Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Nat Rev Drug Discov ; 23(7): 501-524, 2024 07.
Article in English | MEDLINE | ID: mdl-38839912

ABSTRACT

Despite major progress in the treatment of autoimmune diseases in the past two decades, most therapies do not cure disease and can be associated with increased risk of infection through broad suppression of the immune system. However, advances in understanding the causes of autoimmune disease and clinical data from novel therapeutic modalities such as chimeric antigen receptor T cell therapies provide evidence that it may be possible to re-establish immune homeostasis and, potentially, prolong remission or even cure autoimmune diseases. Here, we propose a 'sequential immunotherapy' framework for immune system modulation to help achieve this ambitious goal. This framework encompasses three steps: controlling inflammation; resetting the immune system through elimination of pathogenic immune memory cells; and promoting and maintaining immune homeostasis via immune regulatory agents and tissue repair. We discuss existing drugs and those in development for each of the three steps. We also highlight the importance of causal human biology in identifying and prioritizing novel immunotherapeutic strategies as well as informing their application in specific patient subsets, enabling precision medicine approaches that have the potential to transform clinical care.


Subject(s)
Autoimmune Diseases , Autoimmunity , Immunotherapy , Humans , Immunotherapy/methods , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Autoimmunity/immunology , Animals , Precision Medicine/methods
3.
Nat Protoc ; 14(12): 3426-3444, 2019 12.
Article in English | MEDLINE | ID: mdl-31748751

ABSTRACT

Phenotypes are the foundation for clinical and genetic studies of disease risk and outcomes. The growth of biobanks linked to electronic medical record (EMR) data has both facilitated and increased the demand for efficient, accurate, and robust approaches for phenotyping millions of patients. Challenges to phenotyping with EMR data include variation in the accuracy of codes, as well as the high level of manual input required to identify features for the algorithm and to obtain gold standard labels. To address these challenges, we developed PheCAP, a high-throughput semi-supervised phenotyping pipeline. PheCAP begins with data from the EMR, including structured data and information extracted from the narrative notes using natural language processing (NLP). The standardized steps integrate automated procedures, which reduce the level of manual input, and machine learning approaches for algorithm training. PheCAP itself can be executed in 1-2 d if all data are available; however, the timing is largely dependent on the chart review stage, which typically requires at least 2 weeks. The final products of PheCAP include a phenotype algorithm, the probability of the phenotype for all patients, and a phenotype classification (yes or no).


Subject(s)
Data Analysis , Electronic Health Records/statistics & numerical data , High-Throughput Screening Assays/methods , Algorithms , Data Interpretation, Statistical , Humans , Machine Learning , Natural Language Processing , Phenotype
4.
Nat Genet ; 51(7): 1073-1075, 2019 07.
Article in English | MEDLINE | ID: mdl-31253975
6.
PLoS One ; 13(12): e0208240, 2018.
Article in English | MEDLINE | ID: mdl-30566451

ABSTRACT

OBJECTIVES: There is limited information on the epidemiology and treatment patterns of rheumatoid arthritis (RA) across the Arab region. We aim in this study to describe the demographic characteristics, clinical profile, and treatment patterns of patients of Arab ancestry with RA. METHODS: This is a cross sectional study of 895 patients with established rheumatoid arthritis enrolled from five sites (Jordan, Lebanon, Qatar, Kingdom of Saudi Arabia (KSA), and United Arab Emirates). Demographic characteristics, clinical profile, and treatment patterns are compared between the five countries. RESULTS: The majority of our patients are women, have an average disease duration of 10 years, are married and non-smokers, with completed secondary education. We report a high (>80%) ever-use of methotrexate (MTX) and steroids among our RA population, while the ever-use of disease modifying anti-rheumatic drugs (DMARDs) and TNF-inhibitors average around 67% and 33%, respectively. There are variations in RA treatment use between the five country sites. Highest utilization of steroids is identified in Jordan and KSA (p-value < 0.001), while the highest ever-use of TNF-inhibitors is reported in KSA (p-value < 0.001). CONCLUSION: Disparities in usage of RA treatments among Arab patients are noted across the five countries. National gross domestic product (GDP), as well as some other unique features in each country likely affect these. Developing treatment guidelines specific to this region could contribute in delivering standardized therapies to RA patients.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/epidemiology , Adult , Cross-Sectional Studies , Etanercept/therapeutic use , Female , Humans , Jordan/epidemiology , Lebanon/epidemiology , Male , Methotrexate/therapeutic use , Middle Aged , Odds Ratio , Qatar/epidemiology , Saudi Arabia/epidemiology , United Arab Emirates/epidemiology
7.
Nat Commun ; 9(1): 4285, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30327483

ABSTRACT

Phenome-wide association studies (PheWAS) have been proposed as a possible aid in drug development through elucidating mechanisms of action, identifying alternative indications, or predicting adverse drug events (ADEs). Here, we select 25 single nucleotide polymorphisms (SNPs) linked through genome-wide association studies (GWAS) to 19 candidate drug targets for common disease indications. We interrogate these SNPs by PheWAS in four large cohorts with extensive health information (23andMe, UK Biobank, FINRISK, CHOP) for association with 1683 binary endpoints in up to 697,815 individuals and conduct meta-analyses for 145 mapped disease endpoints. Our analyses replicate 75% of known GWAS associations (P < 0.05) and identify nine study-wide significant novel associations (of 71 with FDR < 0.1). We describe associations that may predict ADEs, e.g., acne, high cholesterol, gout, and gallstones with rs738409 (p.I148M) in PNPLA3 and asthma with rs1990760 (p.T946A) in IFIH1. Our results demonstrate PheWAS as a powerful addition to the toolkit for drug discovery.


Subject(s)
Drug Discovery/methods , Genome-Wide Association Study/methods , Polymorphism, Single Nucleotide , Asthma/genetics , Cohort Studies , Databases, Factual , Genetic Association Studies , Genetic Pleiotropy , Genetic Predisposition to Disease , Humans , Interferon-Induced Helicase, IFIH1/genetics , Lipase/genetics , Membrane Proteins/genetics , Molecular Targeted Therapy/methods , Phenotype , Reproducibility of Results , Thromboembolism/genetics , United Kingdom
8.
Nature ; 558(7708): 73-79, 2018 06.
Article in English | MEDLINE | ID: mdl-29875488

ABSTRACT

Although plasma proteins have important roles in biological processes and are the direct targets of many drugs, the genetic factors that control inter-individual variation in plasma protein levels are not well understood. Here we characterize the genetic architecture of the human plasma proteome in healthy blood donors from the INTERVAL study. We identify 1,927 genetic associations with 1,478 proteins, a fourfold increase on existing knowledge, including trans associations for 1,104 proteins. To understand the consequences of perturbations in plasma protein levels, we apply an integrated approach that links genetic variation with biological pathway, disease, and drug databases. We show that protein quantitative trait loci overlap with gene expression quantitative trait loci, as well as with disease-associated loci, and find evidence that protein biomarkers have causal roles in disease using Mendelian randomization analysis. By linking genetic factors to diseases via specific proteins, our analyses highlight potential therapeutic targets, opportunities for matching existing drugs with new disease indications, and potential safety concerns for drugs under development.


Subject(s)
Blood Proteins/genetics , Genomics , Proteome/genetics , Female , Hepatocyte Growth Factor/genetics , Humans , Inflammatory Bowel Diseases/genetics , Male , Mutation, Missense/genetics , Myeloblastin/genetics , Positive Regulatory Domain I-Binding Factor 1/genetics , Proto-Oncogene Proteins/genetics , Quantitative Trait Loci/genetics , Vasculitis/genetics , alpha 1-Antitrypsin/genetics
9.
Nature ; 544(7649): 171-172, 2017 04 12.
Article in English | MEDLINE | ID: mdl-28406199
10.
Arthritis Rheumatol ; 69(5): 976-985, 2017 05.
Article in English | MEDLINE | ID: mdl-28118524

ABSTRACT

OBJECTIVE: Genetic factors underlying susceptibility to rheumatoid arthritis (RA) in Arab populations are largely unknown. This genome-wide association study (GWAS) was undertaken to explore the generalizability of previously reported RA loci to Arab subjects and to discover new Arab-specific genetic loci. METHODS: The Genetics of Rheumatoid Arthritis in Some Arab States Study was designed to examine the genetics and clinical features of RA patients from Jordan, the Kingdom of Saudi Arabia, Lebanon, Qatar, and the United Arab Emirates. In total, >7 million single-nucleotide polymorphisms (SNPs) were tested for association with RA overall and with seropositive or seronegative RA in 511 RA cases and 352 healthy controls. In addition, replication of 15 signals was attempted in 283 RA cases and 221 healthy controls. A genetic risk score of 68 known RA SNPs was also examined in this study population. RESULTS: Three loci (HLA region, intergenic 5q13, and 17p13 at SMTNL2/GGT6) reached genome-wide significance in the analyses of association with RA and with seropositive RA, and for all 3 loci, evidence of independent replication was demonstrated. Consistent with the findings in European and East Asian populations, the association of RA with HLA-DRB1 amino acid position 11 conferred the strongest effect (P = 4.8 × 10-16 ), and a weighted genetic risk score of previously associated RA loci was found to be associated with RA (P = 3.41 × 10-5 ) and with seropositive RA (P = 1.48 × 10-6 ) in this population. In addition, 2 novel associations specific to Arab populations were found at the 5q13 and 17p13 loci. CONCLUSION: This first RA GWAS in Arab populations confirms that established HLA-region and known RA risk alleles contribute strongly to the risk and severity of disease in some Arab groups, suggesting that the genetic architecture of RA is similar across ethnic groups. Moreover, this study identified 2 novel RA risk loci in Arabs, offering further population-specific insights into the pathophysiology of RA.


Subject(s)
Arthritis, Rheumatoid/genetics , HLA-DRB1 Chains/genetics , Phosphoproteins/genetics , gamma-Glutamyltransferase/genetics , Adult , Arabs/genetics , Arthritis, Rheumatoid/immunology , Case-Control Studies , Chromosomes, Human, Pair 3/genetics , Chromosomes, Human, Pair 5/genetics , DNA, Intergenic/genetics , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , HLA Antigens/genetics , Humans , Jordan , Lebanon , Male , Middle Aged , Peptides, Cyclic/immunology , Polymorphism, Single Nucleotide , Qatar , Rheumatoid Factor/immunology , Saudi Arabia , United Arab Emirates
11.
Arthritis Rheumatol ; 69(4): 735-741, 2017 04.
Article in English | MEDLINE | ID: mdl-27788309

ABSTRACT

OBJECTIVE: In many rheumatoid arthritis (RA) patients, disease is controlled with anti-tumor necrosis factor (anti-TNF) biologic therapies. However, in a significant number of patients, the disease fails to respond to anti-TNF therapy. We undertook the present study to examine the hypothesis that rare and low-frequency genetic variants might influence response to anti-TNF treatment. METHODS: We sequenced the coding region of 750 genes in 1,094 RA patients of European ancestry who were treated with anti-TNF. After quality control, 690 genes were included in the analysis. We applied single-variant association and gene-based association tests to identify variants associated with anti-TNF treatment response. In addition, given the key mechanistic role of TNF, we performed gene set analyses of 27 TNF pathway genes. RESULTS: We identified 14,420 functional variants, of which 6,934 were predicted as nonsynonymous 2,136 of which were further predicted to be "damaging." Despite the fact that the study was well powered, no single variant or gene showed study-wide significant association with change in the outcome measures disease activity or European League Against Rheumatism response. Intriguingly, we observed 3 genes, of 27 with nominal signals of association (P < 0.05), that were involved in the TNF signaling pathway. However, when we performed a rigorous gene set enrichment analysis based on association P value ranking, we observed no evidence of enrichment of association at genes involved in the TNF pathway (Penrichment = 0.15, based on phenotype permutations). CONCLUSION: Our findings suggest that rare and low-frequency protein-coding variants in TNF signaling pathway genes or other genes do not contribute substantially to anti-TNF treatment response in patients with RA.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Female , Genetic Variation , Humans , Male , Middle Aged , Open Reading Frames , Treatment Outcome
12.
PLoS Genet ; 12(9): e1006292, 2016 09.
Article in English | MEDLINE | ID: mdl-27626929

ABSTRACT

Understanding the implications of genome-wide association studies (GWAS) for disease biology requires both identification of causal variants and definition of how these variants alter gene function. The non-coding triallelic dinucleotide polymorphism CCR6DNP is associated with risk for rheumatoid arthritis, and is considered likely causal because allelic variation correlates with expression of the chemokine receptor CCR6. Using transcription activator-like effector nuclease (TALEN) gene editing, we confirmed that CCR6DNP regulates CCR6. To identify the associated transcription factor, we applied a novel assay, Flanking Restriction Enhanced Pulldown (FREP), to identify specific association of poly (ADP-ribose) polymerase 1 (PARP-1) with CCR6DNP consistent with the established allelic risk hierarchy. Correspondingly, manipulation of PARP-1 expression or activity impaired CCR6 expression in several lineages. These findings show that CCR6DNP is a causal variant through which PARP-1 regulates CCR6, and introduce a highly efficient approach to interrogate non-coding genetic polymorphisms associated with human disease.


Subject(s)
Arthritis, Rheumatoid/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Polymorphism, Genetic , Receptors, CCR6/genetics , Cell Line , HCT116 Cells , Humans , Poly (ADP-Ribose) Polymerase-1/metabolism , Receptors, CCR6/metabolism
13.
Sci Transl Med ; 8(349): 349ps15, 2016 07 27.
Article in English | MEDLINE | ID: mdl-27464747

ABSTRACT

Our modern health care system demands therapeutic interventions that improve the lives of patients. Unfortunately, decreased productivity in therapeutics research and development (R&D) has driven drug costs up while delivering insufficient value to patients. Here, I discuss a model of translational medicine that connects four components of the early R&D pipeline-causal human biology, therapeutic modality, biomarkers of target modulation, and proof-of-concept clinical trials. Whereas the individual components of this model are not new, technological advances and a disciplined approach to integrating all four areas offer hope for improving R&D productivity.


Subject(s)
Drug Discovery , Models, Theoretical , Drug Design , Humans , Translational Research, Biomedical
14.
Eur J Hum Genet ; 24(2): 263-70, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25966632

ABSTRACT

Segmental duplications (SDs) comprise about 5% of the human genome and are enriched for immune genes. SD loci often show copy numbers variations (CNV), which are difficult to tag with genotyping methods. CNV in the Fcγ receptor region (FCGR) has been suggested to be associated with rheumatic diseases. The objective of this study was to delineate association of FCGR-CNV with rheumatoid arthritis (RA), coeliac disease and Inflammatory bowel disease incidence. We developed a method to accurately quantify CNV in SD loci based on the intensity values from the Immunochip platform and applied it to the FCGR locus. We determined the method's validity using three independent assays: segregation analysis in families, arrayCGH, and whole genome sequencing. Our data showed the presence of two separate CNVs in the FCGR locus. The first region encodes FCGR2A, FCGR3A and part of FCGR2C gene, the second encodes another part of FCGR2C, FCGR3B and FCGR2B. Analysis of CNV status in 4578 individuals with RA and 5457 controls indicated association of duplications in the FCGR3B gene in antibody-negative RA (P=0.002, OR=1.43). Deletion in FCGR3B was associated with increased risk of antibody-positive RA, consistently with previous reports (P=0.023, OR=1.23). A clear genotype-phenotype relationship was observed: CNV polymorphisms of the FCGR3A gene correlated to CD16A expression (encoded by FCGR3A) on CD8 T-cells. In conclusion, our method allows determining the CNV status of the FCGR locus, we identified association of CNV in FCGR3B to RA and showed a functional relationship between CNV in the FCGR3A gene and CD16A expression.


Subject(s)
Arthritis, Rheumatoid/genetics , Inflammatory Bowel Diseases/genetics , Receptors, IgG/biosynthesis , Receptors, IgG/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , DNA Copy Number Variations/genetics , Female , GPI-Linked Proteins/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Inflammatory Bowel Diseases/pathology , Male , Phenotype , Polymorphism, Single Nucleotide
15.
PLoS One ; 10(10): e0140815, 2015.
Article in English | MEDLINE | ID: mdl-26517366

ABSTRACT

Every person carries a vast repertoire of CD4+ T-helper cells and CD8+ cytotoxic T cells for a healthy immune system. Somatic VDJ recombination at genomic loci that encode the T-cell receptor (TCR) is a key step during T-cell development, but how a single T cell commits to become either CD4+ or CD8+ is poorly understood. To evaluate the influence of TCR sequence variation on CD4+/CD8+ lineage commitment, we sequenced rearranged TCRs for both α and ß chains in naïve T cells isolated from healthy donors and investigated gene segment usage and recombination patterns in CD4+ and CD8+ T-cell subsets. Our data demonstrate that most V and J gene segments are strongly biased in the naïve CD4+ and CD8+ subsets with some segments increasing the odds of being CD4+ (or CD8+) up to five-fold. These V and J gene associations are highly reproducible across individuals and independent of classical HLA genotype, explaining ~11% of the observed variance in the CD4+ vs. CD8+ propensity. In addition, we identified a strong independent association of the electrostatic charge of the complementarity determining region 3 (CDR3) in both α and ß chains, where a positively charged CDR3 is associated with CD4+ lineage and a negatively charged CDR3 with CD8+ lineage. Our findings suggest that somatic variation in different parts of the TCR influences T-cell lineage commitment in a predominantly additive fashion. This notion can help delineate how certain structural features of the TCR-peptide-HLA complex influence thymic selection.


Subject(s)
Genes, T-Cell Receptor/genetics , HLA Antigens/genetics , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Genes, T-Cell Receptor alpha/genetics , Genes, T-Cell Receptor beta/genetics , Genetic Variation , Humans , Receptors, Antigen, T-Cell/genetics
16.
PLoS One ; 10(8): e0136651, 2015.
Article in English | MEDLINE | ID: mdl-26301417

ABSTRACT

BACKGROUND: Typically, algorithms to classify phenotypes using electronic medical record (EMR) data were developed to perform well in a specific patient population. There is increasing interest in analyses which can allow study of a specific outcome across different diseases. Such a study in the EMR would require an algorithm that can be applied across different patient populations. Our objectives were: (1) to develop an algorithm that would enable the study of coronary artery disease (CAD) across diverse patient populations; (2) to study the impact of adding narrative data extracted using natural language processing (NLP) in the algorithm. Additionally, we demonstrate how to implement CAD algorithm to compare risk across 3 chronic diseases in a preliminary study. METHODS AND RESULTS: We studied 3 established EMR based patient cohorts: diabetes mellitus (DM, n = 65,099), inflammatory bowel disease (IBD, n = 10,974), and rheumatoid arthritis (RA, n = 4,453) from two large academic centers. We developed a CAD algorithm using NLP in addition to structured data (e.g. ICD9 codes) in the RA cohort and validated it in the DM and IBD cohorts. The CAD algorithm using NLP in addition to structured data achieved specificity >95% with a positive predictive value (PPV) 90% in the training (RA) and validation sets (IBD and DM). The addition of NLP data improved the sensitivity for all cohorts, classifying an additional 17% of CAD subjects in IBD and 10% in DM while maintaining PPV of 90%. The algorithm classified 16,488 DM (26.1%), 457 IBD (4.2%), and 245 RA (5.0%) with CAD. In a cross-sectional analysis, CAD risk was 63% lower in RA and 68% lower in IBD compared to DM (p<0.0001) after adjusting for traditional cardiovascular risk factors. CONCLUSIONS: We developed and validated a CAD algorithm that performed well across diverse patient populations. The addition of NLP into the CAD algorithm improved the sensitivity of the algorithm, particularly in cohorts where the prevalence of CAD was low. Preliminary data suggest that CAD risk was significantly lower in RA and IBD compared to DM.


Subject(s)
Coronary Artery Disease/epidemiology , Diabetes Mellitus/epidemiology , Electronic Health Records , Adult , Aged , Algorithms , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/epidemiology , Arthritis, Rheumatoid/physiopathology , Coronary Artery Disease/complications , Coronary Artery Disease/physiopathology , Diabetes Mellitus/physiopathology , Female , Humans , Hyperlipidemias/complications , Hyperlipidemias/epidemiology , Hyperlipidemias/physiopathology , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/epidemiology , Inflammatory Bowel Diseases/physiopathology , Male , Middle Aged , Natural Language Processing , Phenotype , Risk Factors
17.
Biomed Res Int ; 2015: 490295, 2015.
Article in English | MEDLINE | ID: mdl-25834819

ABSTRACT

BACKGROUND: The aim of our work was to replicate, in a Southern European population, the association reported in Northern populations between PTPRC locus and response to anti-tumor necrosis factor (anti-TNF) treatment in rheumatoid arthritis (RA). We also looked at associations between five RA risk alleles and treatment response. METHODS: We evaluated associations between anti-TNF treatment responses assessed by DAS28 change and by EULAR response at six months in 383 Portuguese patients. Univariate and multivariate linear and logistic regression analyses were performed. In a second step to confirm our findings, we pooled our population with 265 Spanish patients. RESULTS: No association was found between PTPRC rs10919563 allele and anti-TNF treatment response, neither in Portuguese modeling for several clinical variables nor in the overall population combining Portuguese and Spanish patients. The minor allele for RA susceptibility, rs3761847 SNP in TRAF1/C5 region, was associated with a poor response in linear and logistic univariate and multivariate regression analyses. No association was observed with the other allellic variants. Results were confirmed in the pooled analysis. CONCLUSION: This study did not replicate the association between PTPRC and the response to anti-TNF treatment in our Southern European population. We found that TRAF1/C5 risk RA variants potentially influence anti-TNF treatment response.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Genetic Association Studies , Leukocyte Common Antigens/genetics , TNF Receptor-Associated Factor 1/genetics , Adalimumab/administration & dosage , Adult , Aged , Alleles , Antibodies, Monoclonal/administration & dosage , Arthritis, Rheumatoid/pathology , Etanercept/administration & dosage , HLA-DRB1 Chains/genetics , Humans , Infliximab/administration & dosage , Middle Aged , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics
18.
Ann Rheum Dis ; 74(3): e13, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24532676

ABSTRACT

OBJECTIVE: A highly polygenic aetiology and high degree of allele-sharing between ancestries have been well elucidated in genetic studies of rheumatoid arthritis. Recently, the high-density genotyping array Immunochip for immune disease loci identified 14 new rheumatoid arthritis risk loci among individuals of European ancestry. Here, we aimed to identify new rheumatoid arthritis risk loci using Korean-specific Immunochip data. METHODS: We analysed Korean rheumatoid arthritis case-control samples using the Immunochip and genome-wide association studies (GWAS) array to search for new risk alleles of rheumatoid arthritis with anticitrullinated peptide antibodies. To increase power, we performed a meta-analysis of Korean data with previously published European Immunochip and GWAS data for a total sample size of 9299 Korean and 45,790 European case-control samples. RESULTS: We identified eight new rheumatoid arthritis susceptibility loci (TNFSF4, LBH, EOMES, ETS1-FLI1, COG6, RAD51B, UBASH3A and SYNGR1) that passed a genome-wide significance threshold (p<5×10(-8)), with evidence for three independent risk alleles at 1q25/TNFSF4. The risk alleles from the seven new loci except for the TNFSF4 locus (monomorphic in Koreans), together with risk alleles from previously established RA risk loci, exhibited a high correlation of effect sizes between ancestries. Further, we refined the number of single nucleotide polymorphisms (SNPs) that represent potentially causal variants through a trans-ethnic comparison of densely genotyped SNPs. CONCLUSIONS: This study demonstrates the advantage of dense-mapping and trans-ancestral analysis for identification of potentially causal SNPs. In addition, our findings support the importance of T cells in the pathogenesis and the fact of frequent overlap of risk loci among diverse autoimmune diseases.


Subject(s)
Arthritis, Rheumatoid/genetics , Asian People/genetics , White People/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Case-Control Studies , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Male , Middle Aged , Republic of Korea/ethnology , Young Adult
19.
Cell Rep ; 9(4): 1417-29, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25453756

ABSTRACT

A large portion of common variant loci associated with genetic risk for schizophrenia reside within noncoding sequence of unknown function. Here, we demonstrate promoter and enhancer enrichment in schizophrenia variants associated with expression quantitative trait loci (eQTL). The enrichment is greater when functional annotations derived from the human brain are used relative to peripheral tissues. Regulatory trait concordance analysis ranked genes within schizophrenia genome-wide significant loci for a potential functional role, based on colocalization of a risk SNP, eQTL, and regulatory element sequence. We identified potential physical interactions of noncontiguous proximal and distal regulatory elements. This was verified in prefrontal cortex and -induced pluripotent stem cell-derived neurons for the L-type calcium channel (CACNA1C) risk locus. Our findings point to a functional link between schizophrenia-associated noncoding SNPs and 3D genome architecture associated with chromosomal loopings and transcriptional regulation in the brain.


Subject(s)
DNA, Intergenic/genetics , Polymorphism, Single Nucleotide/genetics , Schizophrenia/genetics , Arthritis, Rheumatoid/genetics , Calcium Channels, L-Type/genetics , Databases, Genetic , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Genetic Loci , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Molecular Sequence Annotation , Organ Specificity/genetics , Promoter Regions, Genetic , Protein Binding/genetics , Risk Factors
20.
Arthritis Rheumatol ; 66(12): 3274-82, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25524867

ABSTRACT

OBJECTIVE: To evaluate HLA-DRB1 genetic risk of rheumatoid arthritis (RA) in African Americans by 3 validated allele classification systems and by amino acid position and residue, and to compare genetic risk between African American and European ancestries. METHODS: Four-digit HLA-DRB1 genotyping was performed on 561 autoantibody-positive African American cases and 776 African American controls. Association analysis was performed on Tezenas du Montcel (TdM), de Vries (DV), and Mattey classification system alleles and separately by amino acid position and individual residues. RESULTS: TdM S2 and S3P alleles were associated with RA (odds ratio [95% confidence interval] 2.8 [2.0-3.9] and 2.1 [1.7-2.7], respectively). The DV (P = 3.2 × 10(-12)) and Mattey (P = 6.5 × 10(-13)) system alleles were both protective in African Americans. Amino acid position 11 (permutation P < 0.00001) accounted for nearly all variability explained by HLA-DRB1, although conditional analysis demonstrated that position 57 was also significant (0.01 ≤ permutation P ≤ 0.05). The valine and aspartic acid residues at position 11 conferred the highest risk of RA in African Americans. CONCLUSION: With some exceptions, the genetic risk conferred by HLA-DRB1 in African Americans is similar to that in individuals of European ancestry at multiple levels: classification system (e.g., TdM), amino acid position (e.g., 11), and residue (Val11). Unlike that reported for individuals of European ancestry, amino acid position 57 was associated with RA in African Americans, but positions 71 and 74 were not. Asp11 (odds ratio 1 in European ancestry) corresponds to the 4-digit classical allele *09:01, which is also a risk allele for RA in Koreans.


Subject(s)
Arthritis, Rheumatoid/genetics , Black or African American/genetics , HLA-DRB1 Chains/genetics , White People/genetics , Adult , Alleles , Amino Acid Motifs/genetics , Amino Acids , Case-Control Studies , Epitopes/genetics , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Odds Ratio
SELECTION OF CITATIONS
SEARCH DETAIL
...