Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
BMC Biol ; 21(1): 156, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37443005

ABSTRACT

BACKGROUND: Prime editing (PE) is the most recent gene editing technology able to introduce targeted alterations to the genome, including single base pair changes, small insertions, and deletions. Several improvements to the PE machinery have been made in the past few years, and these have been tested in a range of model systems including immortalized cell lines, stem cells, and animal models. While double nicking RNA (dncRNA) PE systems PE3 and PE5 currently show the highest editing rates, they come with reduced accuracy as undesired indels or SNVs arise at edited loci. Here, we aimed to improve single ncRNA (sncRNA) systems PE2 and PE4max by generating novel all-in-one (pAIO) plasmids driven by an EF-1α promoter, which is especially suitable for human-induced pluripotent stem cell (hiPSC) models. RESULTS: pAIO-EF1α-PE2 and pAIO-EF1α-PE4max were used to edit the voltage gated potassium channel gene KCNQ2 and voltage gated sodium channel gene SCN1A. Two clinically relevant mutations were corrected using pAIO-EF1α-PE2 including the homozygous truncating SCN1A R612* variant in HEK293T cells and the heterozygous gain-of-function KCNQ2 R201C variant in patient-derived hiPSC. We show that sncRNA PE yielded detectable editing rates in hiPSC ranging between 6.4% and 9.8%, which was further increased to 41% after a GFP-based fluorescence-activated cell sorting (FACS) cell sorting step. Furthermore, we show that selecting the high GFP expressing population improved editing efficiencies up to 3.2-fold compared to the low GFP expressing population, demonstrating that not only delivery but also the number of copies of the PE enzyme and/or pegRNA per cell are important for efficient editing. Edit rates were not improved when an additional silent protospacer-adjacent motif (PAM)-removing alteration was introduced in hiPSC at the target locus. Finally, there were no genome-wide off-target effects using pAIO-EF1α-PE2 and no off-target editing activity near the edit locus highlighting the accuracy of snc prime editors. CONCLUSION: Taken together, our study shows an improved efficacy of EF-1α driven sncRNA pAIO-PE plasmids in hiPSC reaching high editing rates, especially after FACS sorting. Optimizing these sncRNA PE systems is of high value when considering future therapeutic in vivo use, where accuracy will be extremely important.


Subject(s)
CRISPR-Cas Systems , RNA, Small Untranslated , Animals , Humans , HEK293 Cells , Peptide Elongation Factor 1/genetics , Plasmids/genetics , KCNQ2 Potassium Channel/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics
2.
Eur J Neurol ; 26(6): 856-864, 2019 06.
Article in English | MEDLINE | ID: mdl-30223294

ABSTRACT

BACKGROUND AND PURPOSE: Juvenile myoclonic epilepsy (JME) is a common epilepsy syndrome for which treatment response is generally assumed to be good. We aimed to determine the prevalence and prognostic risk factors for refractoriness of JME. METHODS: We systematically searched PubMed and EMBASE and included 43 eligible studies, reporting seizure outcome after antiepileptic drug (AED) treatment in JME cohorts. We defined refractory JME as persistence of any seizure despite AED treatment and performed a random-effects meta-analysis to assess the prevalence of refractory JME and of seizure recurrence after AED withdrawal in individuals with well-controlled seizures. Studies reporting potential prognostic risk factors in relation to seizure outcome were included for subsequent meta-analysis of risk factors for refractoriness. RESULTS: Overall, 35% (95% confidence interval, 29-41%) of individuals (n = 3311) were refractory. There was marked heterogeneity between studies. Seizures recurred in 78% (95% confidence interval, 52-94%) of individuals who attempted to withdraw from treatment after a period of seizure freedom (n = 246). Seizure outcome by publication year suggested that prognosis did not improve over time. Meta-analysis suggested six variables as prognostic factors for refractoriness, i.e. having three seizure types, absence seizures, psychiatric comorbidities, earlier age at seizure onset, history of childhood absence epilepsy and praxis-induced seizures. CONCLUSION: One-third of people with JME were refractory, which is a higher prevalence than expected. Risk factors were identified and can be used to guide treatment and counselling of people with JME.


Subject(s)
Myoclonic Epilepsy, Juvenile/epidemiology , Anticonvulsants/therapeutic use , Humans , Myoclonic Epilepsy, Juvenile/drug therapy , Myoclonic Epilepsy, Juvenile/etiology , Prevalence , Prognosis , Risk Factors
3.
Genes Immun ; 18(3): 176-183, 2017 09.
Article in English | MEDLINE | ID: mdl-28794505

ABSTRACT

Tolerogenic dendritic cells (tolDCs) are assessed as immunomodulatory adjuvants to regulate autoimmunity. The underlying gene expression endorsing their regulatory features remains ill-defined. Using deep mRNA sequencing, we compared transcriptomes of 1,25-dihydroxyvitaminD3/dexametasone-modulated tolDCs with that of non-modulated mature inflammatory DCs (mDCs). Differentially expressed genes controlled cellular interactions, metabolic pathways and endorse tolDCs with the capacity to regulate cell activation through nutrient and signal deprivation, collectively gearing tolDCs into tolerogenic immune regulators. Gene expression differences correlated with protein expression, designating low CD86 and high CD52 on the cell surface as superior discriminators between tolDCs and mDCs. Of 37 candidate genes conferring risk to developing type 1 diabetes (T1D), 11 genes differentially expressed in tolDCs and mDCs regulated immune response and antigen-presenting activity. Differential-expressed transcripts of candidate risk loci for T1D suggest a role of these 'risk genes' in immune regulation, which targeting may modulate the genetic contribution to autoimmunity.


Subject(s)
Autoimmunity/genetics , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/genetics , Immune Tolerance/genetics , Transcriptome , Antigen Presentation/genetics , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , CD52 Antigen/genetics , CD52 Antigen/metabolism , Calcitriol/pharmacology , Cell Line , Cells, Cultured , Dendritic Cells/drug effects , Dexamethasone/pharmacology , Diabetes Mellitus, Type 1/immunology , Humans
4.
Neurogenetics ; 18(3): 147-153, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28669061

ABSTRACT

Pathogenic variants in the PCDH19 gene are associated with epilepsy, intellectual disability (ID) and behavioural disturbances. Only heterozygous females and mosaic males are affected, likely due to a disease mechanism named cellular interference. Until now, only four affected mosaic male patients have been described in literature. Here, we report five additional male patients, of which four are older than the oldest patient reported so far. All reported patients were selected for genetic testing because of developmental delay and/or epilepsy. Custom-targeted next generation sequencing gene panels for epilepsy genes were used. Clinical data were collected from medical records. All patients were mosaic in blood for likely pathogenic variants in the PCDH19 gene. In most, clinical features were very similar to the female phenotype, with normal development before seizure onset, which occurred between 5 and 10 months of age, clustering of seizures and sensitivity to fever. Four out of five patients had mild to severe ID and behavioural problems. We reaffirm the similarity between male and female PCDH19-related phenotypes, now also in a later phase of the disorder (ages 10-14 years).


Subject(s)
Cadherins/genetics , Epilepsy/genetics , Genetic Predisposition to Disease , Intellectual Disability/genetics , Mutation/genetics , Female , Heterozygote , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Protocadherins , Seizures/complications , Sex Factors
5.
Genes Immun ; 17(6): 342-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27467285

ABSTRACT

Autoreactive CD8(+) T cells recognizing autoantigens expressed by pancreatic islets lead to the destruction of insulin-producing beta cells in type 1 diabetes (T1D), but these T cells also occur in healthy subjects. We tested the hypothesis that uncontrolled expansion of diabetogenic T cells in patients occurs, resulting from failure to activate apoptosis. We compared function, transcriptome and epigenetic regulation thereof in relation with fate upon repeated exposure to islet-autoantigen of islet autoreactive T cells from healthy and type 1 diabetic donors with identical islet epitope specificity and HLA-A2 restriction. Patient's T cells proliferated exponentially, whereas those of non-diabetic origin succumbed to cell death. Transcriptome analysis revealed reduced expression of TRAIL, TRAIL-R2, FAS and FASLG (members of the extrinsic apoptosis pathway) in patient-derived compared with healthy donor-derived T cells. This was mirrored by increased expression of microRNAs predicted to regulate these particular genes, namely miR-98, miR-23b and miR-590-5p. Gene-specific targeting by these microRNAs was confirmed using dual-luciferase reporter assays. Finally, transfection of these microRNAs into primary T cells reduced FAS and TRAIL mRNA underscoring their functional relevance. We propose that repression of pro-apoptotic pathways by microRNAs contributes to unrestricted expansion of diabetogenic cytotoxic T cells, implicating microRNA-mediated gene silencing in islet autoimmunity in T1D.


Subject(s)
Apoptosis , Autoimmunity , CD8-Positive T-Lymphocytes/physiology , Diabetes Mellitus, Type 1/immunology , MicroRNAs/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , fas Receptor/metabolism , Apoptosis/genetics , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cell Survival , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Humans , Insulin-Secreting Cells , Transcriptome
6.
Eur J Neurol ; 23(9): 1393-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27333872

ABSTRACT

The use of carbamazepine (CBZ) and oxcarbazepine (OXC) as first-line antiepileptic drugs in the treatment of focal epilepsy is limited by hyponatremia, a known adverse effect. Hyponatremia occurs in up to half of people taking CBZ or OXC and, although often assumed to be asymptomatic, it can lead to symptoms ranging from unsteadiness and mild confusion to seizures and coma. Hyponatremia is probably due to the antidiuretic properties of CBZ and OXC that are, at least partly, explained by stimulation of the vasopressin 2 receptor/aquaporin 2 pathway. No known genetic risk variants for CBZ- and OXC-induced hyponatremia exist, but likely candidate genes are part of the vasopressin water reabsorption pathway.


Subject(s)
Anticonvulsants/adverse effects , Carbamazepine/analogs & derivatives , Carbamazepine/adverse effects , Hyponatremia/chemically induced , Animals , Humans , Hyponatremia/epidemiology , Hyponatremia/genetics , Hyponatremia/physiopathology , Oxcarbazepine , Pharmacogenetics
7.
J Neurol Neurosurg Psychiatry ; 87(7): 693-6, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26272027

ABSTRACT

BACKGROUND: Case-control studies have reported multiple genetic loci to be associated with sporadic brain arteriovenous malformations (AVMs) but most of these have not been replicated in independent populations. The aim of this study was to find additional evidence for these reported associations and perform a meta-analysis including all previously published results. METHODS: We included 167 Dutch patients and 1038 Dutch controls. Case genotyping was performed by KASPar assays. Controls had been previously genotyped with a genome wide single nucleotide polymorphisms (SNP) array. Differences in genotype frequencies between cases and controls were estimated by χ(2) testing in Plink V.1.07. Meta-analysis was performed in RevMan V.5.3. RESULTS: In our case-control study we found no significant association with brain AVM (BAVM) for previously discovered SNPs near ANGPTL4, IL-1ß, GPR124, VEGFA and MMP-3. The meta-analysis revealed a statistically significant association with BAVMs for the polymorphism rs11672433 near ANGPTL4 (OR 1.39; 95% CI 1.10 to 1.75, p value 0.005). CONCLUSIONS: The results of this study support a role for the previously identified SNP near ANGPTL4 in the pathogenesis of AVMs. Previously found associations with SNPs near IL-1ß, GPR124, VEGFA and MMP-3 genes could not be substantiated in our replication cohort or in the meta-analysis.


Subject(s)
Genetic Loci/genetics , Genetic Predisposition to Disease/genetics , Intracranial Arteriovenous Malformations/genetics , Adult , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Case-Control Studies , Female , Gene Frequency/genetics , Genome-Wide Association Study , Genotype , Humans , Interleukin-1beta/genetics , Male , Matrix Metalloproteinase 3/genetics , Middle Aged , Polymorphism, Single Nucleotide/genetics , Receptors, G-Protein-Coupled/genetics , Reproducibility of Results , Vascular Endothelial Growth Factor A/genetics
8.
Genes Immun ; 17(1): 75-8, 2016.
Article in English | MEDLINE | ID: mdl-26656450

ABSTRACT

Cytotoxic T-lymphocyte-associated protein 4 (CTLA4) is a protein receptor that downregulates the immune system. CTLA4 gene variants associate with various autoimmune diseases, including type 1 diabetes. Fine mapping of the genetic risk has shown that the genomic region near CTLA4 marked by the single-nucleotide polymorphism (SNP) CT60A/G (rs3087243) acts as a susceptibility factor. Yet, the functional basis for the increased susceptibility conferred by rs3087243 remains unclear. We demonstrate that the length of the dinucleotide (AT)n repeat within the CTLA4 3' untranslated region (3'UTR) strongly associates with the risk of SNP CT60A/G (P<6.5 × 10(-72)). Genomic (AT)n repeat length inversely correlated with CTLA4 messenger RNA (mRNA) and protein levels in islet autoreactive T-cell lines. Transfer of a long (AT)n element into T cells lead to a reduction of mRNA compared to a short (AT)n element. Thus, this study provides evidence for a role of the CTLA4 3'UTR (AT)n repeat in the increased genetic risk for islet autoimmunity associated with the CTLA4 locus.


Subject(s)
3' Untranslated Regions , CTLA-4 Antigen/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Dinucleotide Repeats , Genetic Predisposition to Disease , T-Lymphocytes/immunology , Humans , RNA Processing, Post-Transcriptional
9.
J Neurol Neurosurg Psychiatry ; 86(5): 524-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25053769

ABSTRACT

BACKGROUND: In genome-wide association studies (GWAS) five putative risk loci are associated with intracranial aneurysm. As brain arteriovenous malformations (AVM) and intracranial aneurysms are both intracranial vascular diseases and AVMs often have associated aneurysms, we investigated whether these loci are also associated with sporadic brain AVM. METHODS: We included 506 patients (168 Dutch, 338 American) and 1548 controls, all Caucasians. Controls had been recruited as part of previous GWAS. Dutch patients were genotyped by KASPar assay and US patients by Affymetrix SNP 6.0 array. Associations in each cohort were tested by univariable logistic regression modelling, with subgroup analysis in 205 American cases with aneurysm data. Meta-analysis was performed by a Mantel-Haenszel fixed-effect method. RESULTS: In the Dutch cohort none of the single nucleotide polymorphisms (SNPs) were associated with AVMs. In the American cohort, genotyped SNPs near SOX-17 (OR 0.74; 95% CI 0.56-0.98), RBBP8 (OR 0.76; 95% CI 0.62-0.94) and an imputed SNP near CDKN2B-AS1 (OR 0.79; 95% CI 0.64-0.98) were significantly associated with AVM. The association with SNPs near SOX-17 and CDKN2B-AS1 but not RBBP8 were strongest in patients with AVM with associated aneurysms. In the meta-analysis we found no significant associations between allele frequencies and AVM occurrence, but rs9298506, near SOX-17 approached statistical significance (OR 0.77; 95% CI 0.57-1.03, p=0.08). CONCLUSIONS: Our meta-analysis of two Caucasian cohorts did not show an association between five aneurysm-associated loci and sporadic brain AVM. Possible involvement of SOX-17 and RBBP8, genes involved in cell cycle progression, deserves further investigation.


Subject(s)
Genetic Predisposition to Disease/genetics , Intracranial Aneurysm/complications , Intracranial Aneurysm/genetics , Intracranial Arteriovenous Malformations/complications , Intracranial Arteriovenous Malformations/genetics , Carrier Proteins/genetics , Case-Control Studies , Cation Transport Proteins , Cyclins/genetics , Endodeoxyribonucleases , GTPase-Activating Proteins , Gene Frequency/genetics , Genome-Wide Association Study , Humans , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , RNA, Long Noncoding/genetics , SOXF Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , White People/genetics
10.
Ann Rheum Dis ; 74(3): e15, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24532677

ABSTRACT

INTRODUCTION: Rheumatoid arthritis (RA) patients can be classified based on presence or absence of anticitrullinated peptide antibodies (ACPA) in their serum. This heterogeneity among patients may reflect important biological differences underlying the disease process. To date, the majority of genetic studies have focused on the ACPA-positive group. Therefore, our goal was to analyse the genetic risk factors that contribute to ACPA-negative RA. METHODS: We performed a large-scale genome-wide association study (GWAS) in three Caucasian European cohorts comprising 1148 ACPA-negative RA patients and 6008 controls. All patients were screened using the Illumina Human Cyto-12 chip, and controls were genotyped using different genome-wide platforms. Population-independent analyses were carried out by means of logistic regression. Meta-analysis with previously published data was performed as follow-up for selected signals (reaching a total of 1922 ACPA-negative RA patients and 7087 controls). Imputation of classical HLA alleles, amino acid residues and single nucleotide polymorphisms was undertaken. RESULTS: The combined analysis of the studied cohorts resulted in identification of a peak of association in the HLA-region and several suggestive non-HLA associations. Meta-analysis with previous reports confirmed the association of the HLA region with this subset and an observed association in the CLYBL locus remained suggestive. The imputation and deep interrogation of the HLA region led to identification of a two amino acid model (HLA-B at position 9 and HLA-DRB1 at position 11) that accounted for the observed genome-wide associations in this region. CONCLUSIONS: Our study shed light on the influence of the HLA region in ACPA-negative RA and identified a suggestive risk locus for this condition.


Subject(s)
Arthritis, Rheumatoid/genetics , HLA Antigens/genetics , Alleles , Arthritis, Rheumatoid/immunology , Autoantibodies/immunology , Case-Control Studies , Citrulline/immunology , Genome-Wide Association Study , HLA Antigens/immunology , HLA-B Antigens/genetics , HLA-DRB1 Chains/genetics , Humans , Logistic Models , Peptides/immunology , Polymorphism, Single Nucleotide , Principal Component Analysis , White People/genetics
11.
J Thromb Haemost ; 11(4): 715-23, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23387557

ABSTRACT

BACKGROUND: Normal protein C (PC) plasma levels range widely in the general population. Factors influencing normal PC levels are thought to influence the risk of venous thrombosis. Little is known about the underlying genetic variants. OBJECTIVES: We performed a genome scan of normal PC levels to identify genes that regulate normal PC levels. PATIENTS/METHODS: We performed a variance components linkage analysis for normal PC levels in 275 individuals from a single, large family. We then sequenced candidate genes under the identified linkage peak in eight family members: four with high and four with low, but normal, PC levels. For variants showing a difference in carriers between those with high and low PC levels, we re-evaluated linkage in the 275 family members conditional on the measured genotype effect. Genotype-specific mean PC levels were determined using likelihood analysis. Findings were replicated in the Leiden Thrombophilia Study (LETS). RESULTS: We identified a quantitative trait locus at chromosome 5q14.1 affecting normal PC plasma level variability. Next-generation sequencing of 113 candidate genes under the linkage peak revealed four SNPs in BHMT2, ACOT12, SSBP2 and XRCC4, which significantly increased PC levels in our thrombophilic family, but not in LETS. CONCLUSIONS: We identified four genes at chromosome 5q14.1 that might influence normal PC levels. BHMT2 seems the most likely candidate to regulate PC levels via homocysteine, a competitive inhibitor to thrombin. Failure to replicate our findings in LETS might be due to differences between the studies in genetic background and linkage disequilibrium patterns.


Subject(s)
Polymorphism, Single Nucleotide , Protein C/metabolism , Case-Control Studies , Genetic Linkage , Humans , Protein C/genetics
12.
Neurogenetics ; 14(1): 23-34, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23334464

ABSTRACT

Epilepsy and mental retardation limited to females (EFMR), caused by PCDH19 mutations, has a variable clinical expression that needs further exploration. Onset of epilepsy may be provoked by fever and can resemble Dravet syndrome. Furthermore, transmitting males have no seizures, but are reported to have rigid personalities suggesting possible autism spectrum disorders (ASD). Therefore, this study aimed to determine the phenotypic spectrum associated with PCDH19 mutations in Dravet-like and EFMR female patients and in males with ASD. We screened 120 females suffering from Dravet-like epilepsy, 136 females with EFMR features and 20 males with ASD. Phenotypes and genotypes of the PCDH19 mutation carriers were compared with those of 125 females with EFMR reported in the literature. We report 15 additional patients with a PCDH19 mutation. Review of clinical data of all reported patients showed that the clinical picture of EFMR is heterogeneous, but epilepsy onset in infancy, fever sensitivity and occurrence of seizures in clusters are key features. Seizures remit in the majority of patients during teenage years. Intellectual disability and behavioural disturbances are common. Fifty percent of all mutations are missense mutations, located in the extracellular domains only. Truncating mutations have been identified in all protein domains. One ASD proband carried one missense mutation predicted to have a deleterious effect, suggesting that ASD in males can be associated with PCDH19 mutations.


Subject(s)
Cadherins/genetics , Child Development Disorders, Pervasive/epidemiology , Child Development Disorders, Pervasive/genetics , Epilepsy/epidemiology , Epilepsy/genetics , Mutation/physiology , Adolescent , Cadherins/physiology , Child , Child Development Disorders, Pervasive/complications , Child, Preschool , Cohort Studies , Epilepsies, Myoclonic/epidemiology , Epilepsies, Myoclonic/genetics , Epilepsy/complications , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Intellectual Disability/complications , Intellectual Disability/epidemiology , Intellectual Disability/genetics , Male , Penetrance , Protocadherins , Sex Characteristics , Syndrome
13.
Genes Immun ; 14(1): 58-61, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22932817

ABSTRACT

The genetic variation causal for predisposition to type 1 diabetes (T1D) remains unidentified for the majority of known T1D risk loci. MicroRNAs function as post-transcriptional gene regulators by targeting microRNA-binding sites in the 3' untranslated regions (UTR) of mRNA. Genetic variation within the 3'-UTR of T1D-associated genes may contribute to T1D development by altering microRNA-mediated gene regulation. In silico analysis of variable sites predicted altered microRNA binding in established T1D loci. Functional implications were assessed for variable sites in the 3'-UTR of T1D candidate risk genes CTLA4 and IL10, both involved in immune regulation. We confirmed that in these genes 3'-UTR variation either disrupted or introduced a microRNA-binding site, affecting the repressive capacity of miR-302a* and miR-523, respectively. Our study points to the potential of 3'-UTR variation to affect T1D pathogenesis by altering post-transcriptional gene regulation by microRNAs.


Subject(s)
CTLA-4 Antigen/genetics , Diabetes Mellitus, Type 1/genetics , Interleukin-10/genetics , MicroRNAs/metabolism , Polymorphism, Single Nucleotide , 3' Untranslated Regions/genetics , Binding Sites , CTLA-4 Antigen/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , HEK293 Cells , Humans , Interleukin-10/metabolism , MicroRNAs/genetics
14.
Ann Rheum Dis ; 71(10): 1651-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22440823

ABSTRACT

BACKGROUND: Interleukin (IL)-15 levels are increased in serum, synovium and bone marrow of patients with rheumatoid arthritis (RA). IL-15 influences both the innate and the adaptive immune response; its major role is activation and proliferation of T cells. There are also emerging data that IL-15 affects osteoclastogenesis. The authors investigated the association of genetic variants in IL15 with the rate of joint destruction in RA. METHOD: 1418 patients with 4885 x-ray sets of both hands and feet of four independent data sets were studied. First, explorative analyses were performed on 600 patients with early RA enrolled in the Leiden Early Arthritis Clinic. Twenty-five single-nucleotide polymorphisms (SNPs) tagging IL-15 were tested. Second, SNPs with significant associations in the explorative phase were genotyped in data sets from Groningen, Sheffield and Lund. In each data set, the relative increase of the progression rate per year in the presence of a genotype was assessed. Subsequently, data were summarised in an inverse weighting meta-analysis. RESULTS: Five SNPs were significantly associated with rate of joint destruction in phase 1 and typed in the other data sets. Patients homozygous for rs7667746, rs7665842, rs2322182, rs6821171 and rs4371699 had respectively 0.94-, 1.04-, 1.09-, 1.09- and 1.09-fold rate of joint destruction compared to other patients (p=4.0×10(-6), p=3.8×10(-4), p=5.0×10(-3), p=5.0×10(-3) and p=9.4×10(-3)). DISCUSSION: Independent replication was not obtained, possibly due to insufficient power. Meta-analyses of all data sets combined resulted in significant results for four SNPs (rs7667746, p<0.001; rs7665842, p<0.001; rs4371699, p=0.01; rs6821171, p=0.01). These SNPs were also significant after correction for multiple testing. CONCLUSION: Genetic variants in IL-15 are associated with progression of joint destruction in RA.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Genetic Predisposition to Disease/genetics , Interleukin-15/genetics , Arthritis, Rheumatoid/diagnostic imaging , Cohort Studies , Disease Progression , Female , Foot/diagnostic imaging , Foot/pathology , Genotype , Hand/diagnostic imaging , Hand/pathology , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Radiography
15.
Genes Immun ; 13(2): 191-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22012429

ABSTRACT

Regulatory T cells (T(regs)) are crucial in the maintenance of the immune tolerance and seem to have an important role in systemic sclerosis (SSc). The interleukin 2 receptor α (IL2RA) is an important T(reg) marker, and polymorphisms of IL2RA gene are associated with a number of autoimmune diseases. Therefore, we aimed to investigate for the first time the association of the IL2RA locus in SSc. For this purpose, a total of 3023 SSc patients and 2735 matched healthy controls, from six European Caucasian cohorts, were genotyped for the IL2RA gene variants rs11594656, rs2104286 and rs12722495 using the TaqMan allelic discrimination technology. The overall meta-analysis reached statistical significance when the three polymorphisms were tested for association with SSc, the limited subtype (lcSSc) and anti-centromere auto-antibodies (ACAs). However, no significant P-values were obtained when the ACA-positive patients were removed from the SSc and lcSSc groups, suggesting that these associations rely on ACA positivity. The strongest association signal with ACA production was detected for rs2104286 (P(FDR)=2.07 × 10(-4), odds ratio=1.30 (1.14-1.47)). The associations of rs11594656 and rs12722495 were lost after conditioning to rs2104286, and allelic combination tests did not evidence a combined effect, indicating that rs2104286 best described the association between IL2RA and ACA presence in SSc.


Subject(s)
Autoimmune Diseases/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Scleroderma, Systemic/genetics , Adult , Autoimmune Diseases/immunology , Genetic Loci , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Middle Aged , Polymorphism, Single Nucleotide , Scleroderma, Systemic/immunology
16.
Diabet Med ; 29(6): 734-41, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22150609

ABSTRACT

AIMS: The relation of disease progression and age, serum interleukin 10 (IL-10) and interferon gamma (IFNγ) and their genetic correlates were studied in paediatric patients with newly diagnosed Type 1 diabetes. METHODS: Two hundred and twenty-seven patients from the Hvidoere Study Group were classified in four different progression groups as assessed by change in stimulated C-peptide from 1 to 6 months. CA repeat variants of the IL-10 and IFNγ gene were genotyped and serum levels of IL-10 and IFNγ were measured at 1, 6 and 12 months. RESULTS: IL-10 decreased (P < 0.001) by 7.7% (1 month), 10.4% (6 months) and 8.6% (12 months) per year increase in age of child, while a twofold higher C-peptide concentration at 1 month (p = 0.06), 6 months (P = 0.0003) and 12 months (P = 0.02) was associated with 9.7%, 18.6% and 9.7% lower IL-10 levels, independent of each other. IL-10 concentrations did not associate with the disease progression groups. By contrast, IFNγ concentrations differed between the four progression groups at 6 and 12 months (P = 0.02 and P = 0.01, respectively); patients with rapid progressing disease had the highest levels at both time points. Distribution of IL-10 and IFNγ genotypes was equal among patients from the progression groups. CONCLUSION: IL-10 serum levels associate inversely with age and C-peptide. As age and C-peptide also associate, a triangular association is proposed. Genetic influence on IL-10 production seems to be masked by distinct disease mechanisms. Increased serum IFNγ concentrations associate with rapid disease progression. Functional genetic variants do not associate with a single progression pattern group, implying that disease processes override genetically predisposed cytokine production.


Subject(s)
Aging/blood , C-Peptide/blood , Diabetes Mellitus, Type 1/blood , Interferon-gamma/blood , Interleukin-10/blood , Adolescent , Aging/genetics , Biomarkers/blood , C-Peptide/genetics , Child , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/physiopathology , Disease Progression , Fasting , Female , Genetic Predisposition to Disease , Humans , Infant , Interferon-gamma/genetics , Interleukin-10/genetics , Male
17.
Genes Immun ; 12(6): 415-27, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21562577

ABSTRACT

Human leukocyte antigen (HLA) class II haplotypes are established risk factors in type 1 diabetes (T1D). The heterozygous DQ2/8 genotype confers the highest risk, whereas the DQ6/8 genotype is protective. We hypothesized that DQ2/8 trans-molecules composed of α and ß chains from DQ2 and DQ8 express unique ß-cell epitopes, whereas DQ6 may interfere with peptide binding to DQ8. Here we show that a single insulin epitope (InsB13-21) within the T1D prototype antigenic InsB6-22 peptide can bind to both cis- and trans-dimers, although these molecules display different peptide binding patterns. DQ6 binds a distinct insulin epitope (InsB6-14). The phenotype of DQ8-restricted T cells from a T1D patient changed from proinflammatory to anti-inflammatory in the presence of DQ6. Our data provide new insights into both susceptible and protective mechanism of DQ, where protecting HLA molecules bind autoantigens in a different (competing) binding register leading to 'epitope stealing', thereby inducing a regulatory, rather than a pathogenic immune response.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , HLA-DQ Antigens/genetics , Islets of Langerhans/immunology , Adolescent , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Epitopes, B-Lymphocyte/immunology , Genetic Predisposition to Disease , Heterozygote , Homozygote , Humans , Insulin/genetics , Male , Protein Binding , Syndecans/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymosin/metabolism
18.
Tissue Antigens ; 77(4): 338-40, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21388358

ABSTRACT

An extreme genetic risk for type 1 diabetes (T1D) was reported for DR3/4-DQ8 siblings sharing both extended human leukocyte antigen (HLA) haplotypes identical-by-descent (IBD) with their diabetic proband. We attempted to replicate this finding in our prospective Dutch T1D cohort and in families from the Type 1 Diabetes Genetics Consortium (T1DGC). Only 2 of the 14 Dutch siblings, sharing both DR3-DQ2/DR4-DQ8 haplotypes IBD with their diabetic proband, developed T1D in a 12-year follow-up period. No differential sharing of HLA haplotypes or significant transmission distortion in parents homozygous for HLA risk alleles was found in T1DGC material. Therefore, we could not confirm the reported extreme risk for T1D, suggesting that the risk conferred by other HLA complex loci is moderate.


Subject(s)
Alleles , Diabetes Mellitus, Type 1/genetics , HLA-DR3 Antigen/genetics , HLA-DR4 Antigen/genetics , Siblings , Adolescent , Child , Cohort Studies , Female , Genetic Predisposition to Disease , HLA-DQ Antigens , Haplotypes , Humans , Male , Netherlands , Risk Factors
20.
Epilepsy Res ; 89(2-3): 286-94, 2010 May.
Article in English | MEDLINE | ID: mdl-20153606

ABSTRACT

Photoparoxysmal response (PPR) is considered to be a risk factor for idiopathic generalised epilepsy (IGE) and it has a strong genetic basis. Two genome-wide linkage studies have been published before and they identified loci for PPR at 6p21, 7q32, 13q13, 13q31 and 16p13. Here we combine these studies, augmented with additional families, in a mega-analysis of 100 families. Non-parametric linkage analysis identified three suggestive peaks for photosensitivity, two of which are novel (5q35.3 and 8q21.13) and one has been found before (16p13.3). We found no evidence for linkage at four previously detected loci (6p21, 7q32, 13q13 and 13q31). Our results suggest that the different family data sets are not linked to a shared locus. Detailed analysis showed that the peak at 16p13 was mainly supported by a single subset of families, while the peaks at 5q35 and 8q21 had weak support from multiple subsets. Family studies clearly support the role of PPR as a risk factor for IGE. This mega-analysis shows that distinct loci seem to be linked to subsets of PPR-positive families that may differ in subtle clinical phenotypes or geographic origin. Further linkage studies of PPR should therefore include in-depth phenotyping to make appropriate subsets and increase genetic homogeneity.


Subject(s)
Epilepsy, Reflex/genetics , Genetic Linkage/genetics , Genome, Human/genetics , Chromosome Mapping/methods , Chromosomes, Human, Pair 16/genetics , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 8/genetics , Female , Genetic Predisposition to Disease , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...