Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Ann Rheum Dis ; 82(6): 754-762, 2023 06.
Article in English | MEDLINE | ID: mdl-36858821

ABSTRACT

BACKGROUND: Type I interferons (IFN-Is) play a role in a broad range of rheumatic and musculoskeletal diseases (RMDs), and compelling evidence suggests that their measurement could have clinical value, although testing has not progressed into clinical settings. OBJECTIVE: To develop evidence-based points to consider (PtC) for the measurement and reporting of IFN-I assays in clinical research and to determine their potential clinical utility. METHODS: EULAR standardised operating procedures were followed. A task force including rheumatologists, immunologists, translational scientists and a patient partner was formed. Two systematic reviews were conducted to address methodological and clinical questions. PtC were formulated based on the retrieved evidence and expert opinion. Level of evidence and agreement was determined. RESULTS: Two overarching principles and 11 PtC were defined. The first set (PtC 1-4) concerned terminology, assay characteristics and reporting practices to enable more consistent reporting and facilitate translation and collaborations. The second set (PtC 5-11) addressed clinical applications for diagnosis and outcome assessments, including disease activity, prognosis and prediction of treatment response. The mean level of agreement was generally high, mainly in the first PtC set and for clinical applications in systemic lupus erythematosus. Harmonisation of assay methodology and clinical validation were key points for the research agenda. CONCLUSIONS: IFN-I assays have a high potential for implementation in the clinical management of RMDs. Uptake of these PtC will facilitate the progress of IFN-I assays into clinical practice and may be also of interest beyond rheumatology.


Subject(s)
Musculoskeletal Diseases , Rheumatology , Humans
2.
PLoS Genet ; 16(10): e1009199, 2020 10.
Article in English | MEDLINE | ID: mdl-33104735

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.


Subject(s)
Genetic Predisposition to Disease , HLA-A2 Antigen/genetics , Multiple Sclerosis/genetics , Quantitative Trait Loci/genetics , 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
3.
Rheumatology (Oxford) ; 61(8): 3461-3470, 2022 08 03.
Article in English | MEDLINE | ID: mdl-34888651

ABSTRACT

OBJECTIVE: To identify and characterize genetic loci associated with the risk of developing ANCA-associated vasculitides (AAV). METHODS: Genetic association analyses were performed after Illumina sequencing of 1853 genes and subsequent replication with genotyping of selected single nucleotide polymorphisms in a total cohort of 1110 Scandinavian cases with granulomatosis with polyangiitis or microscopic polyangiitis, and 1589 controls. A novel AAV-associated single nucleotide polymorphism was analysed for allele-specific effects on gene expression using luciferase reporter assay. RESULTS: PR3-ANCA+ AAV was significantly associated with two independent loci in the HLA-DPB1/HLA-DPA1 region [rs1042335, P = 6.3 × 10-61, odds ratio (OR) 0.10; rs9277341, P = 1.5 × 10-44, OR 0.22] and with rs28929474 in the SERPINA1 gene (P = 2.7 × 10-10, OR 2.9). MPO-ANCA+ AAV was significantly associated with the HLA-DQB1/HLA-DQA2 locus (rs9274619, P = 5.4 × 10-25, OR 3.7) and with a rare variant in the BACH2 gene (rs78275221, P = 7.9 × 10-7, OR 3.0), the latter a novel susceptibility locus for MPO-ANCA+ granulomatosis with polyangiitis/microscopic polyangiitis. The rs78275221-A risk allele reduced luciferase gene expression in endothelial cells, specifically, as compared with the non-risk allele. CONCLUSION: We identified a novel susceptibility locus for MPO-ANCA+ AAV and propose that the associated variant is of mechanistic importance, exerting a regulatory function on gene expression in specific cell types.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis , Granulomatosis with Polyangiitis , Microscopic Polyangiitis , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/complications , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/genetics , Antibodies, Antineutrophil Cytoplasmic , Endothelial Cells , Granulomatosis with Polyangiitis/complications , Granulomatosis with Polyangiitis/genetics , Humans , Microscopic Polyangiitis/complications , Microscopic Polyangiitis/genetics , Myeloblastin/genetics , Peroxidase
4.
BMC Pulm Med ; 22(1): 430, 2022 Nov 21.
Article in English | MEDLINE | ID: mdl-36404311

ABSTRACT

BACKGROUND: Ground glass opacity (GGO) is the main HRCT feature representing alveolitis in systemic sclerosis-associated interstitial lung disease (SSc-ILD), but may also represent other conditions such as atelectasis or edema. It is unclear how much this is affected by the HRCT scan protocol used. We aimed to compare the performance of three different HRCT protocols to evaluate the degree of SSc-ILD related changes. METHODS: Eleven patients with SSc underwent chest HRCT scan by three different protocols: First, a supine scan after lying down for 15 minutes, then two scans in alternating order: A prone position scan, and a supine position scan after performing 10 deep breaths using a positive expiratory pressure (PEP) device. The HRCT scans were evaluated by the Warrick score system for ILD-related findings. RESULTS: The three HRCT protocols were compared and resulted in different mean (95% CI) Warrick scores: 9.4 (5.3-13.4) in supine after rest; 7.5 (95% CI 3.8-11.1) in prone and 7.6 (95% CI 4.2-11.1) in supine after PEP. When comparing supine after rest to prone and supine after PEP, the latter two scans had a significantly lower score (p = 0.001 for both comparisons). In all cases, only sub-scores for ground glass opacities differed, while sub-scores for fibrosis-related changes did not change. CONCLUSIONS: Different HRCT scan protocols significantly altered the Warrick severity score for SSc-ILD findings, primarily because of changes in ground glass opacities. These differences may be clinically meaningful.


Subject(s)
Lung Diseases, Interstitial , Scleroderma, Systemic , Humans , Lung Diseases, Interstitial/diagnostic imaging , Lung Diseases, Interstitial/etiology , Scleroderma, Systemic/complications , Tomography, X-Ray Computed/methods
5.
Ann Rheum Dis ; 80(1): 109-117, 2021 01.
Article in English | MEDLINE | ID: mdl-33037003

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.


Subject(s)
Antigen Presentation/genetics , Immunity, Innate/genetics , Interferon Type I/immunology , Lupus Erythematosus, Systemic/genetics , Lymphopoiesis/genetics , T-Lymphocytes/immunology , 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
6.
Ann Rheum Dis ; 79(3): 363-369, 2020 03.
Article in English | MEDLINE | ID: mdl-31826855

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-ß2-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.


Subject(s)
Genetic Predisposition to Disease/epidemiology , Lupus Erythematosus, Systemic/genetics , Lupus Nephritis/genetics , Risk Assessment/statistics & numerical data , Severity of Illness Index , 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
7.
Scand J Immunol ; 91(6): e12880, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32219875

ABSTRACT

Synthetic Toll-like receptor (TLR) 7 agonists have been suggested as immune modulators in a range of conditions. In contrast, self-derived TLR7 activators, such as RNA-containing immune complexes (RNA-IC), can contribute to autoimmune diseases due to endogenous immune activation. The exact difference in immune cell response between synthetic and endogenous TLR7 triggers is only partly known. An understanding of these differences could aid in the development of new therapeutic agents and provide insights into autoimmune disease mechanisms. We therefore compared the stimulatory capacity of two TLR7 agonists, RNA-IC and a synthetic small molecule DSR-6434, on blood leucocytes, plasmacytoid dendritic cells (pDCs) and B cells from healthy individuals. IFN-α, IL-6, IL-8 and TNF levels were measured by immunoassays, and gene expression in pDCs was analysed by an expression array. DSR-6434 triggered 20-fold lower levels of IFN-α by pDCs, but higher production of IL-6, IL-8 and TNF, compared to RNA-IC. Furthermore, IFN-α and TNF production were increased with exogenous IFN-α2b priming, whereas IL-8 synthesis by B cells was reduced for both stimuli. Cocultivation of pDCs and B cells increased the RNA-IC-stimulated IFN-α and TNF levels, while only IL-6 production was enhanced in the DSR-6434-stimulated cocultures. When comparing pDCs stimulated with RNA-IC and DSR-6434, twelve genes were differentially expressed (log2 fold change >2, adjusted P-value <.05). In conclusion, RNA-IC, which mimics an endogenous TLR7 stimulator, and the synthetic TLR7 agonist DSR-6434 trigger distinct inflammatory profiles in immune cells. This demonstrates the importance of using relevant stimuli when targeting the TLR7 pathway for therapeutic purposes.


Subject(s)
Adenine/pharmacology , Antigen-Antibody Complex/pharmacology , B-Lymphocytes/immunology , Dendritic Cells/immunology , Multiprotein Complexes/pharmacology , RNA/pharmacology , Toll-Like Receptor 7/metabolism , Adenine/analogs & derivatives , Adenine/chemistry , Antigen-Antibody Complex/chemistry , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Gene Expression Profiling , Humans , Inflammation Mediators/metabolism , Lymphocyte Activation , Molecular Structure , Multiprotein Complexes/chemistry , RNA/chemistry , Toll-Like Receptor 7/agonists
8.
PLoS Genet ; 13(6): e1006820, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28640813

ABSTRACT

Sjögren's syndrome (SS) is a common, autoimmune exocrinopathy distinguished by keratoconjunctivitis sicca and xerostomia. Patients frequently develop serious complications including lymphoma, pulmonary dysfunction, neuropathy, vasculitis, and debilitating fatigue. Dysregulation of type I interferon (IFN) pathway is a prominent feature of SS and is correlated with increased autoantibody titers and disease severity. To identify genetic determinants of IFN pathway dysregulation in SS, we performed cis-expression quantitative trait locus (eQTL) analyses focusing on differentially expressed type I IFN-inducible transcripts identified through a transcriptome profiling study. Multiple cis-eQTLs were associated with transcript levels of 2'-5'-oligoadenylate synthetase 1 (OAS1) peaking at rs10774671 (PeQTL = 6.05 × 10-14). Association of rs10774671 with SS susceptibility was identified and confirmed through meta-analysis of two independent cohorts (Pmeta = 2.59 × 10-9; odds ratio = 0.75; 95% confidence interval = 0.66-0.86). The risk allele of rs10774671 shifts splicing of OAS1 from production of the p46 isoform to multiple alternative transcripts, including p42, p48, and p44. We found that the isoforms were differentially expressed within each genotype in controls and patients with and without autoantibodies. Furthermore, our results showed that the three alternatively spliced isoforms lacked translational response to type I IFN stimulation. The p48 and p44 isoforms also had impaired protein expression governed by the 3' end of the transcripts. The SS risk allele of rs10774671 has been shown by others to be associated with reduced OAS1 enzymatic activity and ability to clear viral infections, as well as reduced responsiveness to IFN treatment. Our results establish OAS1 as a risk locus for SS and support a potential role for defective viral clearance due to altered IFN response as a genetic pathophysiological basis of this complex autoimmune disease.


Subject(s)
2',5'-Oligoadenylate Synthetase/genetics , Interferon Type I/genetics , Quantitative Trait Loci/genetics , Sjogren's Syndrome/genetics , 2',5'-Oligoadenylate Synthetase/biosynthesis , Alleles , Alternative Splicing/genetics , Female , Gene Expression Regulation , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Interferon Type I/metabolism , Male , Sjogren's Syndrome/metabolism , Sjogren's Syndrome/pathology , Virus Diseases/genetics , Virus Diseases/virology
9.
Hum Genet ; 138(2): 141-150, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30707351

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.


Subject(s)
Genome, Human , Heterozygote , High-Throughput Nucleotide Sequencing , Lupus Erythematosus, Systemic/genetics , Models, Genetic , Mutation, Missense , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Risk Factors
10.
Ann Rheum Dis ; 78(10): 1363-1370, 2019 10.
Article in English | MEDLINE | ID: mdl-31300459

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.


Subject(s)
Citrullination/genetics , Endopeptidases/genetics , Extracellular Traps/genetics , Lupus Erythematosus, Systemic/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , 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
11.
Ann Rheum Dis ; 77(7): 1070-1077, 2018 07.
Article in English | MEDLINE | ID: mdl-29475858

ABSTRACT

OBJECTIVES: Genetic variants in the transcription factor STAT4 are associated with increased susceptibility to systemic lupus erythematosus (SLE) and a more severe disease phenotype. This study aimed to clarify how the SLE-associated intronic STAT4 risk allele rs7574865[T] affects the function of immune cells in SLE. METHODS: Peripheral blood mononuclear cells (PBMCs) were isolated from 52 genotyped patients with SLE. Phosphorylation of STAT4 (pSTAT4) and STAT1 (pSTAT1) in response to interferon (IFN)-α, IFN-γ or interleukin (IL)-12, total levels of STAT4, STAT1 and T-bet, and frequency of IFN-γ+ cells on IL-12 stimulation were determined by flow cytometry in subsets of immune cells before and after preactivation of cells with phytohaemagglutinin (PHA) and IL-2. Cellular responses and phenotypes were correlated to STAT4 risk allele carriership. Janus kinase inhibitors (JAKi) selective for TYK2 (TYK2i) or JAK2 (JAK2i) were evaluated for inhibition of IL-12 or IFN-γ-induced activation of SLE PBMCs. RESULTS: In resting PBMCs, the STAT4 risk allele was neither associated with total levels of STAT4 or STAT1, nor cytokine-induced pSTAT4 or pSTAT1. Following PHA/IL-2 activation, CD8+ T cells from STAT4 risk allele carriers displayed increased levels of STAT4 resulting in increased pSTAT4 in response to IL-12 and IFN-α, and an augmented IL-12-induced IFN-γ production in CD8+ and CD4+ T cells. The TYK2i and the JAK2i efficiently blocked IL-12 and IFN-γ-induced activation of PBMCs from STAT4 risk patients, respectively. CONCLUSIONS: T cells from patients with SLE carrying the STAT4 risk allele rs7574865[T] display an augmented response to IL-12 and IFN-α. This subset of patients may benefit from JAKi treatment.


Subject(s)
Gene Expression Regulation , Interleukin-12/pharmacology , Lupus Erythematosus, Systemic/genetics , STAT1 Transcription Factor/genetics , STAT4 Transcription Factor/genetics , Adult , Alleles , Cells, Cultured , Female , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/blood , Male , Phosphorylation , Sensitivity and Specificity , Signal Transduction
12.
Ann Rheum Dis ; 77(5): 736-743, 2018 05.
Article in English | MEDLINE | ID: mdl-29437559

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.


Subject(s)
DNA Methylation/genetics , Gene Expression Regulation/genetics , Lupus Erythematosus, Systemic/genetics , 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
13.
Ann Rheum Dis ; 77(7): 1063-1069, 2018 07.
Article in English | MEDLINE | ID: mdl-29514802

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.


Subject(s)
Arthritis, Rheumatoid/genetics , Cardiovascular Diseases/genetics , Genetic Predisposition to Disease/epidemiology , Lupus Erythematosus, Systemic/genetics , Polymorphism, Single Nucleotide/genetics , 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
14.
J Autoimmun ; 90: 28-38, 2018 06.
Article in English | MEDLINE | ID: mdl-29398253

ABSTRACT

Non-coding SNPs in the protein tyrosine phosphatase non-receptor type 2 (PTPN2) locus have been linked with several autoimmune diseases, including rheumatoid arthritis, type I diabetes, and inflammatory bowel disease. However, the functional consequences of these SNPs are poorly characterized. Herein, we show in blood cells that SNPs in the PTPN2 locus are highly correlated with DNA methylation levels at four CpG sites downstream of PTPN2 and expression levels of the long non-coding RNA (lncRNA) LINC01882 downstream of these CpG sites. We observed that LINC01882 is mainly expressed in T cells and that anti-CD3/CD28 activated naïve CD4+ T cells downregulate the expression of LINC01882. RNA sequencing analysis of LINC01882 knockdown in Jurkat T cells, using a combination of antisense oligonucleotides and RNA interference, revealed the upregulation of the transcription factor ZEB1 and kinase MAP2K4, both involved in IL-2 regulation. Overall, our data suggests the involvement of LINC01882 in T cell activation and hints towards an auxiliary role of these non-coding SNPs in autoimmunity associated with the PTPN2 locus.


Subject(s)
Autoimmune Diseases/genetics , CD4-Positive T-Lymphocytes/physiology , CpG Islands/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , RNA, Long Noncoding/genetics , Autoimmunity/genetics , DNA Methylation , Gene Knockdown Techniques , Humans , Jurkat Cells , Lymphocyte Activation , MAP Kinase Kinase 4/genetics , Polymorphism, Single Nucleotide , RNA, Small Interfering/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics
15.
Hum Mol Genet ; 24(12): 3571-81, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25779693

ABSTRACT

The type I interferon (IFN) system is persistently activated in systemic lupus erythematosus (SLE) and many other systemic autoimmune diseases. Studies have shown an association between SLE and several gene variants within the type I IFN system. We investigated whether single-nucleotide polymorphisms (SNPs) associated with SLE and other autoimmune diseases affect the IFN-α production in healthy individuals. Plasmacytoid dendritic cells (pDCs), B cells and NK cells were isolated from peripheral blood of healthy individuals and stimulated with RNA-containing immune complexes (ICs), herpes simplex virus (HSV) or the oligonucleotide ODN2216. IFN-α production by pDCs alone or in cocultures with B or NK cells was measured by an immunoassay. All donors were genotyped with the 200K ImmunoChip, and a 5 bp CGGGG length polymorphism in the IFN regulatory factor 5 gene (IRF5) was genotyped by PCR. We found associations between IFN-α production and 18-86 SNPs (P ≤ 0.001), depending on the combination of the stimulated cell types. However, only three of these associated SNPs were shared between the cell-type combinations. Several SNPs showed novel associations to the type I IFN system among all the associated SNPs, whereas some loci have been described earlier for their association with SLE. Furthermore, we found that the SLE-risk variant of the IRF5 CGGGG-indel was associated with lower IFN-α production. We conclude that the genetic variants affecting the IFN-α production highlight the intricate regulation of the type I IFN system and the importance of understanding the mechanisms behind the dysregulated type I IFN system in SLE.


Subject(s)
Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Genetic Loci , Inflammation/etiology , Inflammation/metabolism , Interferon-alpha/biosynthesis , Polymorphism, Genetic , Adult , Aged , Case-Control Studies , Female , Gene Regulatory Networks , Genotype , Humans , Interferon Regulatory Factors/genetics , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Male , Middle Aged , Polymorphism, Single Nucleotide , Young Adult
16.
Hum Genet ; 135(5): 485-497, 2016 May.
Article in English | MEDLINE | ID: mdl-26993500

ABSTRACT

Genome-wide association studies (GWAS) have identified a large number of disease-associated SNPs, but in few cases the functional variant and the gene it controls have been identified. To systematically identify candidate regulatory variants, we sequenced ENCODE cell lines and used public ChIP-seq data to look for transcription factors binding preferentially to one allele. We found 9962 candidate regulatory SNPs, of which 16 % were rare and showed evidence of larger functional effect than common ones. Functionally rare variants may explain divergent GWAS results between populations and are candidates for a partial explanation of the missing heritability. The majority of allele-specific variants (96 %) were specific to a cell type. Furthermore, by examining GWAS loci we found >400 allele-specific candidate SNPs, 141 of which were highly relevant in our cell types. Functionally validated SNPs support identification of an SNP in SYNGR1 which may expose to the risk of rheumatoid arthritis and primary biliary cirrhosis, as well as an SNP in the last intron of COG6 exposing to the risk of psoriasis. We propose that by repeating the ChIP-seq experiments of 20 selected transcription factors in three to ten people, the most common polymorphisms can be interrogated for allele-specific binding. Our strategy may help to remove the current bottleneck in functional annotation of the genome.


Subject(s)
Gene Expression Profiling , Genetic Predisposition to Disease , Neuroblastoma/genetics , Neuroblastoma/metabolism , Polymorphism, Single Nucleotide/genetics , Transcription Factors/metabolism , Alleles , Biomarkers , Genotype , Humans , K562 Cells , Protein Binding , Tumor Cells, Cultured
17.
Mol Med ; 22: 322-328, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27532898

ABSTRACT

OBJECTIVE: In rheumatoid arthritis (RA) several recent efforts have sought to discover means of predicting which patients would benefit from treatment. However, results have been discrepant with few successful replications. Our objective was to build a biobank with DNA, RNA and protein measurements to test the claim that the current state-of-the-art precision medicine will benefit RA patients. METHODS: We collected 451 blood samples from 61 healthy individuals and 185 RA patients initiating treatment, before treatment initiation and at a 3 month follow-up time. All samples were subjected to high-throughput RNA sequencing, DNA genotyping, extensive proteomics and flow cytometry measurements, as well as comprehensive clinical phenotyping. Literature review identified 2 proteins, 52 single-nucleotide polymorphisms (SNPs) and 72 gene-expression biomarkers that had previously been proposed as predictors of TNF inhibitor response (∆DAS28-CRP). RESULTS: From these published TNFi biomarkers we found that 2 protein, 2 SNP and 8 mRNA biomarkers could be replicated in the 59 TNF initiating patients. Combining these replicated biomarkers into a single signature we found that we could explain 51% of the variation in ∆DAS28-CRP. This corresponds to a sensitivity of 0.73 and specificity of 0.78 for the prediction of three month ∆DAS28-CRP better than -1.2. CONCLUSIONS: The COMBINE biobank is currently the largest collection of multi-omics data from RA patients with high potential for discovery and replication. Taking advantage of this we surveyed the current state-of-the-art of drug-response stratification in RA, and identified a small set of previously published biomarkers available in peripheral blood which predicts clinical response to TNF blockade in this independent cohort.

18.
Ann Rheum Dis ; 75(9): 1728-34, 2016 09.
Article in English | MEDLINE | ID: mdl-26493814

ABSTRACT

OBJECTIVES: Patients with systemic lupus erythematosus (SLE) have an ongoing interferon-α (IFN-α) production by plasmacytoid dendritic cells (pDCs). We investigated whether T cells can promote IFN-α production by pDCs. METHODS: Human pDCs were stimulated with immune complexes (ICs) containing U1 small nuclear ribonucleic proteins particles and SLE-IgG (RNA-IC) in the presence of T cells or T cell supernatants. T cells were activated by anti-CD3/CD28 antibodies or in a mixed leucocyte reaction. IFN-α and other cytokines were determined in culture supernatants or patient sera with immunoassays. The effect of interleukin (IL) 3 and granulocyte-macrophage-colony-stimulating factor (GM-CSF) on pDCs was examined by the use of antibodies, and the expression of CD80/CD86 was determined using flow cytometry. RESULTS: Activated T cells and supernatants from activated T cells increased IFN-α production by >20-fold. The stimulatory effect of T cell supernatants was reduced after depletion of GM-CSF (81%) or by blocking the GM-CSF receptor (55%-81%). Supernatant from activated T cells, furthermore, increased the frequency of CD80 and CD86 expressing pDCs stimulated with RNA-IC from 6% to 35% (p<0.05) and from 10% to 26% (p<0.01), respectively. Activated SLE T cells enhanced IFN-α production to the same extent as T cells from healthy individuals and a subset of patients with SLE had increased serum levels of GM-CSF. CONCLUSIONS: Activated T cells enhance IFN-α production by RNA-IC stimulated pDCs via GM-CSF and induce pDC maturation. Given the increased serum levels of GM-CSF in a subset of patients with SLE, these findings suggest that activated T cells may upregulate type I IFN production in SLE.


Subject(s)
Antigen-Antibody Complex/immunology , Dendritic Cells/immunology , Interferon-alpha/biosynthesis , RNA/immunology , T-Lymphocytes/immunology , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology
19.
Ann Rheum Dis ; 75(11): 2029-2036, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26857698

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.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Interferons/genetics , Sjogren's Syndrome/genetics , 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
20.
J Immunol ; 190(5): 2335-44, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23359498

ABSTRACT

Tyk2 belongs to the Janus protein tyrosine kinase family and is involved in signaling of immunoregulatory cytokines (type I and III IFNs, IL-6, IL-10, and IL-12 families) via its interaction with shared receptor subunits. Depending on the receptor complex, Tyk2 is coactivated with either Jak1 or Jak2, but a detailed molecular characterization of the interplay between the two enzymes is missing. In human populations, the Tyk2 gene presents high levels of genetic diversity with >100 nonsynonymous variants being detected. In this study, we characterized two rare Tyk2 variants, I684S and P1104A, which have been associated with susceptibility to autoimmune disease. Specifically, we measured their in vitro catalytic activity and their ability to mediate Stat activation in fibroblasts and genotyped B cell lines. Both variants were found to be catalytically impaired but rescued signaling in response to IFN-α/ß, IL-6, and IL-10. These data, coupled with functional study of an engineered Jak1 P1084A, support a model of nonhierarchical activation of Janus kinases in which one catalytically competent Jak is sufficient for signaling provided that its partner behaves as proper scaffold, even if inactive. Through the analysis of IFN-α and IFN-γ signaling in cells with different Jak1 P1084A levels, we also illustrate a context in which a hypomorphic Jak can hamper signaling in a cytokine-specific manner. Given the multitude of Tyk2-activating cytokines, the cell context-dependent requirement for Tyk2 and the catalytic defect of the two disease-associated variants studied in this paper, we predict that these alleles are functionally significant in complex immune disorders.


Subject(s)
Autoimmune Diseases/genetics , B-Lymphocytes/metabolism , Janus Kinase 1/genetics , Polymorphism, Single Nucleotide , Signal Transduction/immunology , TYK2 Kinase/genetics , Alleles , Amino Acid Sequence , Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Line, Transformed , Gene Expression , Genetic Predisposition to Disease , Genetic Vectors , Herpesvirus 4, Human/genetics , Humans , Interferon Type I/immunology , Interferon Type I/pharmacology , Interleukin-10/immunology , Interleukin-10/pharmacology , Interleukin-6/immunology , Interleukin-6/pharmacology , Janus Kinase 1/immunology , Janus Kinase 1/metabolism , Models, Molecular , Molecular Sequence Data , Plasmids , TYK2 Kinase/immunology , TYK2 Kinase/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL