Characteristics of B cell‑associated gene expression in patients with coronary artery disease

  • Authors:
    • Wenwen Yan
    • Haoming Song
    • Jinfa Jiang
    • Wenjun Xu
    • Zhu Gong
    • Qianglin Duan
    • Chuangrong Li
    • Yuan Xie
    • Lemin Wang
  • View Affiliations

  • Published online on: March 21, 2016     https://doi.org/10.3892/mmr.2016.5029
  • Pages: 4113-4121
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The current study aimed to identify differentially expressed B cell‑associated genes in peripheral blood mononuclear cells and observe the changes in B cell activation at different stages of coronary artery disease. Groups of patients with acute myocardial infarction (AMI) and stable angina (SA), as well as healthy volunteers, were recruited into the study (n=20 per group). Whole human genome microarray analysis was performed to examine the expression of B cell‑associated genes among these three groups. The mRNA expression levels of 60 genes associated with B cell activity and regulation were measured using reverse transcription‑quantitative polymerase chain reaction. The mRNA expression of the B cell antigen receptor (BCR)‑associated genes, CD45, NFAM, SYK and LYN, were significantly upregulated in patients with AMI; however, FCRL3, CD79B, CD19, CD81, FYN, BLK, CD22 and CD5 mRNA expression levels were significantly downregulated, compared with patients in the SA and control group. The mRNA levels of the T‑independent B cell‑associated genes, CD16, CD32, LILRA1 and TLR9, were significantly increased in AMI patients compared with SA and control patients. The mRNA expression of genes associated with T‑dependent B cells were also measured: EMR2 and CD97 were statistically upregulated, whereas SLAMF1, LY9, CD28, CD43, CD72, ICOSL, PD1, CD40 and CD20 mRNAs were significantly downregulated in AMI group patients compared with the two other groups. Additionally the gene expression levels of B cell regulatory genes were measured. In patients with AMI, CR1, LILRB2, LILRB3 and VAV1 mRNA expression levels were statistically increased, whereas, CS1 and IL4I1 mRNAs were significantly reduced compared with the SA and control groups. There was no statistically significant difference in B cell‑associated gene expression levels between patients with SA and the control group. The present study identified the downregulation of genes associated with BCRs, B2 cells and B cell regulators in patients with AMI, indicating a weakened T cell‑B cell interaction and reduced B2 cell activation during AMI. Thus, improving B2 cell‑mediated humoral immunity may be a potential target for medical intervention in patients with AMI.

Introduction

Atherosclerosis is a major cause of coronary artery disease, which is a leading cause of mortality in numerous countries (1). The presence of B lymphocytes during inflammatory infiltration in humans (2,3) and mouse models (4,5) has led to the investigation of their importance in atherosclerosis. However, the association between B cells and atherogenesis remains controversial. T-independent B cells (B1 cells) have atheroprotective properties (6,7), whereas T-dependent B cells (B2 cells) exhibit pro-atherogenic activity (8,9). The activation of B cells requires two signals. The first signal is the binding of an antigen to the B cell antigen receptor (BCR) expressed on the surface of B cells. If the antigen is thymus-independent, B cells can stimulate the second signal directly. However, thymus-dependent antigens require an interaction between T cells and B cells for the second signal to be initiated (10). B1 and B2 cells constitute the two predominant B cell types, and are considered to be part of the innate and adaptive immune systems, respectively (11). B1 cells are a minor group of B cells that produce natural antibodies against common microbial epitopes and self-determinants. By contrast, B2 cells are considered to be the conventional B lymphocytes, requiring signals from T helper cells to recognize specific antigens, and to produce and regulate antibody-mediated humoral immunity (12,13).

In the present study, the expression levels of a range of B cell-associated genes involved in B cell activation were examined. Human cDNA microarray analysis was used to detect the variations in gene expression levels at different stages of B cell activation and in subsets of peripheral blood mononuclear cells (PBMCs) isolated from patients with AMI and stable angina (SA), as well as healthy controls. The in vitro study was designed to investigate differential gene expression levels in B cells, and analyze the differences in humoral immunity in patients with AMI and SA.

Patients and methods

Patient information

The current study consisted of 3 groups of subjects: 20 patients with AMI, 20 patients with SA and 20 healthy volunteers. The baseline demographic data are presented in Table I. The patients with AMI were admitted to a Coronary Care Unit of Tongji Hospital (Shanghai, China), <12 h after the onset of symptoms between January and June 2013. The AMI group included 18 male and 2 female patients aged 58±12 years [mean ± standard deviation (SD)]. All patients with AMI were diagnosed on the basis of previously described criteria (14). Briefly, selected subjects exhibited a rise in cardiac biomarker parameters (preferably cardiac troponin) with at least one value above the 99th percentile reference limit and with at least one of the following features: i) Symptoms of ischemia; ii) new or presumed new significant ST-segment-T wave changes or new left bundle branch block; iii) development of pathological Q waves in the electrocardiograph; iv) imaging evidence of new loss of viable myocardium or new regional wall motion abnormality; and v) identification of an intracoronary thrombus by angiography.

Table I

Baseline demographic data in AMI, SA and control groups.

Table I

Baseline demographic data in AMI, SA and control groups.

Patient characteristicAMISAControlP-value
All groupsAMI vs. SA
Age, years57.8±11.963.6±9.928.8±3.30.0000.251
Gender (male/female), n18/218/217/30.8531.000
Body mass index, kg/m223.6±2.622.8±2.721.3±1.80.1020.560
Smoking history, n/day13.6±12.29.8±10.30±00.0000.648
Systolic blood pressure, mmHg128.6±15.3123.0±12.1120.8±7.20.1150.501
Diastolic blood pressure, mmHg67.0±8.073.0±8.071.6±3.20.0170.064
LDL-C, mmol/l2.5±1.02.1±0.82.9±0.50.3270.548
Triglycerides, mmol/l1.6±1.11.5±1.41.2±0.40.7300.762
HDL-C, mmol/l0.8±0.70.9±0.21.3±0.20.0000.803
Fasting blood glucose, mmol/l5.4±0.95.0±0.84.9±0.50.6100.082

[i] Data are presented as the mean ± standard deviation (excluding Gender). n=20 for all groups. AMI, acute myocardial infarction; SA, stable angina; LDL-C, low-density lipoprotein cholesterol; HDL-C, high-density lipoprotein cholesterol.

In the SA group, 18 male and 2 female patients of age 64±10 years (mean ± SD) with exclusively effort-associated angina were analyzed. Each patient exhibited positive exercise stress test results and at least one coronary stenosis was detected during angiography (>70% reduction of lumen diameter). There was no significant difference between AMI and SA patients (Table I) regarding age, gender, smoking status, body mass index, systolic blood pressure, diastolic blood pressure, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, triglycerides or fasting glucose.

The control group consisted of 17 male and 3 female healthy volunteers of age 29±3 years (mean ± SD), recruited during the same time period as patients with AMI and SA. Family histories, physical examination, electrocardiograph, chest radiography and routine chemical analysis demonstrated that the controls had no evidence of coronary heart disease.

SA group candidates were outpatients with risk factors of cardiovascular disease and verified by electrocardiogram, cardio angiography or treadmill exercise test. The control candidates were individuals living in nearby communities without any risk factors or history of cardiovascular diseases. The exclusion criteria for the three groups were venous thrombosis, hematological disorders, acute or chronic inflammatory diseases, intake of hormones or immunosuppressors and malignancy.

The study protocol was approved by the ethics committee of Tongji University (Shanghai, China) and informed consent forms were obtained.

Microarray gene expression analysis

A Human Gene Expression Microarray kit was purchased from Agilent Technologies, Inc. (cat no. G4112F; Santa Clara, CA, USA). The microarray was composed of >41,000 genes or transcripts, including 19,596 targeted Entrez gene RNAs. The sequence information used in the microarrays was derived from the latest databases of RefSeq (www.ncbi.nlm.nih.gov/refseq), GoldenPath (genome.ucsc.edu/index.html), Ensembl (www.ensembl.org) and Unigene (www.ncbi.nlm.nih.gov/unigene). The functions of >70% of the genes in the microarray had been previously established. All patients were subjected to the microarray analysis.

Total RNA isolation

Peripheral blood samples (5 ml) were collected from patients with AMI and SA in PAXgene tubes (BD Biosciences, San Jose, CA, USA) immediately after admission. The control group blood samples were collected from nearby communities during the same period as AMI/SA blood collection in the hospital. Briefly, the leukocytes were isolated through density gradient centrifugation with Ficoll solution (Sigma Aldrich, St. Louis, MO, USA) and the remaining red blood cells were destroyed with erythrocyte lysis buffer (Qiagen GmbH, Hilden, Germany). Total RNA was extracted and purified using a PAXgene Blood RNA kit (cat no. 762174; Qiagen GmbH), according to the manufacturer's protocol. RNA integrity number (RIN) was measured to analyze RNA integration using an Agilent Bioanalyzer 2100 (Agilent Technologies, Inc.). Samples were considered to be high quality when RIN was ≥7.0 and 28S/18S was ≥1.7.

RNA amplification and labeling

Total RNA was amplified and labeled with Cy3 using a by Low Input Quick Amp Labeling kit, one-color (cat no. 5190-2305; Agilent Technologies, Inc.) according to the manufacturer's protocol. Labeled cRNA was purified using an RNeasy Mini kit (cat no. 74106, Qiagen GmbH).

Microarray hybridization

Each microarray slide was hybridized with 1.65 µg Cy3-labeled cRNA using a Gene Expression Hybridization kit (cat no. 5188-5242) in a hybridization oven (cat no. G2545A) at 37°C, both purchased from Agilent Technologies, Inc., according to the manufacturer's protocol. After 17 h of hybridization, the slides were washed in staining dishes (cat no. 121; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with a Gene Expression Wash Buffer kit (cat no. 5188-5327; Agilent Technologies, Inc.), according to the manufacturer's protocol.

Microarray scanning and data acquisition

The microarray slides were scanned with an Agilent Microarray Scanner (cat no. G2565CA; Agilent Technologies, Inc.) using the default settings (dye channel, green; scan resolution, 3 µm; 20 bit). Data were extracted using Feature Extraction software (version 10.7; Agilent Technologies, Inc.). Raw data were normalized using the Quantile algorithm function of Gene Spring software (version 11.0; Agilent Technologies, Inc.).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

The spots on the microarray were randomly selected and their mRNA expression levels were confirmed by RT-qPCR. Briefly, reverse transcription was performed using the iScript reverse transcription supermix kit (Bio-Rad Laboratories, Inc., Hercules, CA, USA; cat. no. 1708841). The Ambion TURBO DNA-free DNase treatment kit (Thermo Fisher Scientific, Inc., Waltham, MA, USA; cat. no. AM1907) was utilized to remove contaminating DNA from RNA preparations according to the manufacturer's instructions. Real-time PCR was performed on a CFX96 Touch Real-time PCR detection system (Bio-Rad Laboratories, Inc.; cat. no. 1855195) using the iQ SYBR Green Supermix, (Bio-Rad Laboratories, Inc.; cat. no. 1708880) according to the manufacturer's instructions. The cycling conditions for RT-qPCR were as follows: 95°C for 30 sec, 95°C for 5 sec, 60°C for 5 sec and plate read, 40 cycles at 95°C for 30 sec, melting curve at 65–95°C, increment 0.5°C for 5 sec and plate read. Of the genes with differential expression levels, three were randomly selected, and these genes and a reference gene (GAPDH) were subjected to RT-qPCR. The relative expression levels were indicated as the expression of the target genes normalized to the expression of GAPDH (2−ΔΔCq). The melting curve and the 2−ΔΔCq method (15) were used to detect differences in gene expression levels among the three groups. The results from RT-qPCR were consistent with the microarray analysis (data not shown).

Statistical analysis

Values are expressed as the mean ± SD. A one-way analysis of variance (ANOVA) was used to examine differences between the groups. Pair-wise group comparisons following ANOVA were performed using Tukey's multiple comparison tests. Data were analyzed using SPSS version 17.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

mRNA expression levels of genes associated with BCR

The present study measured the expression levels of 22 genes associated with the BCR in patients with AMI and SA, as well as controls subjects (Table II; Fig. 1). In PBMCs from the three groups, the expression levels of 15 genes were statistically different (P<0.05): CD45, NFAM1, BTK, SYK, LYN, FCRL3, CD79A, CD79B, CD19, CD21, CD81, FYN, BLK, CD22 and CD5. Compared with the control subjects, the gene expression levels of CD45, NFAM1, BTK, SYK, and LYN were significantly upregulated (P<0.05), whereas CD79A, CD21 (P<0.05), FCRL3, CD79B, CD19, CD81, FYN, BLK, CD22 and CD5 (P<0.01) mRNA expression were downregulated in the AMI group. In PBMCs from AMI patients, the expression of CD45, NFAM1, SYN and LYN were statistically increased (P<0.01), whereas FCRL3, CD79B, CD19, CD81, FYN, BLK, CD22 and CD5 expression were significantly reduced (P<0.05) compared with the levels in PBMCs from the SA group. There were no significant differences in BCR-associated mRNA expression levels between the SA and control group.

Table II

Expression of genes associated with B cell antigen receptor in B cell activation.

Table II

Expression of genes associated with B cell antigen receptor in B cell activation.

Gene IDGene expressiona
P-value
AMISAControlAll groupsAMI vs. controlAMI vs. SASA vs. control
BLK8.85±0.669.39±0.529.64±0.540.0000.0000.0090.359
BLNK6.31±0.376.50±0.556.63±0.430.0860.0720.3640.655
BTK8.58±0.268.38±0.318.33±0.320.0270.0290.1020.846
NFAM17.29±0.606.59±0.466.51±0.440.0000.0000.0000.873
LYN14.52±0.3613.74±0.4513.49±0.280.0000.0000.0000.099
CD59.26±0.529.80±0.349.82±0.290.0000.0000.0000.984
CD1910.49±0.5911.12±0.5111.53±0.550.0000.0000.0020.057
CD219.09±1.739.69±1.3510.23±1.000.0420.0320.3720.436
CD4513.63±0.7513.01±0.4812.81±0.200.0000.0000.0010.233
CD226.38±0.556.93±0.577.17±0.600.0000.0000.0090.517
CD79A13.87±1.6714.59±1.2115.08±0.770.0140.0200.3390.347
CD79B8.76±0.399.29±0.529.51±0.440.0000.0000.0010.293
CD8111.34±0.4111.84±0.3311.87±0.330.0000.0000.0000.962
FYN11.94±0.3312.33±0.3212.34±0.220.0000.0000.0000.099
FCRL17.08±0.427.33±0.567.35±0.530.1880.2300.2760.993
FCRL27.60±0.387.80±0.498.00±0.490.1800.1540.6700.566
FCRL37.43±0.687.96±0.598.04±0.520.0040.0060.0200.892
FCRL42.66±1.152.70±0.812.81±1.150.9010.8990.9930.944
FCRL55.39±0.825.57±0.685.64±0.810.6070.5970.7650.960
LAX5.85±0.556.12±0.705.90±0.630.3650.9820.3900.493
PIR3.12±1.012.47±0.512.69±0.530.1060.0540.2730.456
SYK12.26±0.2511.94±0.3111.87±0.210.0000.0000.0010.802

a Data are presented as the mean ± standard deviation. n=20 for all groups. AMI, acute myocardial infarction; SA stable angina.

mRNA expression levels of genes associated with B1 cells

The present study measured the expression levels of 7 genes associated with B1 cell activation in PBMCs from the AMI, SA and control groups (Table III; Fig. 2). In separate comparisons, CD16, CD32, LILRA1 and TLR9 mRNA levels were significantly increased in patients with AMI compared with SA and control patients (P<0.05). There was no statistical difference in the expression levels of genes associated with T1 cell activation between patients with SA and the control group.

Table III

Expression of genes associated with T-independent B cell activation.

Table III

Expression of genes associated with T-independent B cell activation.

Gene IDGene expressiona
P-value
AMISAControlAll groupsAMI vs. controlAMI vs. SASA vs. control
SLAMF87.90±0.407.98±0.457.65±0.450.0580.1630.8320.052
UBD2.63±0.472.48±0.352.35±0.250.0610.0720.5780.518
CD1616.48±0.6015.70±0.5615.50±0.430.0000.0000.0000.580
CD329.76±0.659.34±0.539.20±0.350.0040.0060.0330.708
CD1806.82±0.406.81±0.336.87±0.410.8840.9280.9940.884
LILRA111.90±0.2511.40±0.2611.40±0.340.0000.0000.0000.999
TLR97.14±0.366.71±0.326.75±0.260.0000.0010.0000.890

a Data are presented as the mean ± standard deviation. n=20 for all groups. AMI, acute myocardial infarction; SA stable angina.

mRNA expression levels of genes associated with B2 cells

The expression levels of 20 genes associated with B2 cell activation were examined in PBMCs from patients with AMI and SA, as well as control subjects (Table IV; Fig. 3). In PBMCs from the three groups, the expression levels of the following 11 genes were statistically different (P<0.05): EMR2, CD97, SLAMF1, LY9, CD28, CD43, CD72, ICOSL, PD1, CD40 and CD20. The mRNA expression levels of EMR2 and CD97 were significantly upregulated (P<0.01), whereas SLAMF1, LY9, CD28, CD43, CD72, ICOSL, PD1, CD40 and CD20 mRNAs were significantly downregulated (P<0.05), in the AMI group compared with the SA and control groups. There was no significant difference in the mRNA expression levels of genes associated with B2 cell activation between the SA and control groups.

Table IV

Expression of genes associated with T-dependent B cell activation.

Table IV

Expression of genes associated with T-dependent B cell activation.

Gene IDGene expressiona
P-value
AMISAControlAll groupsAMI vs. controlAMI vs. SASA vs. control
ICOSL7.96±0.488.32±0.358.59±0.340.0000.0000.0050.092
BTLA6.57±0.636.64±0.666.48±0.570.7260.9040.9260.704
CD208.83±0.409.37±0.539.45±0.400.0000.0000.0010.852
CD288.12±0.498.75±0.648.69±0.400.0010.0020.0050.926
CD3712.20±0.2712.30±0.2612.38±0.200.2140.2490.3170.988
CD406.56±0.456.99±0.387.07±0.300.0000.0000.0030.789
CD4310.00±0.5110.60±0.3410.70±0.300.0000.0000.0000.425
CD726.35±0.456.84±0.476.97±0.500.0000.0000.0070.667
CD805.20±1.255.26±1.315.73±1.000.3080.3400.9860.425
CD866.64±0.616.94±0.436.87±0.500.1650.3350.1670.911
CD9712.9±0.4912.3±0.4812.30±0.200.0000.0000.0011.000
CD2268.68±0.488.87±0.278.80±0.400.3000.5890.2750.839
CD2762.05±0.102.24±0.302.41±0.300.1200.2130.1450.976
LY98.35±0.348.84±0.208.94±0.280.0000.0000.0000.507
CTLA44.68±0.464.89±0.344.76±0.260.2010.7600.1780.521
EMR211.62±0.6110.60±0.5810.30±0.460.0000.0000.0000.175
TNFSF47.26±0.566.79±0.537.05±0.590.0510.3240.0530.324
TNFSF93.88±0.873.20±0.883.47±0.650.0540.1360.9300.063
PD18.96±0.419.49±0.499.30±0.400.0010.0420.0010.469
SLAMF18.62±0.319.36±0.329.20±0.430.0000.0000.0000.336

a Data are presented as the mean ± standard deviation. n=20 for all groups. AMI, acute myocardial infarction; SA stable angina.

mRNA expression levels of regulatory genes associated with B cell activation

In PBMCs from the AMI, SA and control groups, the expression levels of 11 regulatory genes associated with B cell activation were examined (Table V; Fig. 4), 8 of which exhibited significant differences between all groups (P<0.05). In patients with AMI, the expression levels of LILRB1, LILRA3 (P<0.05), CR1, LILRB2, LILRB3 and VAV1 (P<0.01) were significantly increased, whereas the expression of CS1 and IL4I1 were significantly reduced (P<0.05), compared with the levels in the control group. When compared with the SA group patients, the mRNA levels of CR1, LILRB2, LILRB3 and VAV1 were significantly upregulated in patients with AMI (P<0.01), while CS1 and IL4I1 mRNAs were significantly downregulated (P<0.05). There were no significant differences in the gene expression levels of regulators of B cell activation between patients with SA and control subjects.

Table V

Gene expression of regulators associated with B cell activation.

Table V

Gene expression of regulators associated with B cell activation.

Gene IDGene expressiona
P-value
AMISAControlAll groupsAMI vs. controlAMI vs. SASA vs. control
CR18.75±0.557.71±0.717.51±0.620.0000.0000.0000.577
BAFF10.35±0.3810.00±0.459.78±0.290.0700.0590.1110.055
LILRB17.75±0.437.60±0.297.45±0.350.0340.0250.3980.359
LILRB213.93±0.513.10±0.4313.09±0.390.0000.0000.0000.879
LILRB316.09±0.4615.30±0.4815.11±0.320.0000.0000.0000.313
LILRB45.66±0.585.28±0.625.55±0.600.1320.8300.1230.343
LILRA38.31±1.277.84±0.947.32±0.930.0170.0120.3460.272
LAIR19.79±0.349.81±0.299.85±0.220.8190.8170.9880.890
VAV112.41±0.3012.00±0.3112.03±0.210.0000.0000.0010.893
CS111.19±0.5711.61±0.4211.67±0.510.0070.0100.0250.941
IL4I17.50±0.507.92±0.448.16±0.510.0000.0000.0220.276

a Data are presented as the mean ± standard deviation. n=20 for all groups. AMI, acute myocardial infarction; SA stable angina.

Discussion

The BCR is composed of a membrane-bound immunoglobulin (Ig) sheathed by an Igα/Igβ heterodimer. The receptor is critical in mediating the development and activation of B cells (16). Ig recognizes antigens, and the induced signals are transmitted by Igα (CD79A) and Igβ. It was previously reported that high CD45 expression may reduce the expression of the BAFF and inhibit B cell survival (17,18). Furthermore, in vivo studies in mice indicated that overexpression of NFAM1 may severely impair early B cell development (19,20). LYN and FCRL3 appear to mediate positive and negative signaling during B cell activation (21,22), with a genetic ablation study indicating that LYN has an important inhibitory role in B cell signaling (23). In addition, CD19, CD21, CD81 and FYN are B cell co-receptors that enhance BCR signal transduction (2426). By contrast, the B cell specific Src-family kinase, BLK, and CD5, which specifically bind the ligand of B cell surface Ig, are dispensable during B cell development and activation (27,28).

In patients with AMI, the expression levels of CD45, NFAM1 and LYN were significantly increased, while CD79A, CD79B, CD19, CD21, CD81, FYN, BLK and CD5 were significantly decreased compared with the control group (Fig. 1). The results of the current study indicate that the expression of mRNAs associated with BCR signals were downregulated in patients with AMI, possibly leading to inhibition of B cell activation and development. A comparison between SA patients and the control group demonstrated no significant difference in the mRNA levels of genes involved in BCR antigen recognition, which indicates that there may be differential BCR antigen recognition activity in patients with AMI and SA.

B1 cells are considered to function in the innate response to infection by viruses and bacteria, and typically demonstrate preferential responses to T cell-independent antigens (10,12). Among the 7 B1 cell-associated genes investigated, the expression levels of CD16, CD32, LILRA1 and TLR9 mRNAs were significantly upregulated in PBMCs from AMI patients compared with the SA and control groups (Fig. 2). Previous studies have demonstrated that Fc γ receptors CD16 and CD32 may influence the growth and differentiation of B cells (29,30). LILRA1 mRNA transcripts were detected in B cells and the LILRA1 protein has been observed to activate cells by associating with the γ chain of Fc γ receptors (31,32). Furthermore, previous studies demonstrated that human B cells were activated by a TLR9 agonist (33,34). It was therefore proposed that B1 cell activation is enhanced in patients with AMI, suggesting that an innate-like immune response occurs in AMI. However, there was no significant difference in B1 cell-associated gene expression between the SA patients and the control group. Thus, the results suggest that B1 cell activity is different in AMI and SA.

B2 cells are important in B cell-specific humoral immunity (35). Among the 20 genes examined, the mRNA expression levels of EMR2 and CD97 were significantly upregulated in AMI patients compared with the SA and control groups (Fig. 3). EMR2 and CD97 are important for the interaction between T cells and B cells (36). The SLAM family, including SLAMF1 and LY9, together with CD28 and CD43, are essential in the development and maturation of B cells, and the control of the humoral immune response (3739). The ICOS-ICOSL and PD1-PDL pathways are important for the stimulation of effector T cell and B2 cell responses (4042). The association between CD40 and CD40L, which are expressed on the surface of activated T cells, promotes the activation of B cells (43). CD20 is a B cell-specific integral membrane protein that regulates B cell proliferation (44). In the present study, the levels of SLAMF1, LY9, CD28, CD43, ICOSL, PD1, CD40 and CD20 mRNAs were lower in AMI patients compared with the control group (Fig. 3). These results demonstrate that the T cell-B cell interaction was weakened during the B2 cell activation in AMI. No significant differences were observed between the SA and control group patients, indicating that the potential insufficiency of the humoral response only occurred in the B2 subset during AMI.

Among the 11 regulatory genes associated with B cell activation examined, the mRNA expression levels of CR1, LILRB2, LILRB3, LILRA3 and VAV1 were significantly increased in AMI patients compared with those in the SA and control groups (Fig. 4). CR1 is an inhibitor of BCR-mediated B cell activation (45) and LILRB negatively regulates the activation of antigen presenting cells (46,47). Additionally, it has been previously demonstrated that mice lacking VAV1 exhibit defects in B cell activation (48). Previous studies also observed that CS1 promotes the proliferation of human B lymphocytes and increases the expression of autocrine cytokines (20,4951). Thus, the results of the current study indicate that B cell activation is inhibited in patients with AMI. Furthermore, there was no significant difference in the mRNA levels of genes associated with B cell activation between the SA and control groups.

During the current study, standard gene testing procedures and appropriate statistical analyses were performed using randomly selected patients and controls, however, the study was not without limitations. Notably, there was an age difference between patients with AMI/SA and the control subjects. It is unclear whether aging affected the humoral response or whether the young control group had a normal immunity status compared with AMI/SA patients. However, the current data indicates that there was no significant difference in humoral immunity between patients with SA and the control group. The second limitation was the enhanced B1 cell activation observed in patients with AMI. B1 cells are a minor component of the total B cell population, which is a part of the innate immune response and produces natural antibodies. Thus, further in vitro studies are required to elucidate the pathological mechanisms of antigen processing pathways in the B cell system.

In conclusion, the statistical downregulation of genes associated with the BCR, B2 cells and B cell regulators in patients with AMI indicates that there is a weakened T cell-B cell interaction, as well as reduced B2 cell activation and development, during AMI. Notably, there were no statistical differences in the expression levels of B cell-associated genes between the SA and control groups, demonstrating that B2 cell dysfunction is only observable in AMI and not in SA. B2 cells are considered to be the conventional B lymphocytes, producing and regulating the antibody-mediated humoral immunity (12,13). Consequently, improving B2 cell-mediated humoral immunity may be considered as a potential target for the treatment of patients with AMI.

Acknowledgments

The present study was supported by the Shanghai Traditional Chinese Medicine 3-Year Development Program (2014–2016; grant no. ZY3-CCCX-3-3047).

Abbreviations:

BCR

B cell antigen receptor

AMI

acute myocardial infarction

PBMCs

peripheral blood mononuclear cells

SA

stable angina

Ig

immunoglobulin

References

1 

Murray CJ, Vos T, Lozano R, Naghavi M, Flaxman AD, Michaud C, Ezzati M, Shibuya K, Salomon JA, Abdalla S, et al: Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990–2010: A systematic analysis for the Global Burden of Disease Study 2010. Lancet. 380:2197–2223. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Kyaw T, Tay C, Khan A, Dumouchel V, Cao A, To K, Kehry M, Dunn R, Agrotis A, et al: Conventional B2 B cell depletion ameliorates whereas its adoptive transfer aggravates atherosclerosis. J Immunol. 185:4410–4419. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Clement M, Guedj K, Andreata F, Morvan M, Bey L, Khallou-Laschet J, Gaston AT, Delbosc S, Alsac JM, Bruneval P, et al: Control of the T follicular helper-germinal center B-cell axis by CD8+ regulatory T cells limits atherosclerosis and tertiary lymphoid organ development. Circulation. 131:560–570. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Paigen B, Morrow A, Holmes PA, Mitchell D and Williams A: Quantitative assessment of atherosclerotic lesions in mice. Atherosclerosis. 68:231–240. 1987. View Article : Google Scholar : PubMed/NCBI

5 

Thorp E, Cui D, Schrijvers DM, Kuriakose G and Tabas I: Mertk receptor mutation reduces efferocytosis efficiency and promotes apoptotic cell accumulation and plaque necrosis in atherosclerotic lesions of apoe−/− mice. Arterioscler Thromb Vasc Biol. 28:1421–1428. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Tsiantoulas D, Diehl CJ, Witztum JL and Binder CJ: B cells and humoral immunity in atherosclerosis. Circ Res. 114:1743–1756. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Kyaw T, Tay C, Krishnamurthi S, Kanellakis P, Agrotis A, Tipping P, Bobik A and Toh BH: B1a B lymphocytes are atheroprotective by secreting natural IgM that increases IgM deposits and reduces necrotic cores in atherosclerotic lesions. Circ Res. 109:830–840. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Kyaw T, Tay C, Hosseini H, Kanellakis P, Gadowski T, MacKay F, Tipping P, Bobik A and Toh BH: Depletion of B2 but not B1a B cells in BAFF receptor-deficient ApoE mice attenuates atherosclerosis by potently ameliorating arterial inflammation. PLoS One. 7:e293712012. View Article : Google Scholar : PubMed/NCBI

9 

Ait-Oufella H, Herbin O, Bouaziz JD, Binder CJ, Uyttenhove C, Laurans L, Taleb S, Van Vré E, Esposito B, Vilar J, et al: B cell depletion reduces the development of atherosclerosis in mice. J Exp Med. 207:1579–1587. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Janeway CA Jr, Travers P, Walport M and Shlomchik MJ: Immunobiology. 5th edition. Garland Science; New York: 2001

11 

Sage AP and Mallat Z: Multiple potential roles for B cells in atherosclerosis. Ann Med. 46:297–303. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Cerutti A, Cols M and Puga I: Marginal zone B cells: Virtues of innate-like antibody-producing lymphocytes. Nat Rev Immunol. 13:118–132. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Tsiantoulas D, Sage AP, Mallat Z and Binder CJ: Targeting B cells in atherosclerosis: Closing the gap from bench to bedside. Arterioscler Thromb Vasc Biol. 35:296–302. 2015. View Article : Google Scholar

14 

Thygesen K, Alpert JS, Jaffe AS, Simoons ML, Chaitman BR and White HD: Joint ESC/ACCF/AHA/WHF Task Force for Universal Definition of Myocardial Infarction: Third universal definition of myocardial infarction. J Am Coll Cardiol. 60:1581–1598. 2012. View Article : Google Scholar : PubMed/NCBI

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. View Article : Google Scholar

16 

Kurosaki T: Regulation of BCR signaling. Mol Immunol. 48:1287–1291. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Huntington ND, Xu Y, Puthalakath H, Light A, Willis SN, Strasser A and Tarlinton DM: CD45 links the B cell receptor with cell survival and is required for the persistence of germinal centers. Nat Immunol. 7:190–198. 2006. View Article : Google Scholar

18 

Zikherman J, Doan K, Parameswaran R, Raschke W and Weiss A: Quantitative differences in CD45 expression unmask functions for CD45 in B-cell development, tolerance, and survival. Proc Natl Acad Sci USA. 109:E3–E12. 2012. View Article : Google Scholar :

19 

Ohtsuka M, Arase H, Takeuchi A, Yamasaki S, Shiina R, Suenaga T, Sakurai D, Yokosuka T, Arase N, Iwashima M, et al: NFAM1, an immunoreceptor tyrosine-based activation motif-bearing molecule that regulates B cell development and signaling. Proc Natl Acad Sci USA. 101:8126–8131. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Lv W, Duan Q, Wang L, Gong Z, Yang F and Song Y: Expression of B-cell-associated genes in peripheral blood mononuclear cells of patients with symptomatic pulmonary embolism. Mol Med Rep. 11:2299–2305. 2015.

21 

Scapini P, Pereira S, Zhang H and Lowell CA: Multiple roles of Lyn kinase in myeloid cell signaling and function. Immunol Rev. 228:23–40. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Li FJ, Schreeder DM, Li R, Wu J and Davis RS: FCRL3 promotes TLR9-induced B-cell activation and suppresses plasma cell differentiation. Eur J Immunol. 43:2980–2992. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Stepanek O, Draber P, Drobek A, Horejsi V and Brdicka T: Nonredundant roles of Src-family kinases and Syk in the initiation of B-cell antigen receptor signaling. J Immunol. 190:1807–1818. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Barrington RA, Schneider TJ, Pitcher LA, Mempel TR, Ma M, Barteneva NS and Carroll MC: Uncoupling CD21 and CD19 of the B-cell coreceptor. Proc Natl Acad Sci USA. 106:14490–14495. 2009. View Article : Google Scholar : PubMed/NCBI

25 

van Zelm MC, Smet J, Adams B, Mascart F, Schandené L, Janssen F, Ferster A, Kuo CC, Levy S, van Dongen JJ and van der Burg M: CD81 gene defect in humans disrupts CD19 complex formation and leads to antibody deficiency. J Clin Invest. 120:1265–1274. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Barua D, Hlavacek WS and Lipniacki TA: Computational model for early events in B cell antigen receptor signaling: Analysis of the roles of Lyn and Fyn. J Immunol. 189:646–658. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Texido G, Su IH, Mecklenbräuker I, Saijo K, Malek SN, Desiderio S, Rajewsky K and Tarakhovsky A: The B-cell-specific Src-family kinase Blk is dispensable for B-cell development and activation. Mol Cell Biol. 20:1227–1233. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Pospisil R, Silverman GJ, Marti GE, Aruffo A, Bowen MA and Mage RG: CD5 is A potential selecting ligand for B-cell surface immunoglobulin: A possible role in maintenance and selective expansion of normal and malignant B cells. Leuk Lymphoma. 36:353–365. 2000. View Article : Google Scholar : PubMed/NCBI

29 

de Andres B, Mueller AL, Verbeek S, Sandor M and Lynch RG: A regulatory role for Fcgamma receptors CD16 and CD32 in the development of murine B cells. Blood. 92:2823–2829. 1998.PubMed/NCBI

30 

Horejs-Hoeck J, Hren A, Mudde GC and Woisetschläger M: Inhibition of immunoglobulin E synthesis through Fc gammaRII (CD32) by a mechanism independent of B-cell receptor co-cross-linking. Immunology. 115:407–415. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Tedla N, An H, Borges L, Vollmer-Conna U, Bryant K, Geczy C and McNeil HP: Expression of activating and inhibitory leukocyte immunoglobulin-like receptors in rheumatoid synovium: Correlations to disease activity. Tissue Antigens. 77:305–316. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Fanger NA, Borges L and Cosman D: The leukocyte immunoglobulin-like receptors (LIRs): A new family of immune regulators. J Leukoc Biol. 66:231–236. 1999.PubMed/NCBI

33 

Hanten JA, Vasilakos JP, Riter CL, Neys L, Lipson KE, Alkan SS and Birmachu W: Comparison of human B cell activation by TLR7 and TLR9 agonists. BMC Immunol. 9:392008. View Article : Google Scholar : PubMed/NCBI

34 

Yamazaki K, Yamazaki T, Taki S, Miyake K, Hayashi T, Ochs HD and Agematsu K: Potentiation of TLR9 responses for human naïve B-cell growth through RP105 signaling. Clin Immunol. 135:125–136. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Hilgendorf I, Theurl I, Gerhardt LM, Robbins CS, Weber GF, Gonen A, Iwamoto Y, Degousee N, Holderried TA, Winter C, et al: Innate response activator B cells aggravate atherosclerosis by stimulating T helper-1 adaptive immunity. Circulation. 129:1677–1687. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Kwakkenbos MJ, Pouwels W, Matmati M, Stacey M, Lin HH, Gordon S, van Lier RA and Hamann J: Expression of the largest CD97 and EMR2 isoforms on leukocytes facilitates a specific interaction with chondroitin sulfate on B cells. J Leukoc Biol. 77:112–119. 2005.

37 

De Salort J, Sintes J, Llinàs L, Matesanz-Isabel J and Engel P: Expression of SLAM (CD150) cell-surface receptors on human B-cell subsets: From pro-B to plasma cells. Immunol Lett. 134:129–136. 2011. View Article : Google Scholar

38 

Reiter R and Pfeffer K: Impaired germinal centre formation and humoral immune response in the absence of CD28 and interleukin-4. Immunology. 106:222–228. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Galindo-Albarrán AO, Ramírez-Pliego O, Labastida-Conde RG, Melchy-Pérez EI, Liquitaya-Montiel A, Esquivel-Guadarrama FR, Rosas-Salgado G, Rosenstein Y and Santana MA: CD43 signals prepare human T cells to receive cytokine differentiation signals. J Cell Physiol. 229:172–180. 2014. View Article : Google Scholar

40 

Chen J, Wang F, Cai Q, Shen S, Chen Y, Hao C and Sun J: A novel anti-human ICOSL monoclonal antibody that enhances IgG production of B cells. Monoclon Antib Immunodiagn Immunother. 32:125–131. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Greenwald RJ, Freeman GJ and Sharpe AH: The B7 family revisited. Annu Rev Immunol. 23:515–548. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Mak TW, Shahinian A, Yoshinaga SK, Wakeham A, Boucher LM, Pintilie M, Duncan G, Gajewska BU, Gronski M, Eriksson U, et al: Costimulation through the inducible costimulator ligand is essential for both T helper and B cell functions in T cell-dependent B cell responses. Nat Immunol. 4:765–772. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Nadeau PJ, Roy A, Gervais-St-Amour C, Marcotte MÈ, Dussault N and Néron S: Modulation of CD40-activated B lymphocytes by N-acetylcysteine involves decreased phosphorylation of STAT3. Mol Immunol. 49:582–592. 2012. View Article : Google Scholar

44 

Uchida J, Lee Y, Hasegawa M, Liang Y, Bradney A, Oliver JA, Bowen K, Steeber DA, Haas KM, Poe JC and Tedder TF: Mouse CD20 expression and function. Int Immunol. 16:119–129. 2004. View Article : Google Scholar

45 

Józsi M, Prechl J, Bajtay Z and Erdei A: Complement receptor type 1 (CD35) mediates inhibitory signals in human B lymphocytes. J Immunol. 168:2782–2788. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Young NT, Waller EC, Patel R, Roghanian A, Austyn JM and Trowsdale J: The inhibitory receptor LILRB1 modulates the differentiation and regulatory potential of human dendritic cells. Blood. 111:3090–3096. 2008. View Article : Google Scholar

47 

HoWangYin KY, Loustau M, Wu J, Alegre E, Daouya M, Caumartin J, Sousa S, Horuzsko A, Carosella ED and LeMaoult J: Multimeric structures of HLA-G isoforms function through differential binding to LILRB receptors. Cell Mol Life Sci. 69:4041–4049. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Fujikawa K, Miletic AV, Alt FW, Faccio R, Brown T, Hoog J, Fredericks J, Nishi S, Mildiner S, Moores SL, et al: Vav1/2/3-null mice define an essential role for Vav family proteins in lymphocyte development and activation but a differential requirement in MAPK signaling in T and B cells. J Exp Med. 198:1595–1608. 2003. View Article : Google Scholar : PubMed/NCBI

49 

Lee JK, Mathew SO, Vaidya SV, Kumaresan PR and Mathew PA: CS1 (CRACC, CD319) induces proliferation and autocrine cytokine expression on human B lymphocytes. J Immunol. 179:4672–4678. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Kim JR, Mathew SO, Patel RK, Pertusi RM and Mathew PA: Altered expression of signalling lymphocyte activation molecule (SLAM) family receptors CS1 (CD319) and 2B4 (CD244) in patients with systemic lupus erythematosus. Clin Exp Immunol. 160:348–358. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Hsi ED, Steinle R, Balasa B, Szmania S, Draksharapu A, Shum BP, Huseni M, Powers D, Nanisetti A, Zhang Y, et al: CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clin Cancer Res. 14:2775–2784. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 13 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yan W, Song H, Jiang J, Xu W, Gong Z, Duan Q, Li C, Xie Y and Wang L: Characteristics of B cell‑associated gene expression in patients with coronary artery disease. Mol Med Rep 13: 4113-4121, 2016
APA
Yan, W., Song, H., Jiang, J., Xu, W., Gong, Z., Duan, Q. ... Wang, L. (2016). Characteristics of B cell‑associated gene expression in patients with coronary artery disease. Molecular Medicine Reports, 13, 4113-4121. https://doi.org/10.3892/mmr.2016.5029
MLA
Yan, W., Song, H., Jiang, J., Xu, W., Gong, Z., Duan, Q., Li, C., Xie, Y., Wang, L."Characteristics of B cell‑associated gene expression in patients with coronary artery disease". Molecular Medicine Reports 13.5 (2016): 4113-4121.
Chicago
Yan, W., Song, H., Jiang, J., Xu, W., Gong, Z., Duan, Q., Li, C., Xie, Y., Wang, L."Characteristics of B cell‑associated gene expression in patients with coronary artery disease". Molecular Medicine Reports 13, no. 5 (2016): 4113-4121. https://doi.org/10.3892/mmr.2016.5029