Single‑nucleotide polymorphism rs6592645 confers asthma risk through regulating LRRC32 expression

  • Authors:
    • Yi-Kun Li
    • Hong-Yan Wang
    • Ying Chen
    • Xiao-Qian Shi
    • Xin-Xin Zhang
    • Ke Li
    • Wei-Ping Fu
    • Chang Sun
  • View Affiliations

  • Published online on: August 3, 2023     https://doi.org/10.3892/etm.2023.12150
  • Article Number: 451
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Asthma is a complex disease, often with evident genetic predisposition; for example, the single‑nucleotide polymorphism (SNP) rs7130588 was significantly associated with asthma by genome‑wide association study (GWAS). Analysis of 1000 Genomes Project data suggests that there is another SNP, rs6592645, in complete linkage disequilibrium with rs7130588 and should present the same signal in GWAS. However, the causal SNP and the mechanism for the association between rs7130588 and asthma remain to be elucidated. In the presents study, results from dual‑luciferase assays indicated that the A/G alleles of rs7130588 failed to present significantly different reporter gene expression. By contrast, A allele of rs6592645 presented a significant increase in relative luciferase activity than G allele, thus suggesting that rs6592645 may be a causal SNP. Using chromosome conformation capture, the enhancer region containing rs6592645 was observed to interact with promoter region of leucine‑rich repeat‑containing 32 (LRRC32). Gene expression quantification suggested that LRRC32 expression is significantly increased in lung tissue of patients with asthma and is dependent on the genotype of this locus, thus verifying that LRRC32 may be involved in asthma onset and that rs6592645 can regulate LRRC32 expression. Through chromatin immunoprecipitation, transcription factor 3 (TCF3) was identified to bind to rs6592645 surrounding region and the interaction between TCF3 and rs6592645 surrounding region was investigated. Results from the present study may improve our understanding of the mechanism by which the genetic variation in this locus might influence asthma susceptibility.

Introduction

Asthma is a common complex disease characterized by chronic respiratory tract inflammation, reversible airflow obstruction and airway hyper-responsiveness (1). Asthma can be induced by multiple environmental factors, such as cold and dry air and airborne substances, including chemical fumes, dust and pollen (1). Additionally, genetics plays an important role in the onset and the heritability for asthma is as high as ~80% (1). To discover the potential genetic contribution to asthma, numerous genome-wide association studies (GWASs) have been performed, and one single-nucleotide polymorphism (SNP), rs7130588, located at 11q13.5 was observed to be associated with asthma, including both adult-onset and childhood-onset, in Caucasian populations (2-6).

It had been suggested that there are ~80 million genetic variations in human genome (7). However, owing to the space limit of the microarray, only a small number (usually ~500,000) can be included and genotyped by GWAS. Therefore, it cannot be discounted that the real causal SNP(s) for asthma might not be rs7130588, but the one(s) in linkage disequilibrium (LD) with it. However, this question has not yet been scrutinized. In addition, since rs7130588 is located at the intergenic region between EMSY transcriptional repressor, BRCA2 interacting (EMSY; also known as C11orf30) and leucine-rich repeat-containing 32 (LRRC32; also known as glycoprotein-A repetitions predominant), with the latter gene encoding for a protein involved in immune response, it had been hypothesized that rs7130588 may be associated with asthma by regulating LRRC32 expression (2). However, owing to the relative long distance (~110.0 kb) between this SNP and the LRRC32 promoter, this interaction has never been demonstrated. As a result, this locus was termed as ‘EMSY (or C11orf30)-LRRC32’ in previous studies (8-14).

The present study attempted to reveal the functional variation(s) in 11q13.5 associated with asthma and its mechanism. Through public genotype data analysis, potential SNPs associated with asthma were identified. Dual-luciferase assay were used to characterize the causal SNP. Furthermore, the present study used a functional genomics approach to investigate the underlying mechanism for asthma.

Materials and methods

1000 Genomes Project (1KGP) data analysis

The upstream and downstream 500 kb genetic sequence flanking rs7130588 for three representative populations, including CEU (Utah Residents with European Ancestry), CHB (Han Chinese in Beijing) and YRI (Yoruba in Ibadan; Nigeria), were downloaded from 1KGP website (http://www.internationalgenome.org). The LD pattern was determined using the Genome Variation Server version 150 (http://gvs.gs.washington.edu/GVS150), with r2≥0.8.

Dual luciferase assay

The rs7130588 and rs6592645 flanking region (~1.5 kb; ~750 bp in each side) was amplified by nested PCR with primers listed in Table SI, digested by KpnI and BglII (New England Biolabs) and inserted into pGL3-promoter vector (Promega Corporation). To avoid artificial mutations, PCR was performed using Phusion High-Fidelity DNA Polymerase (Thermo Fisher Scientific, Inc.) with the following thermocycling conditions: 98˚C for 30 sec; followed by 35 cycles of 98˚C for 10 sec, 56˚C for 30 sec and 72˚C for 30 sec. Following sequencing by BigDye terminator (Thermo Fisher Scientific, Inc.), plasmids containing another allele were generated by mutagenesis using the Q5 Site-directed Mutagenesis Kit (New England Biolabs) and primers listed in Table SI. Prior to transfection, all plasmids were sequenced to verify the sequence and the haplotype orientation.

The human lung bronchial epithelial cell line Beas-2B was purchased from Conservation Genetics CAS (Chinese Academy of Science) Kunming Cell Bank (http://www.kmcellbank.com; cat. no. KCB200922YJ) and maintained in Dulbecco's modified Eagle's medium (high glucose; HyClone; Cytiva) with 10% FBS (Biological Industries USA, Inc.) in 5% CO2 at 37˚C. Beas-2B Cells (~105 cells/well) were plated into 24-well plate and 475 ng plasmid constructs were transfected by Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) at 37˚C for 12 h. After transfection, cells were cultured for additional 36 h at 37˚C and harvested. The luciferase activity was determined by Dual-Luciferase Reporter Assay System (Promega Corporation). A total of six independent transfections were performed for each plasmid. In each transfection, the Renilla luciferase control plasmid pRL-TK (25 ng; Promega Corporation) plasmid was cot-transfected and used to normalize the transfection efficiency.

Chromosome conformation capture (3C)

Spatial contacts between the enhancers and the promoters of nearby genes were examined by 3C and quantitative PCR (qPCR). Briefly, ~108 Beas-2B cells were cross-linked by formaldehyde (1% final concentration) and lysed by lysis buffer (10 mM Tris-HCl, 10 mM NaCl, 0.2% NP40, pH 8.0), and the chromatin was digested by restrictive enzyme (HindIII or EcoRI; see below). After ligation, DNA was purified by standard phenol-chloroform method (https://www.thermofisher.cn/cn/zh/home/references/protocols/nucleic-acid-purification-and-analysis/dna-extraction-protocols/phenol-chloroform-extraction.html). Along with the Beas-2B cells, related bacterial artificial chromosomes (BACs) containing the enhancer and nearby region were ordered from BACPAC Resources Center (https://bacpacresources.org/), cultured in E. coli, isolated, digested, ligated and quantified by qPCR with iQ SYBR green (Bio-Rad Laboratories, Inc.; cat. no. 1725122) and the primers in Tables SII and SIII with the following thermocycling conditions: 96˚C for 10 min; followed by 40 cycles of 96˚C for 10 sec and 60˚C for 30 sec. Owing to the distance between rs6592645 and the promoter of nearby genes, in addition to the complex restrictive enzyme map, 3C was performed separately for the upstream and the downstream regions of rs6592645. For upstream region 3C, HindIII (New England Biolabs) was chosen to digest chromatin. The two BACs RP11-269F1 and RP11-795A14 (BACPAC Resources Center) were mixed with the same amount (10 µg for each) and used as control. By contrast, EcoRI (New England Biolabs) and BAC RP11-672A2 were utilized in downstream region 3C. In the current study, unidirectional primers were designed to anchor three protein-coding genes [GVQW motif-containing 3 (GVQW3), THAP domain-containing 12 (THAP12) and EMSY] for the upstream region, and one protein-coding gene (LRRC32) for the downstream one, this enhancer and several random genome regions (Tables SII and SIII). GVQW3 and THAP12 utilize different strands of the same fragment as the promoter. Therefore, these two genes occupied the same position in the assay.

The enrichment for chromatin was evaluated using the 2-ΔΔCq method (15). All PCR products were verified by sequencing. Three repeats were performed for each unidirectional anchor primer.

The potential unknown genes were searched in two lncRNA databases; NONCODE (http://www.noncode.org) and GENCODE (https://www.gencodegenes.org).

Tissue collection and LRRC32 expression quantification

A total of 46 lung tissues were collected from Department of Respiratory Critical Care Medicine, the First Affiliated Hospital of Kunming Medical University (Kunming, China) between January 2016 and June 2016. The detail of the donors can be found in Table I. All donors were suspected of lung cancer and thus tissues were sectioned for pathological examination. After examination, the remaining tissues were used for research. Among the donors, 22 individuals were diagnosed as asthma according to global initiative for asthma (16). None of the patients were eventually diagnosed with lung cancer and no other comorbidities were reported. All available tissues (22 asthma and 24 controls) were included, and no individuals were excluded. All participants were Han Chinese and provided written informed consent. The present study was approved by ethics committee of Shaanxi Normal University (approval number 20170308).

Table I

Characteristics of the tissue donors.

Table I

Characteristics of the tissue donors.

CharacteristicAsthma groupControl group
Sample size2224
Sex (M/F)12/1014/10
Age distribution51-7047-69

[i] M, male; F, female.

RNA was isolated by TRIzol® (Thermo Fisher Scientific, Inc.). cDNA was generated by reverse transcription with RevertAid First Strand cDNA Synthesis Kit (Thermo Fisher Scientific, Inc.). LRRC32 expression was quantified by qPCR with primer pair 5'-GCATAGCAACGTGCTGATGGAC-3' and 5'-GATGCTGTTGCAGCTCAGGTCT-3'; GAPDH expression was also measured as a control with primer pair 5'-GAAGGTGAAGGTCGGAGTC-3' and 5'-GAAGATGGTGATGGGATTTC-3'. qPCR was performed by relative standard curve approach (17); that is, a cDNA mixture was serial diluted and thus a standard curve was generated. Gene expression was determined by the position of Cq in standard curve. The reagent and thermocycling condition were the same as the aforementioned qPCR. A total of three repeats were performed for each individual.

RNA-seq and expression quantitative trait locus (eQTL) analysis

RNA-seq data (SRA format) for lymphoblastoid cell lines (LCL) from two studies (18,19) were obtained from the SRA database (https://www.ncbi.nlm.nih.gov/sra; accession numbers: PRJNA357867 and PRJNA122271) and converted into fastq format by SRA toolkit (https://github.com/ncbi/sra-tools). After alignment with LRRC32 mRNA sequence (GenBank ID NM_005512.3) by bowtie2 (http://bowtie-bio.sourceforge.net/bowtie2/index.shtml), the expression was calculated by eXpress (https://pachterlab.github.io/eXpress) using the default parameters and reported as fragments per kilobase of transcript per million fragments mapped (FPKM). The genotype for LCL was obtained from the HapMap project (https://www.genome.gov/10001688/international-hapmap-project), and linear regression was performed between genotype and LRRC32 expression by SPSS 20.0 Statistics (IBM Corp.).

Chromatin immunoprecipitation (ChIP)

The online program Match (http://www.gene-regulation.com/cgi-bin/pub/programs/match/bin/match.cgi) was used to identify potential transcription factors. ChIP was carried out by EZ ChIP Kit (MilliporeSigma) according to the manufacturer's protocol. Briefly, ~1x107 Beas-2B cells were cross-linked with formaldehyde (1% final concentration) at room temperature for 10 min. After washing with phosphate buffered saline (Beijing Solarbio Science & Technology Co., Ltd.), cells were scraped, lysed by lysis buffer (MilliporeSigma; cat. no. 20-163), sonicated (2% magnitude, 60 sec at 4˚C) into small fragments (~400-800 bp) and precleared with 60 µl protein A beads (MilliporeSigma; cat. no. 16-201C) by centrifuge at 3,000 x g for 1 min at 4˚C. The protein/chromatin complex (1 ml) was captured by adding 2 µg mouse anti-transcription factor 3 (TCF3) antibody (Santa Cruz Biotechnology, Inc.; cat. no. sc-133074) or 2 µg normal mouse IgG (Santa Cruz Biotechnology, Inc.; cat. no. sc-2025) as a control, and precipitated with 60 µl protein A beads. Following washing with 1 ml Low Salt, High Salt, LiCl and TE wash buffers (MilliporeSigma; cat. nos. 20-154, 20-155, 20-156 and 20-157; respectively), the immunoprecipitated protein/chromatin complex was dissolved and de-crosslinked by adding 8 µl 5M NaCl and incubating at 65˚C overnight. Protein was removed by proteinase K (Roche Diagnostics) digestion and DNA was purified by supplied column in aforementioned EZ ChIP Kit. In brief, DNA was binding to the spin filter, washed by reagent A and resolved in reagent B. qPCR was used to evaluate the enrichment of the obtaining DNA with primer pair 5'-ATAGCATTAGATTGTTGTTCTGC-3' and 5'-ACGGAGATGATGGGTGAGA-3'. This primer pair was designed to bind rs6592645 surrounding region. The qPCR was performed by relative standard curve method (17). The reagent and thermocycling condition were identical with the aforementioned qPCR. Three repeats were performed for each experiment.

Electrophoretic mobility shift assay (EMSA)

Nuclear extracts were prepared and quantified using a Nuclear and Cytoplasmic Protein Extraction kit (Beyotime Institute of Biotechnology; cat. no. P0027). The probes for both alleles of rs6592645 are shown in Table SIV; they were synthesized by Sangon Biotech Co., Ltd., labeled with biotin using an EMSA probe biotin labeling kit (Beyotime Institute of Biotechnology, cat. nos. GS008) and incubated with nuclear extracts (5 µg) of Beas-2B cells at 37˚C for 30 min. The probe/protein complex was separated by electrophoresis in nondenaturing polyacrylamide gel (6%) and transferred to nylon membranes (Beyotime Institute of Biotechnology). For each allele, two controls were also included. One control was biotin-labeled probes alone while the other was probe/protein complex incubating with competitor oligonucleotides (non-labeled probes). After incubating with streptavidin-HRP conjugate (Beyotime Institute of Biotechnology; cat. no. GS009), the membrane was visualized in ECL chemiluminescence (MilliporeSigma).

Statistical analysis

One-way ANOVA with Tukey's test was used to compare the data from multiple groups, including luciferase activity, 3C and partial LRRC32 expression. Independent Student's t-tests were utilized to compare the difference of two groups, including gene expression and ChIP results. All statistical analyses were performed using SPSS 20.0 (IBM Corp.). P#x003C;0.05 was considered to indicate a statistically significant difference.

Results

Genetic variations near rs7130588

As the causal SNP(s) for asthma might be those in LD with rs7130588 (20,21), the present study investigated the LD pattern in representative 1KGP populations. Within the 500 kb region surrounding rs7130588, there are 2,253 SNPs for CEU, 2,279 SNPs for CHB and 3,776 SNPs for YRI. Among these SNPs, one was located ~0.3 kb away, rs6592645, and showed complete LD with rs7130588 (r2=1 for all three populations; Fig. S1), which indicated that rs6592645 and rs7130588 appear simultaneously. Therefore, rs6592645 should also present differentially distribution in case and control groups and be associated with asthma.

Another three SNPs were located ~30.6 kb away, including rs7926914, rs7927894 and rs7927997, which are in complete LD with each other (r2=1 for all three populations) and are associated with Crohn's disease (22) and atopic dermatitis (23), presented a strong LD with rs7130588 in CEU (r2=0.937) but relatively weak LD in CHB (r2=0.373) and YRI (r2=0.309) populations. All other SNPs had relatively low LD with rs7130588 in these three populations (all r2#x003C;0.530). Considering this LD pattern and that these three SNPs in the microarray (or the tag one, rs7927894) fail to reach a genome-wide significance level in GWAS for asthma (24), we hypothesized that rs7130588 and rs6592645 may be potential causal SNPs for this disease.

Function of rs7130588 and rs6592645

Since both rs7130588 and rs6592645 are located in intergenic region of EMSY and LRRC32, it was hypothesized that they might regulate gene expression by altering enhancer activity (21). To investigate this, the present study cloned the segment containing rs7130588 and rs6592645 (~1.5 kb), created plasmids with the corresponding alleles, performed transient transfections and subsequent dual-luciferase assays. ANOVA indicated that there was significant difference among the luciferase activity of these three plasmids (P=0.000049; Fig. 1). To explore the functional SNP, the present study further pairwisely compared the luciferase activity by ANOVA with Tukey's test. As shown in Fig. 1, the mutagenesis at rs7130588 failed to alter the relative luciferase activity when compared with the original plasmid construct (P=0.839), thus indicating that rs7130588 is not functional in lung cell and unlikely to be the causal SNP for asthma. By contrast, the A allele of rs6592645 presented a significant increase in relative luciferase activity compared with the G allele (P=0.000093), thus suggesting that rs6592645 may be a causal SNP for asthma. Since the risk allele is G of rs7130588 in GWAS (2) and that G of rs7130588 was in complete LD with A of rs6592645, the causal variation for asthma should be A of rs6592645.

Gene interaction with the enhancer containing rs6592645

Since rs6592645 is within the intergenic region of EMSY and LRRC32, it was hypothesized that this mutation might be within an enhancer region and may have the ability to modify enhancer activity (21). In addition, the histone modification for rs6592645 surrounding region was searched in ENCODE project (https://www.encodeproject.org), which includes different ChIP-seq results for multiple cell lines. Since Beas-2B cell line was not included in ENCODE, lung cancer cell line A549 was searched due to the similar origin of these two cell lines. As shown in Fig. S2, there are evident histone 3 lysine 4 (H3K4) monomethylation and H3K27 acetylation signals, which are two common histone modifications on active enhancers (25), surrounding rs6592645 region in lung cell. However, the regulatory target remained undetermined. To investigated this, 3C was performed to determine the potential spatial contacts. Owing to the long distance between rs6952645 and nearby gene (EMSY and LRRC32) promoter, it was hard to perform 3C in one experiment. Therefore, the nearby region was divided into upstream and downstream regions and these were investigated separately.

As shown in Fig. 2A, in the upstream region, the GVQW3/THAP12 (the fifth point on the x-axis, ~179.1 kb away from rs6592645) and the EMSY (the eighth point on the x-axis, ~115.0 kb away from rs6592645) promoters failed to show any increases in ligation efficiency, thus indicating that none of them are the target of this enhancer. By contrast, the fourth, sixth and seventh point on the x-axis displayed some increase in ligation frequency (Fig. 2A). However, no known protein-coding genes are within these three restriction fragments. Previous studies have suggested that long non-coding RNA (lncRNA; RNA with length >200 base pairs but without protein-coding function) are also involved in the onset of asthma (26,27). Therefore, we hypothesized that there might be a novel lncRNA within this region. To reveal the potential gene, this segment was searched in two lncRNA databases; NONCODE and GENCODE. However, no lncRNAs were observed to be within this segment. Therefore, it was considered that this interaction in space between this segment and the enhancer containing rs6592645 might be a random one and without biological sense, which might result from the affinity between these two genome regions and the same protein complex.

For the downstream region, the promoter of LRRC32 showed a strong increase in ligation efficiency (the seventh point on the x-axis, ~110.0 kb away from rs6592645; Fig. 2B). Using ANOVA to compare the interaction efficiency of this point with nearby ones, a significant difference can be observed (P#x003C;0.001), which indicated that the putative regulation target of this enhancer may be LRRC32.

LRRC32 expression between asthma and controls

Results from the luciferase assay indicated that the putative causal allele, A of rs6592645, may induce high gene expression. If LRRC32 is indeed involved in asthma onset, then this gene should be overexpressed in patients with asthma. To validate this, lung tissues from 22 patients with asthma and 24 healthy controls were collected, and LRRC32 expression levels were quantified. As shown in Fig. 3, LRRC32 expression in patients with asthma was significantly higher compared with that in controls (P=0.0095), thus confirming this hypothesis.

Previous GWAS analysis has suggested that rs7130588 is significantly associated with atopic asthma, thus indicating that this locus may be a risk factor mainly for allergic asthma (2). Therefore, it is useful to compare LRRC32 expression among allergic asthma and controls. Among the cohort of the present study, only six individuals were diagnosed with allergic asthma. ANOVA indicated that there was significant difference among LRRC32 expression of these three groups (P=0.030). Further ANOVA with Tukey's test indicated that no significant difference was observed in LRRC32 expression between allergic asthma and non-allergic asthma (P=0.55) or control (P=0.25; Fig. S3). By contrast, LRRC32 expression in non-allergic asthma was significantly higher compared with the control patients (P=0.0060; Fig. S3). However, there must be caution in interpreting this result, as the sample size was too small for allergic asthma.

LRRC32 expression upon stimulation

A number of environmental factors, including house dust mites (HDMs) and rhinoviruses (RVs), can induce asthma. To investigate whether LRRC32 may be involved in the response of these two environmental factors, two RNA-seq datasets were downloaded from SRA database (accession number PRJNA379624 and PRJNA700069) and LRRC32 expression was analyzed as aforementioned. In one dataset, the authors collected peripheral blood mononuclear cells (PBMCs) from a number of individuals and treated them with HDM extract for 48 h (28). As shown in Fig. S4, LRRC32 expression in HDM extract treatment group was significantly higher compared with that in the control group (P=6.40x10-9). In another cohort, the authors isolated PBMCs and stimulated with RV for 24 h (29). As shown in Fig. S5, LRRC32 expression in RV stimulated group was significantly higher compared with that in the non-stimulated group (P=0.000034). These data indicated that LRRC32 may be involved in the immune response to HDM or RV.

eQTL analysis

If rs6592645 can indeed influence LRRC32 expression, this locus could be an eQTL for this gene (21). Since enough lung tissues with known genotype and expression data were not available for the present study, the well-established model using LCL (30) cells was used to elucidate this. Therefore, two published RNA-seq data for LCL were downloaded for analysis (18,19) and the results are shown in Fig. 4. Since the individuals are not included in 1KGP and rs6592645 genotype is not available in HapMap project for one study (19), rs7130588 genotype was retrieved from HapMap for analysis. This study includes three populations, CEU, YRI and CHS (Southern Han Chinese) (19). Since the genotype for the CHS individuals from that study are not available from 1KGP or HapMap, only the CEU and YRI patient data were included for the present study analysis. In CEU, no association was observed between rs7130588 genotype (A/A and A/G) and LRRC32 expression (P=0.45; data not shown). In YRI, owing to the low frequency (~20%) of rs7130588 G allele, no individuals were identified as homozygous for this allele. Therefore, the present study used independent samples t-test to compared LRRC32 expression levels between A/A and A/G groups. As shown in Fig. 4A, LRRC32 expression in A/G group (n=34) was significantly higher compared with that in the A/A group (n=11; P=0.003), which indicated that G allele of rs7130588 was associated with higher LRRC32 expression. Since G of rs7130588 is in complete LD with A of rs6592645 (Fig. S1), it can be concluded that A of rs6592645 is also associated with higher LRRC32 expression, which is consistent with our luciferase assay (Fig. 1). In another YRI RNA-seq dataset (18), LRRC32 expression in A/G group (n=31) was significantly higher compared with that in A/A group (n=13; P=0.03; Fig. 4B), which verified that this locus may be an eQTL for LRRC32, at least in YRI population.

Transcription factor binding rs6592645

Considering the location of rs6592645, it was hypothesized that this mutation might be within a transcription factor binding site and thus may alter interaction affinity (21). Only TCF3 was predicted to reside in the surrounding region of rs6592645. To verify this, ChIP was performed with anti-TCF3 antibodies and the relative chromatin enrichment quantified for rs6592645 surrounding region. As shown in Fig. 5, the amount of immunoprecipitated chromatin samples were significantly higher in the TCF3 antibody experiment compared with IgG (P=0.025), thus suggesting that TCF3 may interact with rs6592645 surrounding region in lung cells.

TCF3 binding affinity difference between rs6592645 alleles

To investigate the binding efficiency between two alleles of rs6592645, EMSA was performed. As shown in Fig. 6, the two alleles of rs6592645 showed bands with evidently different density, which indicated apparent different affinity with nuclear proteins from Beas-2B cells. The high expression allele, A of rs6592645, possesses a much higher band density, which suggested a stronger binding affinity.

Association between rs11236797 genotype and LRRC32 expression

In the ~10.6-31.1 kb downstream region of rs6592645, there was another LD block containing 16 SNPs (rs2212434, rs61893460, rs7126418, rs7110818, rs7114362, rs7936070, rs7936312, rs7936323, rs7936434, rs4494327, rs11236791, rs10160518, rs2155219, rs11236797, rs7931483 and rs7930763; pairwise r2>0.85; Fig. S1) in CEU. This LD block displays a moderate LD with rs6592645 (r2#x003C;0.62 for all SNPs; Fig. S1) in CEU. In recent GWAS for Caucasians, this LD block is proposed to be associated with grass sensitization, self-reported allergy, allergic sensitization, eosinophilic esophagitis, alopecia areata, giant cell arteritis, atopic march, gut microbiome, food allergy, asthma and the aforementioned autoimmune disorders (https://www.ebi.ac.uk/gwas/variants/rs11236797). Through functional genomics work, rs11236797 was proposed to be a causal SNP that can also regulate LRRC32 expression in Treg cells (31), which is similar with the function of rs6592645. Considering the LD pattern and the 28.6 kb distance between rs6592645 and rs11236797, rs11236797 may be within an independent cis-regulatory element for LRRC32 with rs6592645. However, eQTL analysis was not conducted in a previous study (31). Notably, the GTEx search also failed to verify the association between this locus and LRRC32 expression (data not shown).

Therefore, the present study also used the aforementioned LCL RNA-seq datasets to perform eQTL analysis. Since the genotype of rs11236797 is not available in HapMap, the genotype of tag SNP rs2155219 was retrieved for analysis. In one cohort (19), there was no significant association between rs2155219 genotype and LRRC32 expression in CEU (P=0.776) or YRI (P=0.332; data not shown). In another cohort (18), rs2155219 genotype is strongly associated with LRRC32 expression (linear regression analysis, r=0.895, P#x003C;1x10-6; Fig. S6). In addition, the risk allele, T of rs2155219 (or A in reverse strand; see GWAS catalog), is associated with a higher LRRC32 expression (Fig. S6), which is similar to the results of the present study.

Discussion

The present study identified a potential causal SNP for asthma at 11q13.5 by population genetics and dual luciferase assay. Using multiple functional genomics approaches, the regulation target and underlying mechanism of the putative enhancer were identified. These data established a putative connection between a genetic marker in this locus and asthma susceptibility.

TGF-β is a pleiotropic cytokine involved in immune response, cell proliferation and differentiation, apoptosis, carcinogenesis and other physiological processes (32). Considering the role of immune response in asthma onset, TGF-β has been suggested to be a risk factor and therapeutic target for asthma (33). LRRC32 is a transmembrane cell surface protein and the docking receptor for latent TGF-β (non-covalently bound of mature TGF-β and latency-associated peptide; the latent form of TGF-β cannot bind with TGF-β receptor) (34). Upon binding, LRRC32 can tether latent TGF-β and enhance the activation of mature TGF-β (29). Overexpression of LRRC32 can enhance TGF-β bioactivity, especially in regulatory T lymphocytes (Treg) (35-37). These data suggest an essential role of LRRC32 in immune system (38,39). In this locus, the risk allele A of rs6592645, can induce a significantly higher LRRC32 expression level, which may result in an exaggerated response and further asthma susceptibility. In this regard, it was reported that rs7130588 is significantly associated with serum IgE level (14), which was consistent with the hypothesis of the present study.

eQTL analysis from the present study indicated that this locus is significantly associated with LRRC32 expression only in YRI population. Therefore, we hypothesized that there might be some negative regulatory element(s) in CEU but not in YRI population which can attenuate the association. Notably, a search in GTEx database (https://gtexportal.org) and one previous study (40) also failed to verify this association (data not shown), which might be due to the origin of tissues in the cohorts. Indeed, only European individuals were used to investigate the association between rs7130588 and LRRC32 or EMSY expression (40).

A number of asthma-related loci have been suggested to produce their effect by influencing the immune system and are thus proposed to be associated with some other autoimmune diseases, such as allergic diseases and atopic dermatitis (26,41,42). In recent GWAS, the rs7130588 locus has been reported to be associated with increased white cell count or percentage (43-45), inflammatory bowel disease (4,46-48), Crohn's disease (46-49), ulcerative colitis (46-49), eczema or atopic dermatitis (4,42,50) and allergic rhinitis (4,51), which probably result from the cis-regulation of LRRC32.

Supplementary Material

Linkage disequilibrium pattern in the rs7130588 surrounding region. Results for population (A) CEU, (B) CHB and (C) YRI, respectively. Each square denotes one SNP. The colors from white to black correspond to r2-values between 0 and 1. The vertical arrows indicate the position of rs7130588. CEU, Utah Residents with European Ancestry; CHB, Han Chinese in Beijing; YRI, Yoruba in Ibadan.
Histone modification in lung cancer cell line A549 for the segment surrounding rs6592645. The yellow lines indicate the location of rs6592645. Each part represents one type of histone modification. The x-axis indicates genome coordinate in chromosome 11 and the start and end are shown below. The y-axis denotes the relative enrichment of chromatin immunoprecipitation followed by sequencing. H3K4me1, histone 3 lysine 4 (H3K4) monomethylation; H3K27Ac, histone 3 lysine 27 acetylation.
LRRC32 expression distribution between allergic asthma, patients with non-allergic asthma and normal controls. The data are from the quantitative PCR of the present study and shown as a box plot. **P<0.01. LRRC32, leucine-rich repeat-containing 32.
Boxplot of LRRC32 expression (log transformed) between HDM extract treatment and control group in peripheral blood mononuclear cells. Data from (28) and shown as a box plot. **P<0.01. HDM, house dust mite; LRRC32, leucine-rich repeat-containing 32.
Boxplot of LRRC32 expression (log transformed) between RV stimulation and control group in peripheral blood mononuclear cells. Data from 29 and shown as a box plot. **P<0.01. LRRC32, leucine-rich repeat-containing 32; RV, rhinoviruses.
Association between rs2155219 genotype and relative LRRC32 expression in YRI population from literature. Data from (18) and shown as a box plot. As the data are in log-transformed form, some individuals display negative expression value. **P<0.01. LRRC32, leucine-rich repeat-containing 32.
Primer sequences for plasmid construction and mutagenesis.
Primer sequences for chromosome conformation capture-quantitative PCR in the upstream region of rs6592645.
Primer sequences for chromosome conformation capture-quantitative PCR in downstream region of rs6592645.
Probe sequences for the different alleles of rs6592645 used in electrophoretic mobility shift assays.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by The National Natural Science Foundation of China (grant no. 31370129).

Availability of data and materials

The luciferase activity, 3C, LRRC32 expression, ChIP and genotype from 1KGP datasets generated and/or analyzed during the current study are available in the Jianguoyun repository, https://www.jianguoyun.com/p/DSjM-N4Q_cv3BRj5r4QFIAA. All other datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

CS and WPF conceived and designed the present study and wrote the manuscript. YKL, YC, XQS performed luciferase, 3C and ChIP experiments. YKL analyzed the data. HYW, XXZ and KL performed LRRC32 expression and EMSA experiments. CS performed eQTL analysis and database search. YKL and HYW confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was reviewed and approved by the ethics committee of Shaanxi Normal University (approval no. 20170308; Xi'an, China). Informed consent was obtained from all subjects involved in the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Martinez FD and Vercelli D: Asthma. Lancet. 382:1360–1372. 2013.PubMed/NCBI View Article : Google Scholar

2 

Ferreira MA, Matheson MC, Duffy DL, Marks G, Hui J, Souëf PL, Danoy P, Baltic S, Nyholt DR, Jenkins M, et al: Identification of IL6R and chromosome 11q13.5 as risk loci for asthma. Lancet. 378:1006–1014. 2011.PubMed/NCBI View Article : Google Scholar

3 

Ferreira MAR, Mathur R, Vonk JM, Szwajda A, Brumpton B, Granell R, Brew BK, Ullemar V, Lu Y, Jiang Y, et al: Genetic architectures of childhood- and adult-onset asthma are partly distinct. Am J Hum Genet. 104:665–684. 2019.PubMed/NCBI View Article : Google Scholar

4 

Donertas HM, Fabian DK, Valenzuela MF, Partridge L and Thornton JM: Common genetic associations between age-related diseases. Nat Aging. 1:400–412. 2021.PubMed/NCBI View Article : Google Scholar

5 

Demenais F, Margaritte-Jeannin P, Barnes KC, Cookson WOC, Altmüller J, Ang W, Barr RG, Beaty TH, Becker AB, Beilby J, et al: Multiancestry association study identifies new asthma risk loci that colocalize with immune-cell enhancer marks. Nat Genet. 50:42–53. 2018.PubMed/NCBI View Article : Google Scholar

6 

Valette K, Li Z, Bon-Baret V, Chignon A, Bérubé JC, Eslami A, Lamothe J, Gaudreault N, Joubert P, Obeidat M, et al: Prioritization of candidate causal genes for asthma in susceptibility loci derived from UK Biobank. Commun Biol. 4(700)2021.PubMed/NCBI View Article : Google Scholar

7 

The 1000 Genomes Project Consortium. Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, Korbel JO, Marchini JL, McCarthy S, McVean GA and Abecasis GR: A global reference for human genetic variation. Nature. 526:68–74. 2015.PubMed/NCBI View Article : Google Scholar

8 

Binia A and Kabesch M: Respiratory medicine-genetic base for allergy and asthma. Swiss Med Wkly. 142(w13612)2012.PubMed/NCBI View Article : Google Scholar

9 

Meyers DA, Bleecker ER, Holloway JW and Holgate ST: Asthma genetics and personalised medicine. Lancet Respir Med. 2:405–415. 2014.PubMed/NCBI View Article : Google Scholar

10 

Kim KW and Ober C: Lessons learned from GWAS of asthma. Allergy Asthma Immunol Res. 11:170–187. 2019.PubMed/NCBI View Article : Google Scholar

11 

Willis-Owen SAG, Cookson WOC and Moffatt MF: The genetics and genomics of asthma. Annu Rev Genomics Hum Genet. 19:223–246. 2018.PubMed/NCBI View Article : Google Scholar

12 

Vicente CT, Revez JA and Ferreira MAR: Lessons from ten years of genome-wide association studies of asthma. Clin Transl Immunology. 6(e165)2017.PubMed/NCBI View Article : Google Scholar

13 

Ortiz RA and Barnes KC: Genetics of allergic diseases. Immunol Allergy Clin North Am. 35:19–44. 2015.PubMed/NCBI View Article : Google Scholar

14 

Li X, Ampleford EJ, Howard TD, Moore WC, Li H, Busse WW, Castro M, Erzurum SC, Fitzpatrick AM, Gasto B, et al: The C11orf30-LRRC32 region is associated with total serum IgE levels in asthmatic patients. J Allergy Clin Immunol. 129:575–578, 578 e571-579. 2012.PubMed/NCBI View Article : Google Scholar

15 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

16 

Global Initiative for Asthma: Global Strategy for Asthma Management and Prevention (2018 update). 2018 (https://ginasthma.org/wp-content/uploads/2019/01/2018-GINA.pdf).

17 

Green MR and Sambrook J: Constructing a standard curve for real-time polymerase chain reaction (PCR) experiments. Cold Spring Harb Protoc. 2018:836–842. 2018.PubMed/NCBI View Article : Google Scholar

18 

Pickrell JK, Marioni JC, Pai AA, Degner JF, Engelhardt BE, Nkadori E, Veyrieras JB, Stephens M, Gilad Y and Pritchard JK: Understanding mechanisms underlying human gene expression variation with RNA sequencing. Nature. 464:768–772. 2010.PubMed/NCBI View Article : Google Scholar

19 

Jadhav B, Monajemi R, Gagalova KK, Ho D, Draisma HHM, van de Wiel MA, Franke L, Heijmans BT, van Meurs J, Jansen R, et al: RNA-Seq in 296 phased trios provides a high-resolution map of genomic imprinting. BMC Biol. 17(50)2019.PubMed/NCBI View Article : Google Scholar

20 

Edwards SL, Beesley J, French JD and Dunning AM: Beyond GWASs: Illuminating the dark road from association to function. Am J Hum Genet. 93:779–797. 2013.PubMed/NCBI View Article : Google Scholar

21 

Spitz F and Furlong EE: Transcription factors: From enhancer binding to developmental control. Nat Rev Genet. 13:613–626. 2012.PubMed/NCBI View Article : Google Scholar

22 

Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, Rioux JD, Brant SR, Silverberg MS, Taylor KD, Barmada MM, et al: Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nat Genet. 40:955–962. 2008.PubMed/NCBI View Article : Google Scholar

23 

Esparza-Gordillo J, Weidinger S, Folster-Holst R, Bauerfeind A, Ruschendorf F, Patone G, Rohde K, Marenholz I, Schulz F, Kerscher T, et al: A common variant on chromosome 11q13 is associated with atopic dermatitis. Nat Genet. 41:596–601. 2009.PubMed/NCBI View Article : Google Scholar

24 

Marenholz I, Bauerfeind A, Esparza-Gordillo J, Kerscher T, Granell R, Nickel R, Lau S, Henderson J and Lee YA: The eczema risk variant on chromosome 11q13 (rs7927894) in the population-based ALSPAC cohort: A novel susceptibility factor for asthma and hay fever. Hum Mol Genet. 20:2443–2449. 2011.PubMed/NCBI View Article : Google Scholar

25 

Calo E and Wysocka J: Modification of enhancer chromatin: What, how, and why? Mol Cell. 49:825–837. 2013.PubMed/NCBI View Article : Google Scholar

26 

Li YK, Zhang XX, Yang Y, Gao J, Shi Q, Liu SD, Fu WP and Sun C: Convergent evidence supports TH2LCRR as a novel asthma susceptibility gene. Am J Respir Cell Mol Biol. 66:283–292. 2022.PubMed/NCBI View Article : Google Scholar

27 

Zhu Y, Mao D, Gao W, Han G and Hu H: Analysis of lncRNA expression in patients with eosinophilic and neutrophilic asthma focusing on LNC_000127. Front Genet. 10(141)2019.PubMed/NCBI View Article : Google Scholar

28 

Altman MC, Whalen E, Togias A, O'Connor GT, Bacharier LB, Bloomberg GR, Kattan M, Wood RA, Presnell S, LeBeau P, et al: Allergen-induced activation of natural killer cells represents an early-life immune response in the development of allergic asthma. J Allergy Clin Immunol. 142:1856–1866. 2018.PubMed/NCBI View Article : Google Scholar

29 

Murray LM, Thillaiyampalam G, Xi Y, Cristino AS and Upham JW: Whole transcriptome analysis of high and low IFN-α producers reveals differential response patterns following rhinovirus stimulation. Clin Transl Immunology. 10(e1356)2021.PubMed/NCBI View Article : Google Scholar

30 

Sie L, Loong S and Tan EK: Utility of lymphoblastoid cell lines. J Neurosci Res. 87:1953–1959. 2009.PubMed/NCBI View Article : Google Scholar

31 

Nasrallah R, Imianowski CJ, Bossini-Castillo L, Grant FM, Dogan M, Placek L, Kozhaya L, Kuo P, Sadiyah F, Whiteside SK, et al: A distal enhancer at risk locus 11q13.5 promotes suppression of colitis by Treg cells. Nature. 583:447–452. 2020.PubMed/NCBI View Article : Google Scholar

32 

Morikawa M, Derynck R and Miyazono K: TGF-β and the TGF-β family: Context-dependent roles in cell and tissue physiology. Cold Spring Harb Perspect Biol. 8(a021873)2016.PubMed/NCBI View Article : Google Scholar

33 

Al-Alawi M, Hassan T and Chotirmall SH: Transforming growth factor β and severe asthma: A perfect storm. Respir Med. 108:1409–1423. 2014.PubMed/NCBI View Article : Google Scholar

34 

Wang R, Zhu J, Dong X, Shi M, Lu C and Springer TA: GARP regulates the bioavailability and activation of TGFβ. Mol Biol Cell. 23:1129–1139. 2012.PubMed/NCBI View Article : Google Scholar

35 

Tran DQ, Andersson J, Wang R, Ramsey H, Unutmaz D and Shevach EM: GARP (LRRC32) is essential for the surface expression of latent TGF-beta on platelets and activated FOXP3+ regulatory T cells. Proc Natl Acad Sci USA. 106:13445–13450. 2009.PubMed/NCBI View Article : Google Scholar

36 

Metelli A, Wu BX, Fugle CW, Rachidi S, Sun S, Zhang Y, Wu J, Tomlinson S, Howe PH, Yang Y, et al: Surface expression of TGFbeta docking receptor GARP promotes oncogenesis and immune tolerance in breast cancer. Cancer Res. 76:7106–7117. 2016.PubMed/NCBI View Article : Google Scholar

37 

Probst-Kepper M, Geffers R, Kroger A, Viegas N, Erck C, Hecht HJ, Lünsdorf H, Roubin R, Moharregh-Khiabani D, Wagner K, et al: GARP: A key receptor controlling FOXP3 in human regulatory T cells. J Cell Mol Med. 13:3343–3357. 2009.PubMed/NCBI View Article : Google Scholar

38 

Metelli A, Salem M, Wallace CH, Wu BX, Li A, Li X and Li Z: Immunoregulatory functions and the therapeutic implications of GARP-TGF-β in inflammation and cancer. J Hematol Oncol. 11(24)2018.PubMed/NCBI View Article : Google Scholar

39 

Weissler KA and Frischmeyer-Guerrerio PA: Genetic evidence for the role of transforming growth factor-β in atopic phenotypes. Curr Opin Immunol. 60:54–62. 2019.PubMed/NCBI View Article : Google Scholar

40 

Li X, Hastie AT, Hawkins GA, Moore WC, Ampleford EJ, Milosevic J, Li H, Busse W, Erzurum SC, Kaminski N, et al: eQTL of bronchial epithelial cells and bronchial alveolar lavage deciphers GWAS-identified asthma genes. Allergy. 70:1309–1318. 2015.PubMed/NCBI View Article : Google Scholar

41 

Zhu Z, Lee PH, Chaffin MD, Chung W, Loh PR, Lu Q, Christiani DC and Liang L: A genome-wide cross-trait analysis from UK Biobank highlights the shared genetic architecture of asthma and allergic diseases. Nat Genet. 50:857–864. 2018.PubMed/NCBI View Article : Google Scholar

42 

Weidinger S, Willis-Owen SA, Kamatani Y, Baurecht H, Morar N, Liang L, Edser P, Street T, Rodriguez E, O'Regan GM, et al: A genome-wide association study of atopic dermatitis identifies loci with overlapping effects on asthma and psoriasis. Hum Mol Genet. 22:4841–4856. 2013.PubMed/NCBI View Article : Google Scholar

43 

Chen MH, Raffield LM, Mousas A, Sakaue S, Huffman JE, Moscati A, Trivedi B, Jiang T, Akbari P, Vuckovic D, et al: Trans-ethnic and ancestry-specific blood-cell genetics in 746,667 individuals from 5 global populations. Cell. 182:1198–1213 e1114. 2020.PubMed/NCBI View Article : Google Scholar

44 

Vuckovic D, Bao EL, Akbari P, Lareau CA, Mousas A, Jiang T, Chen MH, Raffield LM, Tardaguila M, Huffman JE, et al: The polygenic and monogenic basis of blood traits and diseases. Cell. 182:1214–1231 e1211. 2020.PubMed/NCBI View Article : Google Scholar

45 

Astle WJ, Elding H, Jiang T, Allen D, Ruklisa D, Mann AL, Mead D, Bouman H, Riveros-Mckay F, Kostadima MA, et al: The allelic landscape of human blood cell trait variation and links to common complex disease. Cell. 167:1415–1429 e1419. 2016.PubMed/NCBI View Article : Google Scholar

46 

Liu JZ, van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, Ripke S, Lee JC, Jostins L, Shah T, et al: Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 47:979–986. 2015.PubMed/NCBI View Article : Google Scholar

47 

Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, Lee JC, Schumm LP, Sharma Y, Anderson CA, et al: Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 491:119–124. 2012.PubMed/NCBI View Article : Google Scholar

48 

Peyrot WJ and Price AL: Identifying loci with different allele frequencies among cases of eight psychiatric disorders using CC-GWAS. Nat Genet. 53:445–454. 2021.PubMed/NCBI View Article : Google Scholar

49 

de Lange KM, Moutsianas L, Lee JC, Lamb CA, Luo Y, Kennedy NA, Jostins L, Rice DL, Gutierrez-Achury J, Ji SG, et al: Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat Genet. 49:256–261. 2017.PubMed/NCBI View Article : Google Scholar

50 

Sliz E, Huilaja L, Pasanen A, Laisk T, Reimann E and Mägi R: FinnGen; Estonian Biobank Research Team. Hannula-Jouppi K, Peltonen S, et al: Uniting biobank resources reveals novel genetic pathways modulating susceptibility for atopic dermatitis. J Allergy Clin Immunol. 149:1105–1112 e1109. 2022.PubMed/NCBI View Article : Google Scholar

51 

Li Y, Chen J, Rui X, Li N, Jiang F and Shen J: The association between sixteen genome-wide association studies-related allergic diseases loci and childhood allergic rhinitis in a Chinese Han population. Cytokine. 111:162–170. 2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

September-2023
Volume 26 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li Y, Wang H, Chen Y, Shi X, Zhang X, Li K, Fu W and Sun C: Single‑nucleotide polymorphism rs6592645 confers asthma risk through regulating <em>LRRC32</em> expression. Exp Ther Med 26: 451, 2023
APA
Li, Y., Wang, H., Chen, Y., Shi, X., Zhang, X., Li, K. ... Sun, C. (2023). Single‑nucleotide polymorphism rs6592645 confers asthma risk through regulating <em>LRRC32</em> expression. Experimental and Therapeutic Medicine, 26, 451. https://doi.org/10.3892/etm.2023.12150
MLA
Li, Y., Wang, H., Chen, Y., Shi, X., Zhang, X., Li, K., Fu, W., Sun, C."Single‑nucleotide polymorphism rs6592645 confers asthma risk through regulating <em>LRRC32</em> expression". Experimental and Therapeutic Medicine 26.3 (2023): 451.
Chicago
Li, Y., Wang, H., Chen, Y., Shi, X., Zhang, X., Li, K., Fu, W., Sun, C."Single‑nucleotide polymorphism rs6592645 confers asthma risk through regulating <em>LRRC32</em> expression". Experimental and Therapeutic Medicine 26, no. 3 (2023): 451. https://doi.org/10.3892/etm.2023.12150