Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
JCI Insight ; 5(17)2020 09 03.
Article in English | MEDLINE | ID: mdl-32879140

ABSTRACT

More than 90% of autoimmune-associated variants are located in noncoding regions, leading to challenges in deciphering the underlying causal roles of functional variants and genes and biological mechanisms. Therefore, to reduce the gap between traditional genetic findings and mechanistic understanding of disease etiologies and clinical drug development, it is important to translate systematically the regulatory mechanisms underlying noncoding variants. Here, we prioritized functional noncoding SNPs with regulatory gene targets associated with 19 autoimmune diseases by incorporating hundreds of immune cell-specific multiomics data. The prioritized SNPs are associated with transcription factor (TF) binding, histone modification, or chromatin accessibility, indicating their allele-specific regulatory roles. Their target genes are significantly enriched in immunologically related pathways and other known immunologically related functions. We found that 90.1% of target genes are regulated by distal SNPs involving several TFs (e.g., the DNA-binding protein CCCTC-binding factor [CTCF]), suggesting the importance of long-range chromatin interaction in autoimmune diseases. Moreover, we predicted potential drug targets for autoimmune diseases, including 2 genes (NFKB1 and SH2B3) with known drug indications on other diseases, highlighting their potential drug repurposing opportunities. Taken together, these findings may provide useful information for future experimental follow-up and drug applications on autoimmune diseases.


Subject(s)
Autoimmune Diseases/genetics , Genome-Wide Association Study/methods , Genomics/methods , Polymorphism, Single Nucleotide , Regulatory Sequences, Nucleic Acid , Adaptor Proteins, Signal Transducing/genetics , CCCTC-Binding Factor/genetics , Humans , NF-kappa B p50 Subunit/genetics , Pharmacogenomic Variants , Software
3.
Bioinformatics ; 36(18): 4739-4748, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32539144

ABSTRACT

MOTIVATION: CircRNAs are an abundant class of non-coding RNAs with widespread, cell-/tissue-specific patterns. Previous work suggested that epigenetic features might be related to circRNA expression. However, the contribution of epigenetic changes to circRNA expression has not been investigated systematically. Here, we built a machine learning framework named CIRCScan, to predict circRNA expression in various cell lines based on the sequence and epigenetic features. RESULTS: The predicted accuracy of the expression status models was high with area under the curve of receiver operating characteristic (ROC) values of 0.89-0.92 and the false-positive rates of 0.17-0.25. Predicted expressed circRNAs were further validated by RNA-seq data. The performance of expression-level prediction models was also good with normalized root-mean-square errors of 0.28-0.30 and Pearson's correlation coefficient r over 0.4 in all cell lines, along with Spearman's correlation coefficient ρ of 0.33-0.46. Noteworthy, H3K79me2 was highly ranked in modeling both circRNA expression status and levels across different cells. Further analysis in additional nine cell lines demonstrated a significant enrichment of H3K79me2 in circRNA flanking intron regions, supporting the potential involvement of H3K79me2 in circRNA expression regulation. AVAILABILITY AND IMPLEMENTATION: The CIRCScan assembler is freely available online for academic use at https://github.com/johnlcd/CIRCScan. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Epigenomics , RNA, Circular , Epigenesis, Genetic , Machine Learning , RNA/genetics , ROC Curve
4.
J Invest Dermatol ; 140(2): 348-360.e11, 2020 02.
Article in English | MEDLINE | ID: mdl-31421124

ABSTRACT

Both systemic lupus erythematosus (SLE) and systemic sclerosis (SSc) are autoimmune diseases sharing similar genetic backgrounds. Genome-wide association studies have constantly disclosed numerous genetic variants conferring to both disease risks at 7q32.1, but the functional mechanisms underlying them are still largely unknown. Through a series of bioinformatics and functional analyses, we prioritized a potential independent functional single-nucleotide polymorphism (rs13239597) within TNPO3 promoter region, residing in a putative enhancer element and validated that IRF5 is the distal target gene (∼118 kb) of rs13239597, which is a key regulator involved in pathogenic autoantibody dysregulation, increasing risk of both SLE and SSc. We experimentally validated the long-range chromatin interactions between rs13239597 and IRF5 using chromosome conformation capture assay. We further demonstrated that rs13239597-A acted as an allele-specific enhancer regulating IRF5 expression, independently of TNPO3 by using dual-luciferase reporter assays and CRISPR-Cas9. Particularly, the transcription factor EVI1 could preferentially bind to rs13239597-A allele and increase the enhancer activity to regulate IRF5 expression. Taken together, our results uncovered a mechanistic insight of a noncoding functional variant acting as an allele-specific distal enhancer to directly modulate IRF5 expression, which might obligate in understanding of complex genetic architectures of SLE and SSc pathogenesis.


Subject(s)
Chromatin/metabolism , Interferon Regulatory Factors/genetics , Lupus Erythematosus, Systemic/genetics , MDS1 and EVI1 Complex Locus Protein/metabolism , Scleroderma, Systemic/genetics , Alleles , Chromatin Immunoprecipitation , Chromosomes, Human, Pair 7/genetics , Computational Biology , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Linkage Disequilibrium , Lupus Erythematosus, Systemic/immunology , Polymorphism, Single Nucleotide/immunology , Promoter Regions, Genetic/genetics , Quantitative Trait Loci/immunology , Scleroderma, Systemic/immunology , beta Karyopherins/genetics
5.
J Cancer ; 9(21): 3858-3866, 2018.
Article in English | MEDLINE | ID: mdl-30410588

ABSTRACT

Although genome-wide association studies (GWASs) have identified some risk single-nucleotide polymorphisms in East Asian never-smoking females, the unexplained missing heritability is still required to be investigated. Runs of homozygosity (ROHs) are thought to be a type of genetic variation acting on human complex traits and diseases. We detected ROHs in 8,881 East Asian never-smoking women. The summed ROHs were used to fit a logistic regression model which noteworthily revealed a significant association between ROHs and the decreased risk of lung cancer (P < 0.05). We identified 4 common ROHs regions located at 2p22.1, which were significantly associated with decreased risk of lung cancer (P = 2.00 × 10-4 - 1.35 × 10-4). Functional annotation was conducted to investigate the regulatory function of ROHs. The common ROHs were overlapped with potential regulatory elements, such as active epigenome elements and chromatin states in lung-derived cell lines. SOS1 and ARHGEF33 were significantly up-regulated as the putative target genes of the identified ROHs in lung cancer samples according to the analysis of differently expressed genes. Our results suggest that ROHs could act as recessive contributing factors and regulatory elements to influence the risk of lung cancer in never-smoking East Asian females.

6.
Am J Hum Genet ; 102(5): 776-793, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29706346

ABSTRACT

Genome-wide association studies (GWASs) have reproducibly associated variants within intergenic regions of 1p36.12 locus with osteoporosis, but the functional roles underlying these noncoding variants are unknown. Through an integrative functional genomic and epigenomic analyses, we prioritized rs6426749 as a potential causal SNP for osteoporosis at 1p36.12. Dual-luciferase assay and CRISPR/Cas9 experiments demonstrate that rs6426749 acts as a distal allele-specific enhancer regulating expression of a lncRNA (LINC00339) (∼360 kb) via long-range chromatin loop formation and that this loop is mediated by CTCF occupied near rs6426749 and LINC00339 promoter region. Specifically, rs6426749-G allele can bind transcription factor TFAP2A, which efficiently elevates the enhancer activity and increases LINC00339 expression. Downregulation of LINC00339 significantly increases the expression of CDC42 in osteoblast cells, which is a pivotal regulator involved in bone metabolism. Our study provides mechanistic insight into how a noncoding SNP affects osteoporosis by long-range interaction, a finding that could indicate promising therapeutic targets for osteoporosis.


Subject(s)
Alleles , Chromosomes, Human, Pair 1/genetics , Enhancer Elements, Genetic , Gene Expression Regulation , Nucleic Acid Conformation , Osteoporosis/genetics , Polymorphism, Single Nucleotide/genetics , RNA, Long Noncoding/genetics , Asian People/genetics , Base Sequence , Bone Density/genetics , Bone and Bones/metabolism , CRISPR-Cas Systems/genetics , Cell Line , Chromatin/metabolism , Genome-Wide Association Study , Humans , Models, Genetic , Promoter Regions, Genetic , Protein Binding , Quantitative Trait Loci/genetics , RNA, Long Noncoding/chemistry , Reproducibility of Results , Risk Factors , Transcription Factors/metabolism
7.
J Bone Miner Res ; 33(7): 1335-1346, 2018 07.
Article in English | MEDLINE | ID: mdl-29528523

ABSTRACT

RANKL is a key regulator involved in bone metabolism, and a drug target for osteoporosis. The clinical diagnosis and assessment of osteoporosis are mainly based on bone mineral density (BMD). Previous powerful genomewide association studies (GWASs) have identified multiple intergenic single-nucleotide polymorphisms (SNPs) located over 100 kb upstream of RANKL and 65 kb downstream of AKAP11 at 13q14.11 for osteoporosis. Whether these SNPs exert their roles on osteoporosis through RANKL is unknown. In this study, we conducted integrative analyses combining expression quantitative trait locus (eQTL), genomic chromatin interaction (high-throughput chromosome conformation capture [Hi-C]), epigenetic annotation, and a series of functional assays. The eQTL analysis identified six potential functional SNPs (rs9533090, rs9594738, r8001611, rs9533094, rs9533095, and rs9594759) exclusively correlated with RANKL gene expression (p < 0.001) at 13q14.11. Co-localization analyses suggested that eQTL signal for RANKL and BMD-GWAS signal shared the same causal variants. Hi-C analysis and functional annotation further validated that the first five osteoporosis SNPs are located in a super-enhancer region to regulate the expression of RANKL via long-range chromosomal interaction. Particularly, dual-luciferase assay showed that the region harboring rs9533090 in the super-enhancer has the strongest enhancer activity, and rs9533090 is an allele-specific regulatory SNP. Furthermore, deletion of the region harboring rs9533090 using CRISPR/Cas9 genome editing significantly reduced RANKL expression in both mRNA level and protein level. Finally, we found that the rs9533090-C robustly recruits transcription factor NFIC, which efficiently elevates the enhancer activity and increases the RANKL expression. In summary, we provided a feasible method to identify regulatory noncoding SNPs to distally regulate their target gene underlying the pathogenesis of osteoporosis by using bioinformatics data analyses and experimental validation. Our findings would be a potential and promising therapeutic target for precision medicine in osteoporosis. © 2018 American Society for Bone and Mineral Research.


Subject(s)
Chromosomes, Human, Pair 13/genetics , Enhancer Elements, Genetic , Genetic Predisposition to Disease , Osteoporosis/genetics , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , RANK Ligand/genetics , Alleles , Bone Density/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Chromatin/genetics , Humans , Models, Genetic , Molecular Sequence Annotation , NFI Transcription Factors/metabolism , Physical Chromosome Mapping , Protein Binding , Reproducibility of Results , Risk Factors
8.
Endocrine ; 59(2): 296-303, 2018 02.
Article in English | MEDLINE | ID: mdl-29299795

ABSTRACT

PURPOSE: Animal-based studies have reported a decrease in bone mass resulting from high level of fibroblast growth factor 21 (FGF21). However, the correlation between plasma FGF21 levels and bone mineral density (BMD) is paradoxical in previous human-based studies, and the associations between FGF21 gene polymorphisms and BMD haven't been reported yet. Therefore, here, we evaluated plasma FGF21 levels with sufficient study samples, and performed genetic association test to reveal the physiological and genetic role of FGF21 on BMD in adults. METHODS: Plasma and genetic samples containing 168 and 569 Han Chinese subjects, respectively, were employed in this study. Fasting plasma FGF21 levels were determined using enzyme-linked immunosorbent assay (ELISA). Regional BMD values were measured by dual energy X-ray absorptiometry (DXA). Five variants of FGF21 gene were successfully genotyped. RESULTS: Physiological association suggested that plasma FGF21 levels were inversely correlated with BMD in femoral neck (Neck-BMD: P = 0.039) and Ward's triangle (Ward's-BMD: P = 0.002) of hip region. A FGF21 gene variant, rs490942, was significantly associated with the increase of Ward's-BMD in total (P = 0.027) and female (P = 0.016) cohorts, as well as Neck-BMD in female cohort (P = 7.45 × 10-3). Meanwhile, eQTL results indicated that this SNP was related to the decreased level of FGF21 gene expression. CONCLUSIONS: Taking together from both physiological and genetic levels, we suggest that FGF21 is inversely associated with regional BMD. And we haven't observed sex-specific effect in this study.


Subject(s)
Bone Density/physiology , Femur Neck/diagnostic imaging , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Absorptiometry, Photon , Aged , Female , Genetic Association Studies , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL
...