx, Boehringer, Eli Lilly, Gilead, Octapharma, Pfizer, UCB. Professor Chinoy has received personal compensation for activities with Novartis, UCB, Eli Lilly, Biogen, Orphazyme, Astra Zeneca, Pfizer, Kezar Life Sciences, Galapagos, Argenx, GSK as a speaker, advisory board member or consultancy; grants via The University of Manchester from Eli Lilly and UCB; travel support from Abbvie and Janssen. Dr Mann has received honoraria from Abbvie, Biogen, BMS, Eli Lilly, MSD, Janssen, Novartis, UCB, Pfizer, SOBI and travel support by Abbvie. Dr Tansley received payment from Boehringer Ingelheim as a speaker. Dr Rönnblom received support from The Swedish Research Council (2018–02399), Reumatikerförbundet (R-939716) and GV:80 (FAI-2020-0658) foundation. Dr Holmqvist received support from The Swedish Research Council, Stockholm Regional Council (ALF), Reumatikerförbundet, Konung Gustaf V:80 year foundation, The Swedish Cancer Association and was a member of Medical Advisory Board of The Myositis Association. Dr Lamb received support from the Medical Research Council UK and Myositis UK. Other authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Strong Association of Combined Genetic Deficiencies in the Classical Complement Pathway With Risk of Systemic Lupus Erythematosus and Primary Sjögren's Syndrome.
- Author
-
Lundtoft C, Sjöwall C, Rantapää-Dahlqvist S, Bengtsson AA, Jönsen A, Pucholt P, Wu YL, Lundström E, Eloranta ML, Gunnarsson I, Baecklund E, Jonsson R, Hammenfors D, Forsblad-d'Elia H, Eriksson P, Mandl T, Bucher S, Norheim KB, Auglaend Johnsen SJ, Omdal R, Kvarnström M, Wahren-Herlenius M, Truedsson L, Nilsson B, Kozyrev SV, Bianchi M, Lindblad-Toh K, Yu CY, Nordmark G, Sandling JK, Svenungsson E, Leonard D, and Rönnblom L
- Subjects
- Humans, Complement Pathway, Classical, DNA Copy Number Variations, Complement System Proteins genetics, Hereditary Complement Deficiency Diseases, Complement C4 genetics, Sjogren's Syndrome genetics, Lupus Erythematosus, Systemic
- Abstract
Objective: Complete genetic deficiency of the complement component C2 is a strong risk factor for monogenic systemic lupus erythematosus (SLE), but whether heterozygous C2 deficiency adds to the risk of SLE or primary Sjögren's syndrome (SS) has not been studied systematically. This study was undertaken to investigate potential associations of heterozygous C2 deficiency and C4 copy number variation with clinical manifestations in patients with SLE and patients with primary SS., Methods: The presence of the common 28-bp C2 deletion rs9332736 and C4 copy number variation was examined in Scandinavian patients who had received a diagnosis of SLE (n = 958) or primary SS (n = 911) and in 2,262 healthy controls through the use of DNA sequencing. The concentration of complement proteins in plasma and classical complement function were analyzed in a subgroup of SLE patients., Results: Heterozygous C2 deficiency-when present in combination with a low C4A copy number-substantially increased the risk of SLE (odds ratio [OR] 10.2 [95% confidence interval (95% CI) 3.5-37.0]) and the risk of primary SS (OR 13.0 [95% CI 4.5-48.4]) when compared to individuals with 2 C4A copies and normal C2. For patients heterozygous for rs9332736 with 1 C4A copy, the median age at diagnosis was 7 years earlier in patients with SLE and 12 years earlier in patients with primary SS when compared to patients with normal C2. Reduced C2 levels in plasma (P = 2 × 10
-9 ) and impaired function of the classical complement pathway (P = 0.03) were detected in SLE patients with heterozygous C2 deficiency. Finally, in a primary SS patient homozygous for C2 deficiency, we observed low levels of anti-Scl-70, which suggests a risk of developing systemic sclerosis or potential overlap between primary SS and other systemic autoimmune diseases., Conclusion: We demonstrate that a genetic pattern involving partial deficiencies of C2 and C4A in the classical complement pathway is a strong risk factor for SLE and for primary SS. Our results emphasize the central role of the complement system in the pathogenesis of both SLE and primary SS., (© 2022 The Authors. Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)- Published
- 2022
- Full Text
- View/download PDF
13. Mer-tyrosine kinase: a novel susceptibility gene for SLE related end-stage renal disease.
- Author
-
Yavuz S, Pucholt P, Sandling JK, Bianchi M, Leonard D, Bolin K, Imgenberg-Kreuz J, Eloranta ML, Kozyrev SV, Lanata CM, Jönsen A, Bengtsson AA, Sjöwall C, Svenungsson E, Gunnarsson I, Rantapää-Dahlqvist S, Nititham J, Criswell LA, Lindblad-Toh K, and Rönnblom L
- Subjects
- Humans, Protein-Tyrosine Kinases, c-Mer Tyrosine Kinase genetics, Lupus Erythematosus, Systemic complications, Lupus Erythematosus, Systemic genetics, Lupus Nephritis complications, Lupus Nephritis genetics, Kidney Failure, Chronic genetics
- Abstract
Objective: Lupus nephritis (LN) is a common and severe manifestation of SLE. The genetic risk for nephritis and progression to end-stage renal disease (ESRD) in patients with LN remains unclear. Herein, we aimed to identify novel genetic associations with LN, focusing on subphenotypes and ESRD., Methods: We analysed genomic data on 958 patients with SLE (discovery cohort: LN=338) with targeted sequencing data from 1832 immunological pathway genes. We used an independent multiethnic cohort comprising 1226 patients with SLE (LN=603) as a replication dataset. Detailed functional annotation and functional epigenomic enrichment analyses were applied to predict functional effects of the candidate variants., Results: A genetic variant (rs56097910) within the MERTK gene was associated with ESRD in both cohorts, meta-analysis OR=5.4 (2.8 to 10.6); p=1.0×10
-6 . We observed decreased methylation levels in peripheral blood cells from SLE patients with ESRD, compared with patients without renal SLE (p=2.7×10-4 ), at one CpG site (cg16333401) in close vicinity to the transcription start site of MERTK and located in a DNAse hypersensitivity region in T and B cells. Rs56097910 is linked to altered MERTK expression in kidney tissue in public eQTL databases. Two loci were replicated for association with proliferative LN: PRDM1 (rs6924535, pmeta =1.6×10-5 , OR=0.58) and APOA1BP ( NAXE ) (rs942960, pmeta =1.2×10-5 , OR=2.64)., Conclusion: We identified a novel genetic risk locus, MERTK , associated with SLE-ESRD using the data from two large SLE cohorts. Through DNA methylation analysis and functional annotation, we showed that the risk could be mediated through regulation of gene expression. Our results suggest that variants in the MERTK gene are important for the risk of developing SLE-ESRD and suggest a role for PRDM1 and APOA1BP in proliferative LN., Competing Interests: Competing interests: PP is now an employee of Olink Proteomics., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ.)- Published
- 2022
- Full Text
- View/download PDF
14. Complement C4 Copy Number Variation is Linked to SSA/Ro and SSB/La Autoantibodies in Systemic Inflammatory Autoimmune Diseases.
- Author
-
Lundtoft C, Pucholt P, Martin M, Bianchi M, Lundström E, Eloranta ML, Sandling JK, Sjöwall C, Jönsen A, Gunnarsson I, Rantapää-Dahlqvist S, Bengtsson AA, Leonard D, Baecklund E, Jonsson R, Hammenfors D, Forsblad-d'Elia H, Eriksson P, Mandl T, Magnusson Bucher S, Norheim KB, Auglaend Johnsen SJ, Omdal R, Kvarnström M, Wahren-Herlenius M, Notarnicola A, Andersson H, Molberg Ø, Diederichsen LP, Almlöf J, Syvänen AC, Kozyrev SV, Lindblad-Toh K, Nilsson B, Blom AM, Lundberg IE, Nordmark G, Diaz-Gallo LM, Svenungsson E, and Rönnblom L
- Subjects
- Autoantibodies genetics, Complement C4 genetics, Complement C4b genetics, DNA Copy Number Variations, Humans, Risk Factors, Lupus Erythematosus, Systemic genetics, Myositis
- Abstract
Objective: Copy number variation of the C4 complement components, C4A and C4B, has been associated with systemic inflammatory autoimmune diseases. This study was undertaken to investigate whether C4 copy number variation is connected to the autoimmune repertoire in systemic lupus erythematosus (SLE), primary Sjögren's syndrome (SS), or myositis., Methods: Using targeted DNA sequencing, we determined the copy number and genetic variants of C4 in 2,290 well-characterized Scandinavian patients with SLE, primary SS, or myositis and 1,251 healthy controls., Results: A prominent relationship was observed between C4A copy number and the presence of SSA/SSB autoantibodies, which was shared between the 3 diseases. The strongest association was detected in patients with autoantibodies against both SSA and SSB and 0 C4A copies when compared to healthy controls (odds ratio [OR] 18.0 [95% confidence interval (95% CI) 10.2-33.3]), whereas a weaker association was seen in patients without SSA/SSB autoantibodies (OR 3.1 [95% CI 1.7-5.5]). The copy number of C4 correlated positively with C4 plasma levels. Further, a common loss-of-function variant in C4A leading to reduced plasma C4 was more prevalent in SLE patients with a low copy number of C4A. Functionally, we showed that absence of C4A reduced the individuals' capacity to deposit C4b on immune complexes., Conclusion: We show that a low C4A copy number is more strongly associated with the autoantibody repertoire than with the clinically defined disease entities. These findings may have implications for understanding the etiopathogenetic mechanisms of systemic inflammatory autoimmune diseases and for patient stratification when taking the genetic profile into account., (© 2022 The Authors. Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)
- Published
- 2022
- Full Text
- View/download PDF
15. Contribution of Rare Genetic Variation to Disease Susceptibility in a Large Scandinavian Myositis Cohort.
- Author
-
Bianchi M, Kozyrev SV, Notarnicola A, Hultin Rosenberg L, Karlsson Å, Pucholt P, Rothwell S, Alexsson A, Sandling JK, Andersson H, Cooper RG, Padyukov L, Tjärnlund A, Dastmalchi M, Meadows JRS, Pyndt Diederichsen L, Molberg Ø, Chinoy H, Lamb JA, Rönnblom L, Lindblad-Toh K, and Lundberg IE
- Subjects
- Case-Control Studies, Cohort Studies, Female, Humans, Male, Scandinavian and Nordic Countries, Genetic Predisposition to Disease, Genetic Variation, Myositis genetics
- Abstract
Objective: Idiopathic inflammatory myopathies (IIMs) are a heterogeneous group of complex autoimmune conditions characterized by inflammation in skeletal muscle and extramuscular compartments, and interferon (IFN) system activation. We undertook this study to examine the contribution of genetic variation to disease susceptibility and to identify novel avenues for research in IIMs., Methods: Targeted DNA sequencing was used to mine coding and potentially regulatory single nucleotide variants from ~1,900 immune-related genes in a Scandinavian case-control cohort of 454 IIM patients and 1,024 healthy controls. Gene-based aggregate testing, together with rare variant- and gene-level enrichment analyses, was implemented to explore genotype-phenotype relations., Results: Gene-based aggregate tests of all variants, including rare variants, identified IFI35 as a potential genetic risk locus for IIMs, suggesting a genetic signature of type I IFN pathway activation. Functional annotation of the IFI35 locus highlighted a regulatory network linked to the skeletal muscle-specific gene PTGES3L, as a potential candidate for IIM pathogenesis. Aggregate genetic associations with AGER and PSMB8 in the major histocompatibility complex locus were detected in the antisynthetase syndrome subgroup, which also showed a less marked genetic signature of the type I IFN pathway. Enrichment analyses indicated a burden of synonymous and noncoding rare variants in IIM patients, suggesting increased disease predisposition associated with these classes of rare variants., Conclusion: Our study suggests the contribution of rare genetic variation to disease susceptibility in IIM and specific patient subgroups, and pinpoints genetic associations consistent with previous findings by gene expression profiling. These features highlight genetic profiles that are potentially relevant to disease pathogenesis., (© 2021 The Authors. Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)
- Published
- 2022
- Full Text
- View/download PDF
16. Interaction between the STAT4 rs11889341(T) risk allele and smoking confers increased risk of myocardial infarction and nephritis in patients with systemic lupus erythematosus.
- Author
-
Reid S, Hagberg N, Sandling JK, Alexsson A, Pucholt P, Sjöwall C, Lerang K, Jönsen A, Gunnarsson I, Syvänen AC, Troldborg AM, Voss A, Bengtsson AA, Molberg Ø, Jacobsen S, Svenungsson E, Rönnblom L, and Leonard D
- Subjects
- Adult, Aged, Female, Humans, Lupus Nephritis epidemiology, Male, Middle Aged, Myocardial Infarction epidemiology, Polymorphism, Single Nucleotide, Gene-Environment Interaction, Interleukin-12 Subunit p35 genetics, Lupus Erythematosus, Systemic epidemiology, Lupus Nephritis genetics, Myocardial Infarction genetics, STAT4 Transcription Factor genetics, Smoking epidemiology
- Abstract
Objective: To investigate how genetics influence the risk of smoking-related systemic lupus erythematosus (SLE) manifestations., Methods: Patients with SLE (n
discovery cohort =776, nreplication cohort =836) were genotyped using the 200K Immunochip single nucleotide polymorphisms (SNP) Array (Illumina) and a custom array. Sixty SNPs with SLE association (p<5.0×10-8 ) were analysed. Signal transducer and activator of transcription 4 (STAT4) activation was assessed in in vitro stimulated peripheral blood mononuclear cells from healthy controls (n=45)., Results: In the discovery cohort, smoking was associated with myocardial infarction (MI) (OR 1.96 (95% CI 1.09 to 3.55)), with a greater effect in patients carrying any rs11889341 STAT4 risk allele (OR 2.72 (95% CI 1.24 to 6.00)) or two risk alleles (OR 8.27 (95% CI 1.48 to 46.27)).Smokers carrying the risk allele also displayed an increased risk of nephritis (OR 1.47 (95% CI 1.06 to 2.03)). In the replication cohort, the high risk of MI in smokers carrying the risk allele and the association between the STAT4 risk allele and nephritis in smokers were confirmed (OR 6.19 (95% CI 1.29 to 29.79) and 1.84 (95% CI 1.05 to 3.29), respectively).The interaction between smoking and the STAT4 risk allele resulted in further increase in the risk of MI (OR 2.14 (95% CI 1.01 to 4.62)) and nephritis (OR 1.53 (95% CI 1.08 to 2.17)), with 54% (MI) and 34% (nephritis) of the risk attributable to the interaction. Levels of interleukin-12-induced phosphorylation of STAT4 in CD8+ T cells were higher in smokers than in non-smokers (mean geometric fluorescence intensity 1063 vs 565, p=0.0063).Lastly, the IL12A rs564799 risk allele displayed association with MI in both cohorts (OR 1.53 (95% CI 1.01 to 2.31) and 2.15 (95% CI 1.08 to 4.26), respectively)., Conclusions: Smoking in the presence of the STAT4 risk gene variant appears to increase the risk of MI and nephritis in SLE. Our results also highlight the role of the IL12-STAT4 pathway in SLE-cardiovascular morbidity., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
17. DNA Methylation-Based Interferon Scores Associate With Sub-Phenotypes in Primary Sjögren's Syndrome.
- Author
-
Imgenberg-Kreuz J, Sandling JK, Norheim KB, Johnsen SJA, Omdal R, Syvänen AC, Svenungsson E, Rönnblom L, Eloranta ML, and Nordmark G
- Subjects
- Female, Humans, Lymphoma immunology, Male, Middle Aged, Phenotype, RNA, Messenger immunology, DNA Methylation immunology, Interferon Type I immunology, Sjogren's Syndrome immunology
- Abstract
Primary Sjögren's syndrome (pSS) is an autoimmune inflammatory disease with profound clinical heterogeneity, where excessive activation of the type I interferon (IFN) system is considered one of the key mechanisms in disease pathogenesis. Here we present a DNA methylation-based IFN system activation score (DNAm IFN score) and investigate its potential associations with sub-phenotypes of pSS. The study comprised 100 Swedish patients with pSS and 587 Swedish controls. For replication, 48 patients with pSS from Stavanger, Norway, were included. IFN scores were calculated from DNA methylation levels at the IFN-induced genes RSAD2, IFIT1 and IFI44L. A high DNAm IFN score, defined as > mean
controls +2SDcontrols (IFN score >4.4), was observed in 59% of pSS patients and in 4% of controls (p=1.3x10-35 ). Patients with a high DNAm IFN score were on average seven years younger at symptom onset (p=0.017) and at diagnosis (p=3x10-3 ). The DNAm IFN score levels were significantly higher in pSS positive for both SSA and SSB antibodies compared to SSA/SSB negative patients (pdiscovery =1.9x10-8 , preplication =7.8x10-4 ). In patients positive for both SSA subtypes Ro52 and Ro60, an increased score was identified compared to single positive patients (p=0.022). Analyzing the discovery and replication cohorts together, elevated DNAm IFN scores were observed in pSS with hypergammaglobulinemia (p=2x10-8 ) and low C4 (p=1.5x10-3 ) compared to patients without these manifestations. Patients < 70 years with ongoing lymphoma at DNA sampling or lymphoma at follow-up (n=7), presented an increased DNAm IFN score compared to pSS without lymphoma (p=0.025). In conclusion, the DNAm-based IFN score is a promising alternative to mRNA-based scores for identification of patients with activation of the IFN system and may be applied for patient stratification guiding treatment decisions, monitoring and inclusion in clinical trials., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Imgenberg-Kreuz, Sandling, Norheim, Johnsen, Omdal, Syvänen, Svenungsson, Rönnblom, Eloranta and Nordmark.)- Published
- 2021
- Full Text
- View/download PDF
18. Variants in BANK1 are associated with lupus nephritis of European ancestry.
- Author
-
Bolin K, Imgenberg-Kreuz J, Leonard D, Sandling JK, Alexsson A, Pucholt P, Haarhaus ML, Almlöf JC, Nititham J, Jönsen A, Sjöwall C, Bengtsson AA, Rantapää-Dahlqvist S, Svenungsson E, Gunnarsson I, Syvänen AC, Lerang K, Troldborg A, Voss A, Molberg Ø, Jacobsen S, Criswell L, Rönnblom L, and Nordmark G
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Cohort Studies, DNA Methylation, Gene Expression Regulation, Genotype, Humans, Membrane Proteins genetics, Lupus Erythematosus, Systemic, Lupus Nephritis genetics
- Abstract
The genetic background of lupus nephritis (LN) has not been completely elucidated. We performed a case-only study of 2886 SLE patients, including 947 (33%) with LN. Renal biopsies were available from 396 patients. The discovery cohort (Sweden, n = 1091) and replication cohort 1 (US, n = 962) were genotyped on the Immunochip and replication cohort 2 (Denmark/Norway, n = 833) on a custom array. Patients with LN, proliferative nephritis, or LN with end-stage renal disease were compared with SLE without nephritis. Six loci were associated with LN (p < 1 × 10
-4 , NFKBIA, CACNA1S, ITGA1, BANK1, OR2Y, and ACER3) in the discovery cohort. Variants in BANK1 showed the strongest association with LN in replication cohort 1 (p = 9.5 × 10-4 ) and proliferative nephritis in a meta-analysis of discovery and replication cohort 1. There was a weak association between BANK1 and LN in replication cohort 2 (p = 0.052), and in the meta-analysis of all three cohorts the association was strengthened (p = 2.2 × 10-7 ). DNA methylation data in 180 LN patients demonstrated methylation quantitative trait loci (meQTL) effects between a CpG site and BANK1 variants. To conclude, we describe genetic variations in BANK1 associated with LN and evidence for genetic regulation of DNA methylation within the BANK1 locus. This indicates a role for BANK1 in LN pathogenesis., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
19. Toll-like receptors revisited; a possible role for TLR1 in lupus nephritis.
- Author
-
Yavuz S, Bianchi M, Kozyrev S, Bolin K, Leonard D, Pucholt P, Sandling JK, Bengtsson A, Jönsen A, Rantapää-Dahlqvist S, Sjöwall C, Svenungsson E, Gunnarsson I, Lindblad-Toh K, and Rönnblom L
- Subjects
- Genetic Predisposition to Disease, Humans, Toll-Like Receptor 1, Toll-Like Receptors, Lupus Erythematosus, Systemic, Lupus Nephritis
- Abstract
Competing Interests: Competing interests: None declared.
- Published
- 2021
- Full Text
- View/download PDF
20. Genetic and clinical basis for two distinct subtypes of primary Sjögren's syndrome.
- Author
-
Thorlacius GE, Hultin-Rosenberg L, Sandling JK, Bianchi M, Imgenberg-Kreuz J, Pucholt P, Theander E, Kvarnström M, Forsblad-d'Elia H, Bucher SM, Norheim KB, Johnsen SJA, Hammenfors D, Skarstein K, Jonsson MV, Baecklund E, Aqrawi LA, Jensen JL, Palm Ø, Morris AP, Meadows JRS, Rantapää-Dahlqvist S, Mandl T, Eriksson P, Lind L, Omdal R, Jonsson R, Lindblad-Toh K, Rönnblom L, Wahren-Herlenius M, and Nordmark G
- Subjects
- Age of Onset, Autoimmunity genetics, Correlation of Data, Female, Genetic Markers genetics, Genetic Predisposition to Disease, Genetic Variation, Humans, Male, Middle Aged, Norway epidemiology, Polymorphism, Single Nucleotide, Sequence Analysis, DNA methods, Sweden epidemiology, Autoantibodies blood, HLA-DQ alpha-Chains genetics, Sjogren's Syndrome classification, Sjogren's Syndrome genetics, Sjogren's Syndrome immunology, Sjogren's Syndrome physiopathology
- Abstract
Objectives: Clinical presentation of primary Sjögren's syndrome (pSS) varies considerably. A shortage of evidence-based objective markers hinders efficient drug development and most clinical trials have failed to reach primary endpoints., Methods: We performed a multicentre study to identify patient subgroups based on clinical, immunological and genetic features. Targeted DNA sequencing of 1853 autoimmune-related loci was performed. After quality control, 918 patients with pSS, 1264 controls and 107 045 single nucleotide variants remained for analysis. Replication was performed in 177 patients with pSS and 7672 controls., Results: We found strong signals of association with pSS in the HLA region. Principal component analysis of clinical data distinguished two patient subgroups defined by the presence of SSA/SSB antibodies. We observed an unprecedented high risk of pSS for an association in the HLA-DQA1 locus of odds ratio 6.10 (95% CI: 4.93, 7.54, P=2.2×10-62) in the SSA/SSB-positive subgroup, while absent in the antibody negative group. Three independent signals within the MHC were observed. The two most significant variants in MHC class I and II respectively, identified patients with a higher risk of hypergammaglobulinaemia, leukopenia, anaemia, purpura, major salivary gland swelling and lymphadenopathy. Replication confirmed the association with both MHC class I and II signals confined to SSA/SSB antibody positive pSS., Conclusion: Two subgroups of patients with pSS with distinct clinical manifestations can be defined by the presence or absence of SSA/SSB antibodies and genetic markers in the HLA locus. These subgroups should be considered in clinical follow-up, drug development and trial outcomes, for the benefit of both subgroups., (© The Author(s) 2020. Published by Oxford University Press on behalf of the British Society for Rheumatology.)
- Published
- 2021
- Full Text
- View/download PDF
21. Contributions of de novo variants to systemic lupus erythematosus.
- Author
-
Almlöf JC, Nystedt S, Mechtidou A, Leonard D, Eloranta ML, Grosso G, Sjöwall C, Bengtsson AA, Jönsen A, Gunnarsson I, Svenungsson E, Rönnblom L, Sandling JK, and Syvänen AC
- Subjects
- Adolescent, Adult, Child, Female, Humans, Male, Middle Aged, Mutation, Missense, Promoter Regions, Genetic, Gene Frequency, Lupus Erythematosus, Systemic genetics, Polymorphism, Single Nucleotide
- Abstract
By performing whole-genome sequencing in a Swedish cohort of 71 parent-offspring trios, in which the child in each family is affected by systemic lupus erythematosus (SLE, OMIM 152700), we investigated the contribution of de novo variants to risk of SLE. We found de novo single nucleotide variants (SNVs) to be significantly enriched in gene promoters in SLE patients compared with healthy controls at a level corresponding to 26 de novo promoter SNVs more in each patient than expected. We identified 12 de novo SNVs in promoter regions of genes that have been previously implicated in SLE, or that have functions that could be of relevance to SLE. Furthermore, we detected three missense de novo SNVs, five de novo insertion-deletions, and three de novo structural variants with potential to affect the expression of genes that are relevant for SLE. Based on enrichment analysis, disease-affecting de novo SNVs are expected to occur in one-third of SLE patients. This study shows that de novo variants in promoters commonly contribute to the genetic risk of SLE. The fact that de novo SNVs in SLE were enriched to promoter regions highlights the importance of using whole-genome sequencing for identification of de novo variants.
- Published
- 2021
- Full Text
- View/download PDF
22. Molecular pathways in patients with systemic lupus erythematosus revealed by gene-centred DNA sequencing.
- Author
-
Sandling JK, Pucholt P, Hultin Rosenberg L, Farias FHG, Kozyrev SV, Eloranta ML, Alexsson A, Bianchi M, Padyukov L, Bengtsson C, Jonsson R, Omdal R, Lie BA, Massarenti L, Steffensen R, Jakobsen MA, Lillevang ST, Lerang K, Molberg Ø, Voss A, Troldborg A, Jacobsen S, Syvänen AC, Jönsen A, Gunnarsson I, Svenungsson E, Rantapää-Dahlqvist S, Bengtsson AA, Sjöwall C, Leonard D, Lindblad-Toh K, and Rönnblom L
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Blood Coagulation genetics, Case-Control Studies, Cluster Analysis, Complement Activation genetics, Female, Humans, Janus Kinases genetics, Lupus Erythematosus, Systemic immunology, Male, Middle Aged, Multifactorial Inheritance, Polymorphism, Single Nucleotide, STAT Transcription Factors genetics, Sequence Analysis, DNA, Signal Transduction genetics, Sweden, White People, Young Adult, Antigen Presentation genetics, Immunity, Innate genetics, Interferon Type I immunology, Lupus Erythematosus, Systemic genetics, Lymphopoiesis genetics, T-Lymphocytes immunology
- Abstract
Objectives: Systemic lupus erythematosus (SLE) is an autoimmune disease with extensive heterogeneity in disease presentation between patients, which is likely due to an underlying molecular diversity. Here, we aimed at elucidating the genetic aetiology of SLE from the immunity pathway level to the single variant level, and stratify patients with SLE into distinguishable molecular subgroups, which could inform treatment choices in SLE., Methods: We undertook a pathway-centred approach, using sequencing of immunological pathway genes. Altogether 1832 candidate genes were analysed in 958 Swedish patients with SLE and 1026 healthy individuals. Aggregate and single variant association testing was performed, and we generated pathway polygenic risk scores (PRS)., Results: We identified two main independent pathways involved in SLE susceptibility: T lymphocyte differentiation and innate immunity, characterised by HLA and interferon, respectively. Pathway PRS defined pathways in individual patients, who on average were positive for seven pathways. We found that SLE organ damage was more pronounced in patients positive for the T or B cell receptor signalling pathways. Further, pathway PRS-based clustering allowed stratification of patients into four groups with different risk score profiles. Studying sets of genes with priors for involvement in SLE, we observed an aggregate common variant contribution to SLE at genes previously reported for monogenic SLE as well as at interferonopathy genes., Conclusions: Our results show that pathway risk scores have the potential to stratify patients with SLE beyond clinical manifestations into molecular subsets, which may have implications for clinical follow-up and therapy selection., Competing Interests: Competing interests: An AstraZeneca research collaboration grant to LR supported the study. LHR is now an employee of Olink Proteomics., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
23. Function of multiple sclerosis-protective HLA class I alleles revealed by genome-wide protein-quantitative trait loci mapping of interferon signalling.
- Author
-
Lundtoft C, Pucholt P, Imgenberg-Kreuz J, Carlsson-Almlöf J, Eloranta ML, Syvänen AC, Nordmark G, Sandling JK, Kockum I, Olsson T, Rönnblom L, and Hagberg N
- Subjects
- Adult, Aged, Aged, 80 and over, Alleles, B-Lymphocytes immunology, B-Lymphocytes pathology, Female, Flow Cytometry, HLA-A2 Antigen immunology, Humans, Interferon Type I genetics, Interferon Type I immunology, Interferon-gamma genetics, Interferon-gamma immunology, Killer Cells, Natural immunology, Male, Middle Aged, Multiple Sclerosis epidemiology, Multiple Sclerosis immunology, Multiple Sclerosis pathology, Polymorphism, Single Nucleotide genetics, Receptor, Interferon alpha-beta genetics, Receptor, Interferon alpha-beta immunology, Receptors, Interferon genetics, Receptors, Interferon immunology, T-Lymphocytes immunology, T-Lymphocytes pathology, Genetic Predisposition to Disease, HLA-A2 Antigen genetics, Multiple Sclerosis genetics, Quantitative Trait Loci genetics
- Abstract
Interferons (IFNs) are cytokines that are central to the host defence against viruses and other microorganisms. If not properly regulated, IFNs may contribute to the pathogenesis of inflammatory autoimmune, or infectious diseases. To identify genetic polymorphisms regulating the IFN system we performed an unbiased genome-wide protein-quantitative trait loci (pQTL) mapping of cell-type specific type I and type II IFN receptor levels and their responses in immune cells from 303 healthy individuals. Seven genome-wide significant (p < 5.0E-8) pQTLs were identified. Two independent SNPs that tagged the multiple sclerosis (MS)-protective HLA class I alleles A*02/A*68 and B*44, respectively, were associated with increased levels of IFNAR2 in B and T cells, with the most prominent effect in IgD-CD27+ memory B cells. The increased IFNAR2 levels in B cells were replicated in cells from an independent set of healthy individuals and in MS patients. Despite increased IFNAR2 levels, B and T cells carrying the MS-protective alleles displayed a reduced response to type I IFN stimulation. Expression and methylation-QTL analysis demonstrated increased mRNA expression of the pseudogene HLA-J in B cells carrying the MS-protective class I alleles, possibly driven via methylation-dependent transcriptional regulation. Together these data suggest that the MS-protective effects of HLA class I alleles are unrelated to their antigen-presenting function, and propose a previously unappreciated function of type I IFN signalling in B and T cells in MS immune-pathogenesis., Competing Interests: I have read the journal's policy and the authors of this manuscript have the following competing interests: Tomas Olsson has received unrestricted MS research grants, and/or advisory board/lecture honoraria from AstraZeneca, Biogen, Novartis, Merck, Sanofi and Roche, none of which has been related to this work.
- Published
- 2020
- Full Text
- View/download PDF
24. High genetic risk score is associated with early disease onset, damage accrual and decreased survival in systemic lupus erythematosus.
- Author
-
Reid S, Alexsson A, Frodlund M, Morris D, Sandling JK, Bolin K, Svenungsson E, Jönsen A, Bengtsson C, Gunnarsson I, Illescas Rodriguez V, Bengtsson A, Arve S, Rantapää-Dahlqvist S, Eloranta ML, Syvänen AC, Sjöwall C, Vyse TJ, Rönnblom L, and Leonard D
- Subjects
- Adult, Antibodies, Anticardiolipin blood, Case-Control Studies, Female, Genotype, Humans, Kidney Failure, Chronic genetics, Kidney Failure, Chronic mortality, Lupus Coagulation Inhibitor blood, Lupus Erythematosus, Systemic blood, Lupus Erythematosus, Systemic mortality, Lupus Nephritis mortality, Male, Middle Aged, Prevalence, Risk, Risk Factors, Survival Rate, beta 2-Glycoprotein I immunology, Genetic Predisposition to Disease epidemiology, Lupus Erythematosus, Systemic genetics, Lupus Nephritis genetics, Risk Assessment statistics & numerical data, Severity of Illness Index
- Abstract
Objectives: To investigate associations between a high genetic disease risk and disease severity in patients with systemic lupus erythematosus (SLE)., Methods: Patients with SLE (n=1001, discovery cohort and n=5524, replication cohort) and healthy controls (n=2802 and n=9859) were genotyped using a 200K Immunochip single nucleotide polymorphism array. A genetic risk score (GRS) was assigned to each individual based on 57 SLE risk loci., Results: SLE was more prevalent in the high, compared with the low, GRS-quartile (OR 12.32 (9.53 to 15.71), p=7.9×10
-86 and OR 7.48 (6.73 to 8.32), p=2.2×10-304 for the discovery and the replication cohorts, respectively). In the discovery cohort, patients in the high GRS-quartile had a 6-year earlier mean disease onset (HR 1.47 (1.22 to 1.75), p=4.3×10-5 ), displayed higher prevalence of damage accrual (OR 1.47 (1.06 to 2.04), p=2.0×10-2 ), renal disorder (OR 2.22 (1.50 to 3.27), p=5.9×10-5 ), anti-dsDNA (OR 1.83 (1.19 to 2.81), p=6.1×10-3 ), end-stage renal disease (ESRD) (OR 5.58 (1.50 to 20.79), p=1.0×10-2 ), proliferative nephritis (OR 2.42 (1.30 to 4.49), p=5.1×10-3 ), anti-cardiolipin-IgG (OR 1.89 (1.13 to 3.18), p=1.6×10-2 ), anti-β2 -glycoprotein-I-IgG (OR 2.29 (1.29 to 4.06), p=4.8×10-3 ) and positive lupus anticoagulant test (OR 2.12 (1.16 to 3.89), p=1.5×10-2 ) compared with patients in the low GRS-quartile. Survival analysis showed earlier onset of the first organ damage (HR 1.51 (1.04 to 2.25), p=3.7×10-2 ), first cardiovascular event (HR 1.65 (1.03 to 2.64), p=2.6×10-2 ), nephritis (HR 2.53 (1.72 to 3.71), p=9.6×10-7 ), ESRD (HR 6.78 (1.78 to 26.86), p=6.5×10-3 ) and decreased overall survival (HR 1.83 (1.02 to 3.30), p=4.3×10-2 ) in high to low quartile comparison., Conclusions: A high GRS is associated with increased risk of organ damage, renal dysfunction and all-cause mortality. Our results indicate that genetic profiling may be useful for predicting outcomes in patients with SLE., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY. Published by BMJ.)- Published
- 2020
- Full Text
- View/download PDF
25. Genetic variations in A20 DUB domain provide a genetic link to citrullination and neutrophil extracellular traps in systemic lupus erythematosus.
- Author
-
Odqvist L, Jevnikar Z, Riise R, Öberg L, Rhedin M, Leonard D, Yrlid L, Jackson S, Mattsson J, Nanda S, Cohen P, Knebel A, Arthur S, Thörn K, Svenungsson E, Jönsen A, Gunnarsson I, Tandre K, Alexsson A, Kastbom A, Rantapää-Dahlqvist S, Eloranta ML, Syvänen AC, Bengtsson A, Johansson P, Sandling JK, Sjöwall C, Rönnblom L, Collins B, and Vaarala O
- Subjects
- Animals, Autoantibodies blood, Autoantibodies immunology, Epitopes immunology, Genetic Predisposition to Disease genetics, Humans, Lupus Erythematosus, Systemic blood, Mice, NF-kappa B metabolism, Neutrophils metabolism, Polymorphism, Genetic, Protein-Arginine Deiminase Type 4 metabolism, Up-Regulation genetics, Citrullination genetics, Endopeptidases genetics, Extracellular Traps genetics, Lupus Erythematosus, Systemic genetics, Tumor Necrosis Factor alpha-Induced Protein 3 genetics
- Abstract
Objectives: Genetic variations in TNFAIP3 (A20) de-ubiquitinase (DUB) domain increase the risk of systemic lupus erythematosus (SLE) and rheumatoid arthritis. A20 is a negative regulator of NF-κB but the role of its DUB domain and related genetic variants remain unclear. We aimed to study the functional effects of A20 DUB-domain alterations in immune cells and understand its link to SLE pathogenesis., Methods: CRISPR/Cas9 was used to generate human U937 monocytes with A20 DUB-inactivating C103A knock-in (KI) mutation. Whole genome RNA-sequencing was used to identify differentially expressed genes between WT and C103A KI cells. Functional studies were performed in A20 C103A U937 cells and in immune cells from A20 C103A mice and genotyped healthy individuals with A20 DUB polymorphism rs2230926. Neutrophil extracellular trap (NET) formation was addressed ex vivo in neutrophils from A20 C103A mice and SLE-patients with rs2230926., Results: Genetic disruption of A20 DUB domain in human and murine myeloid cells did not give rise to enhanced NF-κB signalling. Instead, cells with C103A mutation or rs2230926 polymorphism presented an upregulated expression of PADI4 , an enzyme regulating protein citrullination and NET formation, two key mechanisms in autoimmune pathology. A20 C103A cells exhibited enhanced protein citrullination and extracellular trap formation, which could be suppressed by selective PAD4 inhibition. Moreover, SLE-patients with rs2230926 showed increased NETs and increased frequency of autoantibodies to citrullinated epitopes., Conclusions: We propose that genetic alterations disrupting the A20 DUB domain mediate increased susceptibility to SLE through the upregulation of PADI4 with resultant protein citrullination and extracellular trap formation., Competing Interests: Competing interests: LO, ZR, RR, LÖ, MR, LFY, SJ, JM, KT, PJ, BC and OV were employees at AstraZeneca Group while performing this study and may have stock/stock options in AstraZeneca. AB received research grant from DISSECT, partly funded by AstraZeneca. LR reports grants and personal fees from Astra Zeneca during the conduct of the study and personal fees from Biogen outside the submitted work. AstraZeneca provided funding to DISSECT for the conduct of this study. The remaining authors declare that they have no competing interests. There are no patents involved., (© Author(s) (or their employer(s)) 2019. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2019
- Full Text
- View/download PDF
26. Shared and Unique Patterns of DNA Methylation in Systemic Lupus Erythematosus and Primary Sjögren's Syndrome.
- Author
-
Imgenberg-Kreuz J, Almlöf JC, Leonard D, Sjöwall C, Syvänen AC, Rönnblom L, Sandling JK, and Nordmark G
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, CpG Islands immunology, DNA Methylation immunology, Genome, Human, Lupus Erythematosus, Systemic genetics, Lupus Erythematosus, Systemic immunology, Sjogren's Syndrome genetics, Sjogren's Syndrome immunology
- Abstract
Objectives: To perform a cross-comparative analysis of DNA methylation in patients with systemic lupus erythematosus (SLE), patients with primary Sjögren's syndrome (pSS), and healthy controls addressing the question of epigenetic sharing and aiming to detect disease-specific alterations. Methods: DNA extracted from peripheral blood from 347 cases with SLE, 100 cases with pSS, and 400 healthy controls were analyzed on the Human Methylation 450k array, targeting 485,000 CpG sites across the genome. A linear regression model including age, sex, and blood cell type distribution as covariates was fitted, and association p -values were Bonferroni corrected. A random forest machine learning classifier was designed for prediction of disease status based on DNA methylation data. Results: We established a combined set of 4,945 shared differentially methylated CpG sites (DMCs) in SLE and pSS compared to controls. In pSS, hypomethylation at type I interferon induced genes was mainly driven by patients who were positive for Ro/SSA and/or La/SSB autoantibodies. Analysis of differential methylation between SLE and pSS identified 2,244 DMCs with a majority of sites showing decreased methylation in SLE compared to pSS. The random forest classifier demonstrated good performance in discerning between disease status with an area under the curve (AUC) between 0.83 and 0.96. Conclusions: The majority of differential DNA methylation is shared between SLE and pSS, however, important quantitative differences exist. Our data highlight neutrophil dysregulation as a shared mechanism, emphasizing the role of neutrophils in the pathogenesis of systemic autoimmune diseases. The current study provides evidence for genes and molecular pathways driving common and disease-specific pathogenic mechanisms.
- Published
- 2019
- Full Text
- View/download PDF
27. Circulating Levels of Interferon Regulatory Factor-5 Associates With Subgroups of Systemic Lupus Erythematosus Patients.
- Author
-
Idborg H, Zandian A, Ossipova E, Wigren E, Preger C, Mobarrez F, Checa A, Sohrabian A, Pucholt P, Sandling JK, Fernandes-Cerqueira C, Rönnelid J, Oke V, Grosso G, Kvarnström M, Larsson A, Wheelock CE, Syvänen AC, Rönnblom L, Kultima K, Persson H, Gräslund S, Gunnarsson I, Nilsson P, Svenungsson E, and Jakobsson PJ
- Subjects
- Adult, Cluster Analysis, Cross-Sectional Studies, Female, Humans, Male, Middle Aged, Phenotype, Proteomics, Proto-Oncogene Proteins c-ets metabolism, Biomarkers blood, Interferon Regulatory Factors blood, Lupus Erythematosus, Systemic immunology, Organic Cation Transporter 2 blood, S100A12 Protein blood
- Abstract
Systemic Lupus Erythematosus (SLE) is a heterogeneous autoimmune disease, which currently lacks specific diagnostic biomarkers. The diversity within the patients obstructs clinical trials but may also reflect differences in underlying pathogenesis. Our objective was to obtain protein profiles to identify potential general biomarkers of SLE and to determine molecular subgroups within SLE for patient stratification. Plasma samples from a cross-sectional study of well-characterized SLE patients ( n = 379) and matched population controls ( n = 316) were analyzed by antibody suspension bead array targeting 281 proteins. To investigate the differences between SLE and controls, Mann-Whitney U -test with Bonferroni correction, generalized linear modeling and receiver operating characteristics (ROC) analysis were performed. K-means clustering was used to identify molecular SLE subgroups. We identified Interferon regulating factor 5 (IRF5), solute carrier family 22 member 2 (SLC22A2) and S100 calcium binding protein A12 (S100A12) as the three proteins with the largest fold change between SLE patients and controls (SLE/Control = 1.4, 1.4, and 1.2 respectively). The lowest p -values comparing SLE patients and controls were obtained for S100A12, Matrix metalloproteinase-1 (MMP1) and SLC22A2 (p
adjusted = 3 × 10-9 , 3 × 10-6 , and 5 × 10-6 respectively). In a set of 15 potential biomarkers differentiating SLE patients and controls, two of the proteins were transcription factors, i.e., IRF5 and SAM pointed domain containing ETS transcription factor (SPDEF). IRF5 was up-regulated while SPDEF was found to be down-regulated in SLE patients. Unsupervised clustering of all investigated proteins identified three molecular subgroups among SLE patients, characterized by (1) high levels of rheumatoid factor-IgM, (2) low IRF5, and (3) high IRF5. IRF5 expressing microparticles were analyzed by flow cytometry in a subset of patients to confirm the presence of IRF5 in plasma and detection of extracellular IRF5 was further confirmed by immunoprecipitation-mass spectrometry (IP-MS). Interestingly IRF5, a known genetic risk factor for SLE, was detected extracellularly and suggested by unsupervised clustering analysis to differentiate between SLE subgroups. Our results imply a set of circulating molecules as markers of possible pathogenic importance in SLE. We believe that these findings could be of relevance for understanding the pathogenesis and diversity of SLE, as well as for selection of patients in clinical trials.- Published
- 2019
- Full Text
- View/download PDF
28. A rare regulatory variant in the MEF2D gene affects gene regulation and splicing and is associated with a SLE sub-phenotype in Swedish cohorts.
- Author
-
Farias FHG, Dahlqvist J, Kozyrev SV, Leonard D, Wilbe M, Abramov SN, Alexsson A, Pielberg GR, Hansson-Hamlin H, Andersson G, Tandre K, Bengtsson AA, Sjöwall C, Svenungsson E, Gunnarsson I, Rantapää-Dahlqvist S, Syvänen AC, Sandling JK, Eloranta ML, Rönnblom L, and Lindblad-Toh K
- Subjects
- Adolescent, Adult, Aged, Child, Female, HEK293 Cells, Humans, MEF2 Transcription Factors genetics, MEF2 Transcription Factors metabolism, Male, Middle Aged, Polymorphism, Single Nucleotide, Protein Binding, Lupus Erythematosus, Systemic genetics, Phenotype, RNA Splicing
- Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder with heterogeneous clinical presentation and complex etiology involving the interplay between genetic, epigenetic, environmental and hormonal factors. Many common SNPs identified by genome wide-association studies (GWAS) explain only a small part of the disease heritability suggesting the contribution from rare genetic variants, undetectable in GWAS, and complex epistatic interactions. Using targeted re-sequencing of coding and conserved regulatory regions within and around 215 candidate genes selected on the basis of their known role in autoimmunity and genes associated with canine immune-mediated diseases, we identified a rare regulatory variant rs200395694:G > T located in intron 4 of the MEF2D gene encoding the myocyte-specific enhancer factor 2D transcription factor and associated with SLE in Swedish cohorts (504 SLE patients and 839 healthy controls, p = 0.014, CI = 1.1-10). Fisher's exact test revealed an association between the genetic variant and a triad of disease manifestations including Raynaud, anti-U1-ribonucleoprotein (anti-RNP), and anti-Smith (anti-Sm) antibodies (p = 0.00037) among the patients. The DNA-binding activity of the allele was further studied by EMSA, reporter assays, and minigenes. The region has properties of an active cell-specific enhancer, differentially affected by the alleles of rs200395694:G > T. In addition, the risk allele exerts an inhibitory effect on the splicing of the alternative tissue-specific isoform, and thus may modify the target gene set regulated by this isoform. These findings emphasize the potential of dissecting traits of complex diseases and correlating them with rare risk alleles with strong biological effects.
- Published
- 2019
- Full Text
- View/download PDF
29. Whole-genome sequencing identifies complex contributions to genetic risk by variants in genes causing monogenic systemic lupus erythematosus.
- Author
-
Almlöf JC, Nystedt S, Leonard D, Eloranta ML, Grosso G, Sjöwall C, Bengtsson AA, Jönsen A, Gunnarsson I, Svenungsson E, Rönnblom L, Sandling JK, and Syvänen AC
- Subjects
- Female, Humans, Male, Middle Aged, Predictive Value of Tests, Risk Factors, Genome, Human, Heterozygote, High-Throughput Nucleotide Sequencing, Lupus Erythematosus, Systemic genetics, Models, Genetic, Mutation, Missense
- Abstract
Systemic lupus erythematosus (SLE, OMIM 152700) is a systemic autoimmune disease with a complex etiology. The mode of inheritance of the genetic risk beyond familial SLE cases is currently unknown. Additionally, the contribution of heterozygous variants in genes known to cause monogenic SLE is not fully understood. Whole-genome sequencing of DNA samples from 71 Swedish patients with SLE and their healthy biological parents was performed to investigate the general genetic risk of SLE using known SLE GWAS risk loci identified using the ImmunoChip, variants in genes associated to monogenic SLE, and the mode of inheritance of SLE risk alleles in these families. A random forest model for predicting genetic risk for SLE showed that the SLE risk variants were mainly inherited from one of the parents. In the 71 patients, we detected a significant enrichment of ultra-rare ( ≤ 0.1%) missense and nonsense mutations in 22 genes known to cause monogenic forms of SLE. We identified one previously reported homozygous nonsense mutation in the C1QC (Complement C1q C Chain) gene, which explains the immunodeficiency and severe SLE phenotype of that patient. We also identified seven ultra-rare, coding heterozygous variants in five genes (C1S, DNASE1L3, DNASE1, IFIH1, and RNASEH2A) involved in monogenic SLE. Our findings indicate a complex contribution to the overall genetic risk of SLE by rare variants in genes associated with monogenic forms of SLE. The rare variants were inherited from the other parent than the one who passed on the more common risk variants leading to an increased genetic burden for SLE in the child. Higher frequency SLE risk variants are mostly passed from one of the parents to the offspring affected with SLE. In contrast, the other parent, in seven cases, contributed heterozygous rare variants in genes associated with monogenic forms of SLE, suggesting a larger impact of rare variants in SLE than hitherto reported.
- Published
- 2019
- Full Text
- View/download PDF
30. Epigenetic alterations in primary Sjögren's syndrome - an overview.
- Author
-
Imgenberg-Kreuz J, Sandling JK, and Nordmark G
- Subjects
- DNA Methylation, Histone Code, Humans, Inflammation, RNA, Untranslated, Sjogren's Syndrome immunology, Epigenesis, Genetic, Sjogren's Syndrome genetics
- Abstract
Primary Sjögren's syndrome (pSS) is a chronic autoimmune rheumatic disease characterized by inflammation of exocrine glands, mainly salivary and lacrimal glands. In addition, pSS may affect multiple other organs resulting in systemic manifestations. Although the precise etiology of pSS remains elusive, pSS is considered to be a multi-factorial disease, where underlying genetic predisposition, environmental factors and epigenetic mechanisms contribute to disease development. Epigenetic mechanisms, such as DNA methylation, histone modifications and non-coding RNAs, may constitute a dynamic link between genome, environment and phenotypic manifestation by their modulating effects on gene expression. A growing body of studies reporting altered epigenetic landscapes in pSS suggests that epigenetic mechanisms play a role in the pathogenesis of pSS, and the reversible nature of epigenetic modifications suggests therapeutic strategies targeting epigenetic dysregulation in pSS. This article reviews our current understanding of epigenetic mechanisms in pSS and discusses implications for novel diagnostic and therapeutic approaches., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
31. Cytokine production by activated plasmacytoid dendritic cells and natural killer cells is suppressed by an IRAK4 inhibitor.
- Author
-
Hjorton K, Hagberg N, Israelsson E, Jinton L, Berggren O, Sandling JK, Thörn K, Mo J, Eloranta ML, and Rönnblom L
- Subjects
- Adult, Aged, Aged, 80 and over, Cytokines antagonists & inhibitors, Cytokines immunology, Dendritic Cells drug effects, Dendritic Cells immunology, Female, Humans, Interleukin-1 Receptor-Associated Kinases antagonists & inhibitors, Interleukin-1 Receptor-Associated Kinases immunology, Killer Cells, Natural drug effects, Killer Cells, Natural immunology, Lupus Erythematosus, Systemic immunology, Lupus Erythematosus, Systemic metabolism, Male, Middle Aged, Antigen-Antibody Complex pharmacology, Cytokines metabolism, Dendritic Cells metabolism, Interleukin-1 Receptor-Associated Kinases metabolism, Killer Cells, Natural metabolism
- Abstract
Background: In systemic lupus erythematosus (SLE), immune complexes (ICs) containing self-derived nucleic acids trigger the synthesis of proinflammatory cytokines by immune cells. We asked how an interleukin (IL)-1 receptor-associated kinase 4 small molecule inhibitor (IRAK4i) affects RNA-IC-induced cytokine production compared with hydroxychloroquine (HCQ)., Methods: Plasmacytoid dendritic cells (pDCs) and natural killer (NK) cells were isolated from peripheral blood mononuclear cells (PBMCs) of healthy individuals. PBMCs from SLE patients and healthy individuals were depleted of monocytes. Cells were stimulated with RNA-containing IC (RNA-IC) in the presence or absence of IRAK4i I92 or HCQ, and cytokines were measured by immunoassay or flow cytometry. Transcriptome sequencing was performed on RNA-IC-stimulated pDCs from healthy individuals to assess the effect of IRAK4i and HCQ., Results: In healthy individuals, RNA-IC induced interferon (IFN)-α, tumor necrosis factor (TNF)-α, IL-6, IL-8, IFN-γ, macrophage inflammatory protein (MIP)1-α, and MIP1-β production in pDC and NK cell cocultures. IFN-α production was selective for pDCs, whereas both pDCs and NK cells produced TNF-α. IRAK4i reduced the pDC and NK cell-derived cytokine production by 74-95%. HCQ interfered with cytokine production in pDCs but not in NK cells. In monocyte-depleted PBMCs, IRAK4i blocked cytokine production more efficiently than HCQ. Following RNA-IC activation of pDCs, 975 differentially expressed genes were observed (false discovery rate (FDR) < 0.05), with many connected to cytokine pathways, cell regulation, and apoptosis. IRAK4i altered the expression of a larger number of RNA-IC-induced genes than did HCQ (492 versus 65 genes)., Conclusions: The IRAK4i I92 exhibits a broader inhibitory effect than HCQ on proinflammatory pathways triggered by RNA-IC, suggesting IRAK4 inhibition as a therapeutic option in SLE.
- Published
- 2018
- Full Text
- View/download PDF
32. Novel gene variants associated with cardiovascular disease in systemic lupus erythematosus and rheumatoid arthritis.
- Author
-
Leonard D, Svenungsson E, Dahlqvist J, Alexsson A, Ärlestig L, Taylor KE, Sandling JK, Bengtsson C, Frodlund M, Jönsen A, Eketjäll S, Jensen-Urstad K, Gunnarsson I, Sjöwall C, Bengtsson AA, Eloranta ML, Syvänen AC, Rantapää-Dahlqvist S, Criswell LA, and Rönnblom L
- Subjects
- Age Distribution, Alleles, Arthritis, Rheumatoid epidemiology, Arthritis, Rheumatoid physiopathology, Cardiovascular Diseases epidemiology, Cardiovascular Diseases physiopathology, Case-Control Studies, Comorbidity, Female, Gene Frequency, Genetic Variation, Humans, Incidence, Interleukins, Lupus Erythematosus, Systemic epidemiology, Lupus Erythematosus, Systemic physiopathology, Male, Prognosis, Reference Values, Severity of Illness Index, Sex Distribution, Signal Recognition Particle genetics, Arthritis, Rheumatoid genetics, Cardiovascular Diseases genetics, Genetic Predisposition to Disease epidemiology, Lupus Erythematosus, Systemic genetics, Polymorphism, Single Nucleotide genetics
- Abstract
Objectives: Patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) have increased risk of cardiovascular disease (CVD). We investigated whether single nucleotide polymorphisms (SNPs) at autoimmunity risk loci were associated with CVD in SLE and RA., Methods: Patients with SLE (n=1045) were genotyped using the 200K Immunochip SNP array (Illumina). The allele frequency was compared between patients with and without different manifestations of CVD. Results were replicated in a second SLE cohort (n=1043) and in an RA cohort (n=824). We analysed publicly available genetic data from general population, performed electrophoretic mobility shift assays and measured cytokine levels and occurrence of antiphospholipid antibodies (aPLs)., Results: We identified two new putative risk loci associated with increased risk for CVD in two SLE populations, which remained after adjustment for traditional CVD risk factors. An IL19 risk allele, rs17581834(T) was associated with stroke/myocardial infarction (MI) in SLE (OR 2.3 (1.5 to 3.4), P=8.5×10
-5 ) and RA (OR 2.8 (1.4 to 5.6), P=3.8×10-3 ), meta-analysis (OR 2.5 (2.0 to 2.9), P=3.5×10-7 ), but not in population controls. The IL19 risk allele affected protein binding, and SLE patients with the risk allele had increased levels of plasma-IL10 (P=0.004) and aPL (P=0.01). An SRP54-AS1 risk allele, rs799454(G) was associated with stroke/transient ischaemic attack in SLE (OR 1.7 (1.3 to 2.2), P=2.5×10-5 ) but not in RA. The SRP54-AS1 risk allele is an expression quantitative trait locus for four genes., Conclusions: The IL19 risk allele was associated with stroke/MI in SLE and RA, but not in the general population, indicating that shared immune pathways may be involved in the CVD pathogenesis in inflammatory rheumatic diseases., Competing Interests: Competing interests: None declared., (© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2018. All rights reserved. No commercial use is permitted unless otherwise expressly granted.)- Published
- 2018
- Full Text
- View/download PDF
33. DNA methylation mapping identifies gene regulatory effects in patients with systemic lupus erythematosus.
- Author
-
Imgenberg-Kreuz J, Carlsson Almlöf J, Leonard D, Alexsson A, Nordmark G, Eloranta ML, Rantapää-Dahlqvist S, Bengtsson AA, Jönsen A, Padyukov L, Gunnarsson I, Svenungsson E, Sjöwall C, Rönnblom L, Syvänen AC, and Sandling JK
- Subjects
- Adult, Case-Control Studies, Chromosome Mapping, CpG Islands genetics, Female, Genetic Predisposition to Disease, Genome-Wide Association Study, Genotype, Humans, Interferon Regulatory Factor-1 genetics, Lupus Erythematosus, Systemic blood, Male, Middle Aged, Polymorphism, Single Nucleotide, Quantitative Trait Loci, DNA Methylation genetics, Gene Expression Regulation genetics, Lupus Erythematosus, Systemic genetics
- Abstract
Objectives: Systemic lupus erythematosus (SLE) is a chronic autoimmune condition with heterogeneous presentation and complex aetiology where DNA methylation changes are emerging as a contributing factor. In order to discover novel epigenetic associations and investigate their relationship to genetic risk for SLE, we analysed DNA methylation profiles in a large collection of patients with SLE and healthy individuals., Methods: DNA extracted from blood from 548 patients with SLE and 587 healthy controls were analysed on the Illumina HumanMethylation 450 k BeadChip, which targets 485 000 CpG sites across the genome. Single nucleotide polymorphism (SNP) genotype data for 196 524 SNPs on the Illumina ImmunoChip from the same individuals were utilised for methylation quantitative trait loci ( cis -meQTLs) analyses., Results: We identified and replicated differentially methylated CpGs (DMCs) in SLE at 7245 CpG sites in the genome. The largest methylation differences were observed at type I interferon-regulated genes which exhibited decreased methylation in SLE. We mapped cis -meQTLs and identified genetic regulation of methylation levels at 466 of the DMCs in SLE. The meQTLs for DMCs in SLE were enriched for genetic association to SLE, and included seven SLE genome-wide association study (GWAS) loci: PTPRC (CD45), MHC-class III , UHRF1BP1 , IRF5 , IRF7 , IKZF3 and UBE2L3 . In addition, we observed association between genotype and variance of methylation at 20 DMCs in SLE, including at the HLA-DQB2 locus., Conclusions: Our results suggest that several of the genetic risk variants for SLE may exert their influence on the phenotype through alteration of DNA methylation levels at regulatory regions of target genes., Competing Interests: Competing interests: None declared., (© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2018. All rights reserved. No commercial use is permitted unless otherwise expressly granted.)
- Published
- 2018
- Full Text
- View/download PDF
34. Transcription profiling of peripheral B cells in antibody-positive primary Sjögren's syndrome reveals upregulated expression of CX3CR1 and a type I and type II interferon signature.
- Author
-
Imgenberg-Kreuz J, Sandling JK, Björk A, Nordlund J, Kvarnström M, Eloranta ML, Rönnblom L, Wahren-Herlenius M, Syvänen AC, and Nordmark G
- Subjects
- Adult, Aged, Antigens, CD19 metabolism, Autoantibodies immunology, Autoantigens immunology, Chemokine CCL5 metabolism, Female, Gene Expression Profiling, Gene Expression Regulation immunology, Humans, Middle Aged, RNA, Small Cytoplasmic immunology, Receptors, CCR1 metabolism, Ribonucleoproteins immunology, Signal Transduction immunology, Transcriptional Activation immunology, Transcriptome genetics, B-Lymphocytes immunology, CX3C Chemokine Receptor 1 metabolism, Interferon Type I immunology, Interferon-gamma immunology, OX40 Ligand metabolism, Sjogren's Syndrome immunology
- Abstract
B cells play a key role in the pathogenesis of primary Sjögren's syndrome (pSS). The aim of this study was to analyse the transcriptome of CD19+ B cells from patients with pSS and healthy controls to decipher the B cell-specific contribution to pSS. RNA from purified CD19+ B cells from 12 anti-SSA antibody-positive untreated female patients with pSS and 20 healthy blood donors was subjected to whole transcriptome sequencing. A false discovery rate corrected significance threshold of α < 0.05 was applied to define differential gene expression. As validation, gene expression in B cells from 17 patients with pSS and 16 healthy controls was analysed using a targeted gene panel. RNA-sequencing identified 4047 differentially expressed autosomal genes in pSS B cells. Upregulated expression of type I and type II interferon (IFN)-induced genes was observed, establishing an IFN signature in pSS B cells. Among the top upregulated and validated genes were CX3CR1, encoding the fractalkine receptor involved in regulation of B-cell malignancies, CCL5/RANTES and CCR1. Increased expression of several members of the TNF superfamily was also identified; TNFSF4/Ox40L, TNFSF10/TRAIL, TNFSF13B/BAFF, TNFRSF17/BCMA as well as S100A8 and -A9/calprotectin, TLR7, STAT1 and STAT2. Among genes with downregulated expression in pSS B cells were SOCS1 and SOCS3, CD70 and TNFAIP3/A20. We conclude that B cells from patients with anti-SSA antibody-positive pSS display immune activation with upregulated expression of chemokines, chemokine receptors and a prominent type I and type II IFN signature, while suppressors of cytokine signalling are downregulated. This adds insight into the autoimmune process and suggests potential targets for future functional studies., (© 2018 The Authors. Scandinavian Journal of Immunology published by John Wiley & Sons Ltd on behalf of The Foundation for the Scandinavian Journal of Immunology.)
- Published
- 2018
- Full Text
- View/download PDF
35. Novel risk genes for systemic lupus erythematosus predicted by random forest classification.
- Author
-
Almlöf JC, Alexsson A, Imgenberg-Kreuz J, Sylwan L, Bäcklin C, Leonard D, Nordmark G, Tandre K, Eloranta ML, Padyukov L, Bengtsson C, Jönsen A, Dahlqvist SR, Sjöwall C, Bengtsson AA, Gunnarsson I, Svenungsson E, Rönnblom L, Sandling JK, and Syvänen AC
- Subjects
- Adult, B-Lymphocytes immunology, B-Lymphocytes metabolism, Case-Control Studies, Female, Gene Expression Profiling, Gene Expression Regulation, Genome-Wide Association Study, Humans, Lupus Erythematosus, Systemic immunology, Lupus Erythematosus, Systemic pathology, Male, T-Lymphocytes immunology, T-Lymphocytes metabolism, Autoimmunity genetics, Biomarkers analysis, CDC2 Protein Kinase genetics, Kruppel-Like Transcription Factors genetics, Lupus Erythematosus, Systemic genetics, Nerve Growth Factors genetics, Polymorphism, Single Nucleotide
- Abstract
Genome-wide association studies have identified risk loci for SLE, but a large proportion of the genetic contribution to SLE still remains unexplained. To detect novel risk genes, and to predict an individual's SLE risk we designed a random forest classifier using SNP genotype data generated on the "Immunochip" from 1,160 patients with SLE and 2,711 controls. Using gene importance scores defined by the random forest classifier, we identified 15 potential novel risk genes for SLE. Of them 12 are associated with other autoimmune diseases than SLE, whereas three genes (ZNF804A, CDK1, and MANF) have not previously been associated with autoimmunity. Random forest classification also allowed prediction of patients at risk for lupus nephritis with an area under the curve of 0.94. By allele-specific gene expression analysis we detected cis-regulatory SNPs that affect the expression levels of six of the top 40 genes designed by the random forest analysis, indicating a regulatory role for the identified risk variants. The 40 top genes from the prediction were overrepresented for differential expression in B and T cells according to RNA-sequencing of samples from five healthy donors, with more frequent over-expression in B cells compared to T cells.
- Published
- 2017
- Full Text
- View/download PDF
36. Transancestral mapping and genetic load in systemic lupus erythematosus.
- Author
-
Langefeld CD, Ainsworth HC, Cunninghame Graham DS, Kelly JA, Comeau ME, Marion MC, Howard TD, Ramos PS, Croker JA, Morris DL, Sandling JK, Almlöf JC, Acevedo-Vásquez EM, Alarcón GS, Babini AM, Baca V, Bengtsson AA, Berbotto GA, Bijl M, Brown EE, Brunner HI, Cardiel MH, Catoggio L, Cervera R, Cucho-Venegas JM, Dahlqvist SR, D'Alfonso S, Da Silva BM, de la Rúa Figueroa I, Doria A, Edberg JC, Endreffy E, Esquivel-Valerio JA, Fortin PR, Freedman BI, Frostegård J, García MA, de la Torre IG, Gilkeson GS, Gladman DD, Gunnarsson I, Guthridge JM, Huggins JL, James JA, Kallenberg CGM, Kamen DL, Karp DR, Kaufman KM, Kottyan LC, Kovács L, Laustrup H, Lauwerys BR, Li QZ, Maradiaga-Ceceña MA, Martín J, McCune JM, McWilliams DR, Merrill JT, Miranda P, Moctezuma JF, Nath SK, Niewold TB, Orozco L, Ortego-Centeno N, Petri M, Pineau CA, Pons-Estel BA, Pope J, Raj P, Ramsey-Goldman R, Reveille JD, Russell LP, Sabio JM, Aguilar-Salinas CA, Scherbarth HR, Scorza R, Seldin MF, Sjöwall C, Svenungsson E, Thompson SD, Toloza SMA, Truedsson L, Tusié-Luna T, Vasconcelos C, Vilá LM, Wallace DJ, Weisman MH, Wither JE, Bhangale T, Oksenberg JR, Rioux JD, Gregersen PK, Syvänen AC, Rönnblom L, Criswell LA, Jacob CO, Sivils KL, Tsao BP, Schanberg LE, Behrens TW, Silverman ED, Alarcón-Riquelme ME, Kimberly RP, Harley JB, Wakeland EK, Graham RR, Gaffney PM, and Vyse TJ
- Subjects
- Age of Onset, Case-Control Studies, Hispanic or Latino genetics, Humans, Logistic Models, Multifactorial Inheritance, Mutagenesis, Insertional, Polymorphism, Single Nucleotide, Sequence Deletion, American Indian or Alaska Native genetics, Black People genetics, Genetic Load, HLA Antigens genetics, Lupus Erythematosus, Systemic genetics, White People genetics
- Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with marked gender and ethnic disparities. We report a large transancestral association study of SLE using Immunochip genotype data from 27,574 individuals of European (EA), African (AA) and Hispanic Amerindian (HA) ancestry. We identify 58 distinct non-HLA regions in EA, 9 in AA and 16 in HA (∼50% of these regions have multiple independent associations); these include 24 novel SLE regions (P<5 × 10
-8 ), refined association signals in established regions, extended associations to additional ancestries, and a disentangled complex HLA multigenic effect. The risk allele count (genetic load) exhibits an accelerating pattern of SLE risk, leading us to posit a cumulative hit hypothesis for autoimmune disease. Comparing results across the three ancestries identifies both ancestry-dependent and ancestry-independent contributions to SLE risk. Our results are consistent with the unique and complex histories of the populations sampled, and collectively help clarify the genetic architecture and ethnic disparities in SLE.- Published
- 2017
- Full Text
- View/download PDF
37. Higher Nevus Count Exhibits a Distinct DNA Methylation Signature in Healthy Human Skin: Implications for Melanoma.
- Author
-
Roos L, Sandling JK, Bell CG, Glass D, Mangino M, Spector TD, Deloukas P, Bataille V, and Bell JT
- Subjects
- Adult, Case-Control Studies, Epigenomics, Female, Gene Expression Regulation, Neoplastic, Genotype, Humans, Male, Melanoma epidemiology, Melanoma pathology, Nevus epidemiology, Nevus pathology, Phenotype, Reference Values, Registries, Skin Neoplasms epidemiology, Skin Neoplasms genetics, Skin Neoplasms pathology, United Kingdom, DNA Methylation genetics, Genetic Predisposition to Disease epidemiology, Genome-Wide Association Study methods, Melanoma genetics, Nevus genetics
- Abstract
High nevus count is the strongest risk factor for melanoma, and although gene variants have been discovered for both traits, epigenetic variation is unexplored. We investigated 322 healthy human skin DNA methylomes associated with total body nevi count, incorporating genetic and transcriptomic variation. DNA methylation changes were identified at genes involved in melanocyte biology, such as RAF1 (P = 1.2 × 10
-6 ) and CTC1 (region: P = 6.3 × 10-4 ), and other genes including ARRDC1 (P = 3.1 × 10-7 ). A subset exhibited coordinated methylation and transcription changes within the same biopsy. The total analysis was also enriched for melanoma-associated DNA methylation variation (P = 6.33 × 10-6 ). In addition, we show that skin DNA methylation is associated in cis with known genome-wide association study single nucleotide polymorphisms for nevus count, at PLA2G6 (P = 1.7 × 10-49 ) and NID1 (P = 6.4 × 10-14 ), as well as melanoma risk, including in or near MC1R, MX2, and TERT/CLPTM1L (P < 1 × 10-10 ). Our analysis using a uniquely large dataset comprising healthy skin DNA methylomes identified known and additional regulatory loci and pathways in nevi and melanoma biology. This integrative study improves our understanding of predisposition to nevi and their potential contribution to melanoma pathogenesis., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
38. Epigenome-wide association study of body mass index, and the adverse outcomes of adiposity.
- Author
-
Wahl S, Drong A, Lehne B, Loh M, Scott WR, Kunze S, Tsai PC, Ried JS, Zhang W, Yang Y, Tan S, Fiorito G, Franke L, Guarrera S, Kasela S, Kriebel J, Richmond RC, Adamo M, Afzal U, Ala-Korpela M, Albetti B, Ammerpohl O, Apperley JF, Beekman M, Bertazzi PA, Black SL, Blancher C, Bonder MJ, Brosch M, Carstensen-Kirberg M, de Craen AJ, de Lusignan S, Dehghan A, Elkalaawy M, Fischer K, Franco OH, Gaunt TR, Hampe J, Hashemi M, Isaacs A, Jenkinson A, Jha S, Kato N, Krogh V, Laffan M, Meisinger C, Meitinger T, Mok ZY, Motta V, Ng HK, Nikolakopoulou Z, Nteliopoulos G, Panico S, Pervjakova N, Prokisch H, Rathmann W, Roden M, Rota F, Rozario MA, Sandling JK, Schafmayer C, Schramm K, Siebert R, Slagboom PE, Soininen P, Stolk L, Strauch K, Tai ES, Tarantini L, Thorand B, Tigchelaar EF, Tumino R, Uitterlinden AG, van Duijn C, van Meurs JB, Vineis P, Wickremasinghe AR, Wijmenga C, Yang TP, Yuan W, Zhernakova A, Batterham RL, Smith GD, Deloukas P, Heijmans BT, Herder C, Hofman A, Lindgren CM, Milani L, van der Harst P, Peters A, Illig T, Relton CL, Waldenberger M, Järvelin MR, Bollati V, Soong R, Spector TD, Scott J, McCarthy MI, Elliott P, Bell JT, Matullo G, Gieger C, Kooner JS, Grallert H, and Chambers JC
- Subjects
- Adipose Tissue metabolism, Asian People genetics, Blood metabolism, Cohort Studies, Diabetes Mellitus, Type 2 complications, Europe ethnology, Female, Genetic Markers, Genetic Predisposition to Disease, Humans, India ethnology, Male, Obesity blood, Obesity complications, Overweight blood, Overweight complications, Overweight genetics, White People genetics, Adiposity genetics, Body Mass Index, DNA Methylation genetics, Diabetes Mellitus, Type 2 genetics, Epigenesis, Genetic, Epigenomics, Genome-Wide Association Study, Obesity genetics
- Abstract
Approximately 1.5 billion people worldwide are overweight or affected by obesity, and are at risk of developing type 2 diabetes, cardiovascular disease and related metabolic and inflammatory disturbances. Although the mechanisms linking adiposity to associated clinical conditions are poorly understood, recent studies suggest that adiposity may influence DNA methylation, a key regulator of gene expression and molecular phenotype. Here we use epigenome-wide association to show that body mass index (BMI; a key measure of adiposity) is associated with widespread changes in DNA methylation (187 genetic loci with P < 1 × 10
-7 , range P = 9.2 × 10-8 to 6.0 × 10-46 ; n = 10,261 samples). Genetic association analyses demonstrate that the alterations in DNA methylation are predominantly the consequence of adiposity, rather than the cause. We find that methylation loci are enriched for functional genomic features in multiple tissues (P < 0.05), and show that sentinel methylation markers identify gene expression signatures at 38 loci (P < 9.0 × 10-6 , range P = 5.5 × 10-6 to 6.1 × 10-35 , n = 1,785 samples). The methylation loci identify genes involved in lipid and lipoprotein metabolism, substrate transport and inflammatory pathways. Finally, we show that the disturbances in DNA methylation predict future development of type 2 diabetes (relative risk per 1 standard deviation increase in methylation risk score: 2.3 (2.07-2.56); P = 1.1 × 10-54 ). Our results provide new insights into the biologic pathways influenced by adiposity, and may enable development of new strategies for prediction and prevention of type 2 diabetes and other adverse clinical consequences of obesity.- Published
- 2017
- Full Text
- View/download PDF
39. Epigenetic Patterns in Blood Associated With Lipid Traits Predict Incident Coronary Heart Disease Events and Are Enriched for Results From Genome-Wide Association Studies.
- Author
-
Hedman ÅK, Mendelson MM, Marioni RE, Gustafsson S, Joehanes R, Irvin MR, Zhi D, Sandling JK, Yao C, Liu C, Liang L, Huan T, McRae AF, Demissie S, Shah S, Starr JM, Cupples LA, Deloukas P, Spector TD, Sundström J, Krauss RM, Arnett DK, Deary IJ, Lind L, Levy D, and Ingelsson E
- Subjects
- Aged, Biomarkers blood, Coronary Disease blood, Coronary Disease diagnosis, Coronary Disease epidemiology, CpG Islands, Cross-Sectional Studies, Dyslipidemias blood, Dyslipidemias diagnosis, Dyslipidemias epidemiology, Epigenomics methods, Europe epidemiology, Female, Genetic Predisposition to Disease, Genome-Wide Association Study, Humans, Incidence, Male, Metabolomics methods, Middle Aged, Phenotype, Prognosis, Prospective Studies, Risk Assessment, Risk Factors, United States epidemiology, Coronary Disease genetics, DNA Methylation, Dyslipidemias genetics, Epigenesis, Genetic, Lipid Metabolism genetics, Lipids blood, Quantitative Trait Loci
- Abstract
Background: Genome-wide association studies have identified loci influencing circulating lipid concentrations in humans; further information on novel contributing genes, pathways, and biology may be gained through studies of epigenetic modifications., Methods and Results: To identify epigenetic changes associated with lipid concentrations, we assayed genome-wide DNA methylation at cytosine-guanine dinucleotides (CpGs) in whole blood from 2306 individuals from 2 population-based cohorts, with replication of findings in 2025 additional individuals. We identified 193 CpGs associated with lipid levels in the discovery stage ( P <1.08E-07) and replicated 33 (at Bonferroni-corrected P <0.05), including 25 novel CpGs not previously associated with lipids. Genes at lipid-associated CpGs were enriched in lipid and amino acid metabolism processes. A differentially methylated locus associated with triglycerides and high-density lipoprotein cholesterol (HDL-C; cg27243685; P =8.1E-26 and 9.3E-19) was associated with cis -expression of a reverse cholesterol transporter ( ABCG1; P =7.2E-28) and incident cardiovascular disease events (hazard ratio per SD increment, 1.38; 95% confidence interval, 1.15-1.66; P =0.0007). We found significant cis -methylation quantitative trait loci at 64% of the 193 CpGs with an enrichment of signals from genome-wide association studies of lipid levels ( P
TC =0.004, PHDL-C =0.008 and Ptriglycerides =0.00003) and coronary heart disease ( P =0.0007). For example, genome-wide significant variants associated with low-density lipoprotein cholesterol and coronary heart disease at APOB were cis -methylation quantitative trait loci for a low-density lipoprotein cholesterol-related differentially methylated locus., Conclusions: We report novel associations of DNA methylation with lipid levels, describe epigenetic mechanisms related to previous genome-wide association studies discoveries, and provide evidence implicating epigenetic regulation of reverse cholesterol transport in blood in relation to occurrence of cardiovascular disease events., (© 2017 The Authors.)- Published
- 2017
- Full Text
- View/download PDF
40. Genome-wide DNA methylation analysis in multiple tissues in primary Sjögren's syndrome reveals regulatory effects at interferon-induced genes.
- Author
-
Imgenberg-Kreuz J, Sandling JK, Almlöf JC, Nordlund J, Signér L, Norheim KB, Omdal R, Rönnblom L, Eloranta ML, Syvänen AC, and Nordmark G
- Subjects
- 2',5'-Oligoadenylate Synthetase genetics, Adult, Aged, Antigens genetics, Antigens, CD19 genetics, Case-Control Studies, Cytoskeletal Proteins genetics, Female, Gene Expression, Genome-Wide Association Study, Humans, Male, Middle Aged, Myxovirus Resistance Proteins genetics, Neoplasm Proteins genetics, Poly(ADP-ribose) Polymerases genetics, Salivary Glands, Minor pathology, Sjogren's Syndrome blood, Sjogren's Syndrome pathology, DNA Methylation, Epigenesis, Genetic, Interferons genetics, Sjogren's Syndrome genetics
- Abstract
Objectives: Increasing evidence suggests an epigenetic contribution to the pathogenesis of autoimmune diseases, including primary Sjögren's Syndrome (pSS). The aim of this study was to investigate the role of DNA methylation in pSS by analysing multiple tissues from patients and controls., Methods: Genome-wide DNA methylation profiles were generated using HumanMethylation450K BeadChips for whole blood, CD19+ B cells and minor salivary gland biopsies. Gene expression was analysed in CD19+ B cells by RNA-sequencing. Analysis of genetic regulatory effects on DNA methylation at known pSS risk loci was performed., Results: We identified prominent hypomethylation of interferon (IFN)-regulated genes in whole blood and CD19+ B cells, including at the genes MX1, IFI44L and PARP9, replicating previous reports in pSS, as well as identifying a large number of novel associations. Enrichment for genomic overlap with histone marks for enhancer and promoter regions was observed. We showed for the first time that hypomethylation of IFN-regulated genes in pSS B cells was associated with their increased expression. In minor salivary gland biopsies we observed hypomethylation of the IFN-induced gene OAS2. Pathway and disease analysis resulted in enrichment of antigen presentation, IFN signalling and lymphoproliferative disorders. Evidence for genetic control of methylation levels at known pSS risk loci was observed., Conclusions: Our study highlights the role of epigenetic regulation of IFN-induced genes in pSS where replication is needed for novel findings. The association with altered gene expression suggests a functional mechanism for differentially methylated CpG sites in pSS aetiology., Competing Interests: Conflicts of Interest: None declared., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)
- Published
- 2016
- Full Text
- View/download PDF
41. Immunoseq: the identification of functionally relevant variants through targeted capture and sequencing of active regulatory regions in human immune cells.
- Author
-
Morin A, Kwan T, Ge B, Letourneau L, Ban M, Tandre K, Caron M, Sandling JK, Carlsson J, Bourque G, Laprise C, Montpetit A, Syvanen AC, Ronnblom L, Sawcer SJ, Lathrop MG, and Pastinen T
- Subjects
- Genome-Wide Association Study, Humans, Molecular Sequence Annotation, Quantitative Trait Loci genetics, T-Lymphocytes metabolism, Alleles, DNA Mutational Analysis, Gene Expression Profiling, Sequence Analysis, RNA, T-Lymphocytes immunology
- Abstract
Background: The observation that the genetic variants identified in genome-wide association studies (GWAS) frequently lie in non-coding regions of the genome that contain cis-regulatory elements suggests that altered gene expression underlies the development of many complex traits. In order to efficiently make a comprehensive assessment of the impact of non-coding genetic variation in immune related diseases we emulated the whole-exome sequencing paradigm and developed a custom capture panel for the known DNase I hypersensitive site (DHS) in immune cells - "Immunoseq"., Results: We performed Immunoseq in 30 healthy individuals where we had existing transcriptome data from T cells. We identified a large number of novel non-coding variants in these samples. Relying on allele specific expression measurements, we also showed that our selected capture regions are enriched for functional variants that have an impact on differential allelic gene expression. The results from a replication set with 180 samples confirmed our observations., Conclusions: We show that Immunoseq is a powerful approach to detect novel rare variants in regulatory regions. We also demonstrate that these novel variants have a potential functional role in immune cells.
- Published
- 2016
- Full Text
- View/download PDF
42. Epigenome-wide DNA methylation patterns associated with fatigue in primary Sjögren's syndrome.
- Author
-
Brække Norheim K, Imgenberg-Kreuz J, Jonsdottir K, Janssen EA, Syvänen AC, Sandling JK, Nordmark G, and Omdal R
- Subjects
- Adult, Aged, Cytosine Nucleotides genetics, Female, Genome-Wide Association Study, Humans, Male, Middle Aged, Sjogren's Syndrome genetics, DNA Methylation, Epigenomics, Fatigue Syndrome, Chronic genetics, Sjogren's Syndrome complications
- Abstract
Objective: Chronic fatigue is a common, disabling and poorly understood phenomenon. Recent studies indicate that epigenetic mechanisms may be involved in the expression of fatigue, a prominent feature of primary SS (pSS). The aim of this study was to investigate whether DNA methylation profiles of whole blood are associated with fatigue in patients with pSS., Methods: Forty-eight pSS patients with high (n = 24) or low (n = 24) fatigue as measured by a visual analogue scale were included. Genome-wide DNA methylation was investigated using the Illumina HumanMethylation450 BeadChip array. After quality control, a total of 383 358 Cytosine-phosphate-Guanine (CpG) sites remained for further analysis. Age, sex and differential cell count estimates were included as covariates in the association model. A false discovery rate-corrected P < 0.05 was considered significant, and a cut-off of 3% average difference in methylation levels between high- and low-fatigue patients was applied., Results: A total of 251 differentially methylated CpG sites were associated with fatigue. The CpG site with the most pronounced hypomethylation in pSS high fatigue annotated to the SBF2-antisense RNA1 gene. The most distinct hypermethylation was observed at a CpG site annotated to the lymphotoxin alpha gene. Functional pathway analysis of genes with differently methylated CpG sites in subjects with high vs low fatigue revealed enrichment in several pathways associated with innate and adaptive immunity., Conclusion: Some genes involved in regulation of the immune system and in inflammation are differently methylated in pSS patients with high vs low fatigue. These findings point to functional networks that may underlie fatigue. Epigenetic changes could constitute a fatigue-regulating mechanism in pSS., (© The Author 2016. Published by Oxford University Press on behalf of the British Society for Rheumatology. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
43. Epigenome-wide association study (EWAS) of BMI, BMI change and waist circumference in African American adults identifies multiple replicated loci.
- Author
-
Demerath EW, Guan W, Grove ML, Aslibekyan S, Mendelson M, Zhou YH, Hedman ÅK, Sandling JK, Li LA, Irvin MR, Zhi D, Deloukas P, Liang L, Liu C, Bressler J, Spector TD, North K, Li Y, Absher DM, Levy D, Arnett DK, Fornage M, Pankow JS, and Boerwinkle E
- Subjects
- Black or African American genetics, Aged, Atherosclerosis pathology, Body Mass Index, Female, Genome-Wide Association Study, Humans, Lipid Metabolism genetics, Male, Metabolic Networks and Pathways genetics, Middle Aged, Obesity pathology, Waist Circumference genetics, White People genetics, Atherosclerosis genetics, DNA Methylation genetics, Epigenesis, Genetic genetics, Obesity genetics
- Abstract
Obesity is an important component of the pathophysiology of chronic diseases. Identifying epigenetic modifications associated with elevated adiposity, including DNA methylation variation, may point to genomic pathways that are dysregulated in numerous conditions. The Illumina 450K Bead Chip array was used to assay DNA methylation in leukocyte DNA obtained from 2097 African American adults in the Atherosclerosis Risk in Communities (ARIC) study. Mixed-effects regression models were used to test the association of methylation beta value with concurrent body mass index (BMI) and waist circumference (WC), and BMI change, adjusting for batch effects and potential confounders. Replication using whole-blood DNA from 2377 White adults in the Framingham Heart Study and CD4+ T cell DNA from 991 Whites in the Genetics of Lipid Lowering Drugs and Diet Network Study was followed by testing using adipose tissue DNA from 648 women in the Multiple Tissue Human Expression Resource cohort. Seventy-six BMI-related probes, 164 WC-related probes and 8 BMI change-related probes passed the threshold for significance in ARIC (P < 1 × 10(-7); Bonferroni), including probes in the recently reported HIF3A, CPT1A and ABCG1 regions. Replication using blood DNA was achieved for 37 BMI probes and 1 additional WC probe. Sixteen of these also replicated in adipose tissue, including 15 novel methylation findings near genes involved in lipid metabolism, immune response/cytokine signaling and other diverse pathways, including LGALS3BP, KDM2B, PBX1 and BBS2, among others. Adiposity traits are associated with DNA methylation at numerous CpG sites that replicate across studies despite variation in tissue type, ethnicity and analytic approaches., (© The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
44. DNA methylation of lipid-related genes affects blood lipid levels.
- Author
-
Pfeiffer L, Wahl S, Pilling LC, Reischl E, Sandling JK, Kunze S, Holdt LM, Kretschmer A, Schramm K, Adamski J, Klopp N, Illig T, Hedman ÅK, Roden M, Hernandez DG, Singleton AB, Thasler WE, Grallert H, Gieger C, Herder C, Teupser D, Meisinger C, Spector TD, Kronenberg F, Prokisch H, Melzer D, Peters A, Deloukas P, Ferrucci L, and Waldenberger M
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 1, Adult, Aged, Aged, 80 and over, Cohort Studies, Female, Genetic Loci, Germany, Humans, Male, Middle Aged, ATP-Binding Cassette Transporters biosynthesis, ATP-Binding Cassette Transporters genetics, DNA Methylation genetics, Epigenesis, Genetic, Lipids blood, Myocardial Infarction blood, Myocardial Infarction genetics, Sterol Regulatory Element Binding Protein 1 biosynthesis, Sterol Regulatory Element Binding Protein 1 genetics
- Abstract
Background: Epigenetic mechanisms might be involved in the regulation of interindividual lipid level variability and thus may contribute to the cardiovascular risk profile. The aim of this study was to investigate the association between genome-wide DNA methylation and blood lipid levels high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides, and total cholesterol. Observed DNA methylation changes were also further analyzed to examine their relationship with previous hospitalized myocardial infarction., Methods and Results: Genome-wide DNA methylation patterns were determined in whole blood samples of 1776 subjects of the Cooperative Health Research in the Region of Augsburg F4 cohort using the Infinium HumanMethylation450 BeadChip (Illumina). Ten novel lipid-related CpG sites annotated to various genes including ABCG1, MIR33B/SREBF1, and TNIP1 were identified. CpG cg06500161, located in ABCG1, was associated in opposite directions with both high-density lipoprotein cholesterol (β coefficient=-0.049; P=8.26E-17) and triglyceride levels (β=0.070; P=1.21E-27). Eight associations were confirmed by replication in the Cooperative Health Research in the Region of Augsburg F3 study (n=499) and in the Invecchiare in Chianti, Aging in the Chianti Area study (n=472). Associations between triglyceride levels and SREBF1 and ABCG1 were also found in adipose tissue of the Multiple Tissue Human Expression Resource cohort (n=634). Expression analysis revealed an association between ABCG1 methylation and lipid levels that might be partly mediated by ABCG1 expression. DNA methylation of ABCG1 might also play a role in previous hospitalized myocardial infarction (odds ratio, 1.15; 95% confidence interval=1.06-1.25)., Conclusions: Epigenetic modifications of the newly identified loci might regulate disturbed blood lipid levels and thus contribute to the development of complex lipid-related diseases., (© 2015 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
45. Meta-analysis in more than 17,900 cases of ischemic stroke reveals a novel association at 12q24.12.
- Author
-
Kilarski LL, Achterberg S, Devan WJ, Traylor M, Malik R, Lindgren A, Pare G, Sharma P, Slowik A, Thijs V, Walters M, Worrall BB, Sale MM, Algra A, Kappelle LJ, Wijmenga C, Norrving B, Sandling JK, Rönnblom L, Goris A, Franke A, Sudlow C, Rothwell PM, Levi C, Holliday EG, Fornage M, Psaty B, Gretarsdottir S, Thorsteinsdottir U, Seshadri S, Mitchell BD, Kittner S, Clarke R, Hopewell JC, Bis JC, Boncoraglio GB, Meschia J, Ikram MA, Hansen BM, Montaner J, Thorleifsson G, Stefanson K, Rosand J, de Bakker PI, Farrall M, Dichgans M, Markus HS, and Bevan S
- Subjects
- Cerebral Hemorrhage genetics, Genome-Wide Association Study, Genotype, Humans, Polymorphism, Single Nucleotide genetics, Risk, Stroke etiology, Brain Ischemia genetics, Chromosomes, Human, Pair 12 genetics, Genetic Predisposition to Disease, Stroke genetics
- Abstract
Objectives: To perform a genome-wide association study (GWAS) using the Immunochip array in 3,420 cases of ischemic stroke and 6,821 controls, followed by a meta-analysis with data from more than 14,000 additional ischemic stroke cases., Methods: Using the Immunochip, we genotyped 3,420 ischemic stroke cases and 6,821 controls. After imputation we meta-analyzed the results with imputed GWAS data from 3,548 cases and 5,972 controls recruited from the ischemic stroke WTCCC2 study, and with summary statistics from a further 8,480 cases and 56,032 controls in the METASTROKE consortium. A final in silico "look-up" of 2 single nucleotide polymorphisms in 2,522 cases and 1,899 controls was performed. Associations were also examined in 1,088 cases with intracerebral hemorrhage and 1,102 controls., Results: In an overall analysis of 17,970 cases of ischemic stroke and 70,764 controls, we identified a novel association on chromosome 12q24 (rs10744777, odds ratio [OR] 1.10 [1.07-1.13], p = 7.12 × 10(-11)) with ischemic stroke. The association was with all ischemic stroke rather than an individual stroke subtype, with similar effect sizes seen in different stroke subtypes. There was no association with intracerebral hemorrhage (OR 1.03 [0.90-1.17], p = 0.695)., Conclusion: Our results show, for the first time, a genetic risk locus associated with ischemic stroke as a whole, rather than in a subtype-specific manner. This finding was not associated with intracerebral hemorrhage., (© 2014 American Academy of Neurology.)
- Published
- 2014
- Full Text
- View/download PDF
46. DNA methylation and body-mass index: a genome-wide analysis.
- Author
-
Dick KJ, Nelson CP, Tsaprouni L, Sandling JK, Aïssi D, Wahl S, Meduri E, Morange PE, Gagnon F, Grallert H, Waldenberger M, Peters A, Erdmann J, Hengstenberg C, Cambien F, Goodall AH, Ouwehand WH, Schunkert H, Thompson JR, Spector TD, Gieger C, Trégouët DA, Deloukas P, and Samani NJ
- Subjects
- Apoptosis Regulatory Proteins, Basic Helix-Loop-Helix Transcription Factors genetics, Body Mass Index, Chromosomes, Human, Pair 15 genetics, Chromosomes, Human, Pair 19 genetics, Female, Genome-Wide Association Study, Humans, Male, Middle Aged, Polymorphism, Single Nucleotide genetics, Repressor Proteins, DNA Methylation genetics, Obesity genetics
- Abstract
Background: Obesity is a major health problem that is determined by interactions between lifestyle and environmental and genetic factors. Although associations between several genetic variants and body-mass index (BMI) have been identified, little is known about epigenetic changes related to BMI. We undertook a genome-wide analysis of methylation at CpG sites in relation to BMI., Methods: 479 individuals of European origin recruited by the Cardiogenics Consortium formed our discovery cohort. We typed their whole-blood DNA with the Infinium HumanMethylation450 array. After quality control, methylation levels were tested for association with BMI. Methylation sites showing an association with BMI at a false discovery rate q value of 0·05 or less were taken forward for replication in a cohort of 339 unrelated white patients of northern European origin from the MARTHA cohort. Sites that remained significant in this primary replication cohort were tested in a second replication cohort of 1789 white patients of European origin from the KORA cohort. We examined whether methylation levels at identified sites also showed an association with BMI in DNA from adipose tissue (n=635) and skin (n=395) obtained from white female individuals participating in the MuTHER study. Finally, we examined the association of methylation at BMI-associated sites with genetic variants and with gene expression., Findings: 20 individuals from the discovery cohort were excluded from analyses after quality-control checks, leaving 459 participants. After adjustment for covariates, we identified an association (q value ≤0·05) between methylation at five probes across three different genes and BMI. The associations with three of these probes--cg22891070, cg27146050, and cg16672562, all of which are in intron 1 of HIF3A--were confirmed in both the primary and second replication cohorts. For every 0·1 increase in methylation β value at cg22891070, BMI was 3·6% (95% CI 2·4-4·9) higher in the discovery cohort, 2·7% (1·2-4·2) higher in the primary replication cohort, and 0·8% (0·2-1·4) higher in the second replication cohort. For the MuTHER cohort, methylation at cg22891070 was associated with BMI in adipose tissue (p=1·72 × 10(-5)) but not in skin (p=0·882). We observed a significant inverse correlation (p=0·005) between methylation at cg22891070 and expression of one HIF3A gene-expression probe in adipose tissue. Two single nucleotide polymorphisms--rs8102595 and rs3826795--had independent associations with methylation at cg22891070 in all cohorts. However, these single nucleotide polymorphisms were not significantly associated with BMI., Interpretation: Increased BMI in adults of European origin is associated with increased methylation at the HIF3A locus in blood cells and in adipose tissue. Our findings suggest that perturbation of hypoxia inducible transcription factor pathways could have an important role in the response to increased weight in people., Funding: The European Commission, National Institute for Health Research, British Heart Foundation, and Wellcome Trust., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
47. Association of STAT4 polymorphism with severe renal insufficiency in lupus nephritis.
- Author
-
Bolin K, Sandling JK, Zickert A, Jönsen A, Sjöwall C, Svenungsson E, Bengtsson AA, Eloranta ML, Rönnblom L, Syvänen AC, Gunnarsson I, and Nordmark G
- Subjects
- Adult, Case-Control Studies, Female, Genome-Wide Association Study, Humans, Lupus Nephritis complications, Male, Renal Insufficiency, Chronic etiology, Severity of Illness Index, Genetic Predisposition to Disease, Lupus Nephritis genetics, Polymorphism, Single Nucleotide, Renal Insufficiency, Chronic genetics, STAT4 Transcription Factor genetics
- Abstract
Lupus nephritis is a cause of significant morbidity in systemic lupus erythematosus (SLE) and its genetic background has not been completely clarified. The aim of this investigation was to analyze single nucleotide polymorphisms (SNPs) for association with lupus nephritis, its severe form proliferative nephritis and renal outcome, in two Swedish cohorts. Cohort I (n = 567 SLE cases, n = 512 controls) was previously genotyped for 5676 SNPs and cohort II (n = 145 SLE cases, n = 619 controls) was genotyped for SNPs in STAT4, IRF5, TNIP1 and BLK. Case-control and case-only association analyses for patients with lupus nephritis, proliferative nephritis and severe renal insufficiency were performed. In the case-control analysis of cohort I, four highly linked SNPs in STAT4 were associated with lupus nephritis with genome wide significance with p = 3.7 × 10(-9), OR 2.20 for the best SNP rs11889341. Strong signals of association between IRF5 and an HLA-DR3 SNP marker were also detected in the lupus nephritis case versus healthy control analysis (p <0.0001). An additional six genes showed an association with lupus nephritis with p <0.001 (PMS2, TNIP1, CARD11, ITGAM, BLK and IRAK1). In the case-only meta-analysis of the two cohorts, the STAT4 SNP rs7582694 was associated with severe renal insufficiency with p = 1.6 × 10(-3) and OR 2.22. We conclude that genetic variations in STAT4 predispose to lupus nephritis and a worse outcome with severe renal insufficiency.
- Published
- 2013
- Full Text
- View/download PDF
48. Global analysis of DNA methylation variation in adipose tissue from twins reveals links to disease-associated variants in distal regulatory elements.
- Author
-
Grundberg E, Meduri E, Sandling JK, Hedman AK, Keildson S, Buil A, Busche S, Yuan W, Nisbet J, Sekowska M, Wilk A, Barrett A, Small KS, Ge B, Caron M, Shin SY, Lathrop M, Dermitzakis ET, McCarthy MI, Spector TD, Bell JT, and Deloukas P
- Subjects
- Body Mass Index, Chromosome Mapping, Epigenomics, Female, Gene Expression Profiling, Gene Expression Regulation, Genome, Human, Humans, Hybridization, Genetic, Oligonucleotide Array Sequence Analysis, Phenotype, Quantitative Trait Loci, Sequence Analysis, DNA, Sulfites metabolism, Twins genetics, Adipose Tissue, DNA Methylation, Polymorphism, Single Nucleotide, Regulatory Sequences, Nucleic Acid
- Abstract
Epigenetic modifications such as DNA methylation play a key role in gene regulation and disease susceptibility. However, little is known about the genome-wide frequency, localization, and function of methylation variation and how it is regulated by genetic and environmental factors. We utilized the Multiple Tissue Human Expression Resource (MuTHER) and generated Illumina 450K adipose methylome data from 648 twins. We found that individual CpGs had low variance and that variability was suppressed in promoters. We noted that DNA methylation variation was highly heritable (h(2)median = 0.34) and that shared environmental effects correlated with metabolic phenotype-associated CpGs. Analysis of methylation quantitative-trait loci (metQTL) revealed that 28% of CpGs were associated with nearby SNPs, and when overlapping them with adipose expression quantitative-trait loci (eQTL) from the same individuals, we found that 6% of the loci played a role in regulating both gene expression and DNA methylation. These associations were bidirectional, but there were pronounced negative associations for promoter CpGs. Integration of metQTL with adipose reference epigenomes and disease associations revealed significant enrichment of metQTL overlapping metabolic-trait or disease loci in enhancers (the strongest effects were for high-density lipoprotein cholesterol and body mass index [BMI]). We followed up with the BMI SNP rs713586, a cg01884057 metQTL that overlaps an enhancer upstream of ADCY3, and used bisulphite sequencing to refine this region. Our results showed widespread population invariability yet sequence dependence on adipose DNA methylation but that incorporating maps of regulatory elements aid in linking CpG variation to gene regulation and disease risk in a tissue-dependent manner., (Copyright © 2013 The American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
49. Genes identified in Asian SLE GWASs are also associated with SLE in Caucasian populations.
- Author
-
Wang C, Ahlford A, Järvinen TM, Nordmark G, Eloranta ML, Gunnarsson I, Svenungsson E, Padyukov L, Sturfelt G, Jönsen A, Bengtsson AA, Truedsson L, Eriksson C, Rantapää-Dahlqvist S, Sjöwall C, Julkunen H, Criswell LA, Graham RR, Behrens TW, Kere J, Rönnblom L, Syvänen AC, and Sandling JK
- Subjects
- Asian People genetics, Case-Control Studies, Gene Frequency, Genetic Predisposition to Disease, Genome-Wide Association Study, Humans, Lupus Erythematosus, Systemic ethnology, Polymorphism, Single Nucleotide, White People genetics, Lupus Erythematosus, Systemic genetics
- Abstract
Recent genome-wide association studies (GWASs) conducted in Asian populations have identified novel risk loci for systemic lupus erythematosus (SLE). Here, we genotyped 10 single-nucleotide polymorphisms (SNPs) in eight such loci and investigated their disease associations in three independent Caucasian SLE case-control cohorts recruited from Sweden, Finland and the United States. The disease associations of the SNPs in ETS1, IKZF1, LRRC18-WDFY4, RASGRP3, SLC15A4, TNIP1 and 16p11.2 were replicated, whereas no solid evidence of association was observed for the 7q11.23 locus in the Caucasian cohorts. SLC15A4 was significantly associated with renal involvement in SLE. The association of TNIP1 was more pronounced in SLE patients with renal and immunological disorder, which is corroborated by two previous studies in Asian cohorts. The effects of all the associated SNPs, either conferring risk for or being protective against SLE, were in the same direction in Caucasians and Asians. The magnitudes of the allelic effects for most of the SNPs were also comparable across different ethnic groups. On the contrary, remarkable differences in allele frequencies between Caucasian and Asian populations were observed for all associated SNPs. In conclusion, most of the novel SLE risk loci identified by GWASs in Asian populations were also associated with SLE in Caucasian populations. We observed both similarities and differences with respect to the effect sizes and risk allele frequencies across ethnicities.
- Published
- 2013
- Full Text
- View/download PDF
50. Contribution of IKBKE and IFIH1 gene variants to SLE susceptibility.
- Author
-
Wang C, Ahlford A, Laxman N, Nordmark G, Eloranta ML, Gunnarsson I, Svenungsson E, Padyukov L, Sturfelt G, Jönsen A, Bengtsson AA, Truedsson L, Rantapää-Dahlqvist S, Sjöwall C, Sandling JK, Rönnblom L, and Syvänen AC
- Subjects
- Case-Control Studies, DNA-Binding Proteins metabolism, Gene Frequency, Genetic Association Studies, Haplotypes, Humans, I-kappa B Kinase metabolism, Interferon-Induced Helicase, IFIH1, Protein Binding, RNA Splicing Factors, Transcription Factors metabolism, DEAD-box RNA Helicases genetics, Genetic Predisposition to Disease, I-kappa B Kinase genetics, Lupus Erythematosus, Systemic genetics, Polymorphism, Single Nucleotide
- Abstract
The type I interferon system genes IKBKE and IFIH1 are associated with the risk of systemic lupus erythematosus (SLE). To identify the sequence variants that are able to account for the disease association, we resequenced the genes IKBKE and IFIH1. Eighty-six single-nucleotide variants (SNVs) with potentially functional effect or differences in allele frequencies between patients and controls determined by sequencing were further genotyped in 1140 SLE patients and 2060 controls. In addition, 108 imputed sequence variants in IKBKE and IFIH1 were included in the association analysis. Ten IKBKE SNVs and three IFIH1 SNVs were associated with SLE. The SNVs rs1539241 and rs12142086 tagged two independent association signals in IKBKE, and the haplotype carrying their risk alleles showed an odds ratio of 1.68 (P-value=1.0 × 10(-5)). The risk allele of rs12142086 affects the binding of splicing factor 1 in vitro and could thus influence its transcriptional regulatory function. Two independent association signals were also detected in IFIH1, which were tagged by a low-frequency SNV rs78456138 and a missense SNV rs3747517. Their joint effect is protective against SLE (odds ratio=0.56; P-value=6.6 × 10(-3)). In conclusion, we have identified new SLE-associated sequence variants in IKBKE and IFIH1, and proposed functional hypotheses for the association signals.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.