Skip to main content
Erschienen in: BMC Cancer 1/2017

Open Access 01.12.2017 | Research article

Combination of Helicobacter pylori infection and the interleukin 8 –251 T > A polymorphism, but not the mannose-binding lectin 2 codon 54 G > A polymorphism, might be a risk factor of gastric cancer

verfasst von: Young Woon Chang, Chi Hyuk Oh, Jung-Wook Kim, Jae Won Lee, Mi Ju Park, Jae-Jun Shim, Chang Kyun Lee, Jae-Young Jang, Seok Ho Dong, Hyo Jong Kim, Sung Soo Kim, Byung-Ho Kim

Erschienen in: BMC Cancer | Ausgabe 1/2017

Abstract

Background

Mannose-binding lectin (MBL) acts in the innate immune response to Helicobacter pylori. Interleukin 8 (IL-8) is a potent cytokine produced by gastric epithelial cells in response to H. pylori. We aimed to investigate whether polymorphisms in MBL2 and IL-8 influence susceptibility to H. pylori infection, and the associations of these polymorphisms with the risk of gastroduodenal diseases in a Korean population.

Methods

We consecutively enrolled 176 H. pylori-negative control subjects, 221 subjects with H. pylori-positive non-atrophic gastritis, 52 mild atrophic gastritis (AG), 61 severe AG, 175 duodenal ulcer, and 283 gastric cancer (GC). Allele-specific PCR-RFLP was conducted for polymorphisms in MBL2 exon 1 (codon 52, 54, and 57) and IL-8 -251 T > A. IL-8 levels in gastric mucosal tissues and serum MBL levels were measured by enzyme-linked immunosorbent assay.

Results

MBL2 exon 1 polymorphic variants were found only in codon 54, and the allele frequencies did not differ significantly between the control and disease groups. Although serum MBL levels in codon 54 A/A mutants were markedly low, it did not influence susceptibility to H. pylori infection or the risk of gastroduodenal diseases. IL-8 levels were significantly different between T/T wild type, T/A heterozygote, and A/A mutant genotypes. IL-8 -251 A allele carriers (A/A + T/A) showed increased IL-8 levels, and were significantly associated with the risk of severe AG and GC.

Conclusions

We suggest that a combination of H. pylori infection and the IL-8 -251 T > A polymorphism might increase the risk of severe AG and GC in a Korean population.
Abkürzungen
AG
Atrophic gastritis
CI
Confidence intervals
DU
Duodenal ulcer
ELISA
Enzyme-linked immunosorbent assay
GC
Gastric cancer
Helicobacter pylori
H. pylori
Hp(−)
Helicobacter pylori-negative
Hp(+)
Helicobacter pylori-positive
IL-1β
Interleukin 1 beta
IL-8
Interleukin 8
MBL
Mannose-binding lectin
NAG
Non-atrophic gastritis
OR
Odds ratios
PCR
Polymerase chain reaction
RFLP
Restriction fragment length polymorphism
SNP
Single nucleotide polymorphism
TLR
Toll-like receptor

Background

The Helicobacter pylori infection rate is about 50% among the worldwide adult population [1]. In Korea, the adult H. pylori infection rate was 66.9% in 1998, 59.6% in 2005, and dropped to 54.4% in 2011 [2]. The main cause of this decrease in infection rate is the improvement in unsanitary environmental conditions. Besides environmental factors, bacterial and host factors are involved in the pathogenesis of H. pylori infection. With regard to bacterial factors, H. pylori strains possessing the virulence factors cagA, vacA s1a/m1, and iceA1 are known to be particularly virulent, and are frequently associated with severe gastric epithelial damage [3]. In contrast to Western populations, the cagA protein is commonly found in Korean patients with gastric cancer (GC) and duodenal ulcer (DU) [4]. However, there have been no associations reported between different H. pylori genotypes and clinical outcome in Korean patients [5, 6].
With regard to host factors, host genetic variants may influence susceptibility to H. pylori and the pathogenesis of gastroduodenal diseases. Host factors are mainly related to two processes: recognition of H. pylori by the innate immune system, and the level of the cytokine response [7, 8]. Polymorphisms in pro- and anti-inflammatory cytokines are associated with the risk of atrophic gastritis (AG) and GC. Interleukin 1 beta (IL-1β), tumor necrosis factor-alpha, IL-6, and IL-8 are up-regulated in response to H. pylori infection [9]. Several anti-inflammatory cytokines such as IL-Rα and IL-10 are also related to H. pylori infection [10, 11].
IL-8 is a major neutrophil-activating cytokine and plays a central role in the immuno-pathogenesis of H. pylori-induced gastric mucosal injury. IL-8 levels are 10-fold higher in GC specimens than in normal gastric tissues [12]. The IL-8 -251 T > A polymorphism has been reported to be associated with increased production of IL-8 protein, and higher risks of AG, gastric ulcer, and GC [1317]. However, many other reports are inconsistent with these findings [1823], and a meta-analysis of epidemiological studies revealed no overall association [24].
The innate immune response to H. pylori infection is a further candidate host factor. Toll-like receptors (TLRs) recognize conserved pathogen-associated molecular patterns expressed by many pathogens, including H. pylori [25]. Mannose-binding lectin (MBL), a pattern recognition receptor encoded by the MBL2 gene, recognizes lipopolysaccharide in the cell wall of gram-negative bacteria such as H. pylori [26, 27]. H. pylori activates MBL in vitro, resulting in complement deposition [28, 29]. Some studies have found a possible association of MBL2 haplotype with susceptibility to H. pylori infection, as well as with risk of GC [30, 31]. However, other studies did not find any significant association between MBL genotype and H. pylori infection prevalence or GC risk [32, 33].
Serum MBL levels vary widely between healthy individuals, mainly due to genetic variation [3436]. The variation in serum MBL levels is correlated with point mutations in the coding and promoter regions of MBL2. Three mutations within exon 1 (in codons 52, 54, and 57) interfere with MBL function and are associated with low serum levels of MBL. In African populations, point mutations at codons 52 and 57 occur frequently [36, 37]. In Caucasians, mutations at codons 52 and 54 are common [38]. In Chinese, Japanese, and Korean populations, mutations are predominantly common in codon 54, but not in codons 52 or 57 [3941]. Polymorphisms within the promoter and 5′-untranslated regions of MBL2 also affect serum levels of MBL, but the effects were found to be lower than those of the exon 1 polymorphisms [41].
The aims of this study were: 1) to examine the influence of the polymorphisms in codons 52, 54, and 57 of MBL2 (related to innate immunity) on susceptibility to H. pylori infection; 2) to evaluate the association of the IL-8 -251 T > A polymorphism with the risk of gastroduodenal diseases in a Korean population; and 3) to analyze our and other investigators’ large-scale data regarding the IL-8 -251 T > A polymorphism and GC risk in Korean, Japanese, Chinese, and Caucasian populations.

Methods

From January 2012 to May 2015, H. pylori-negative healthy control subjects (control, n = 176), H. pylori-positive non-atrophic gastritis patients (NAG, n = 108), H. pylori-positive mild AG patients (n = 52), H. pylori-positive severe AG patients (n = 61), DU patients (n = 175), and GC patients (n = 283) were consecutively enrolled.
All participants (n = 855) underwent upper gastrointestinal endoscopy and routine laboratory tests. The controls were asymptomatic subjects who visited the Health Screening Center for a health status check-up, and their endoscopic findings were normal. Exclusion criteria were H. pylori eradication history; use of antibiotics, proton pump inhibitors, nonsteroidal anti-inflammatory drugs, or anticoagulant drugs; and severe systemic illnesses. Age, sex, alcohol consumption (current or never), smoking habits (current or never), salt intake (high, low-moderate), and family history of GC (first-degree relatives) were recorded. Informed consent was obtained from all included subjects. The Institutional Review Board of the Kyung Hee University Hospital approved the study protocol (KMC IRB 1523–04).

Diagnosis of H. pylori infection

The rapid urease test (or urea breath test) and serum anti-H. pylori immunoglobulin G antibody test were performed. A subject was defined as H. pylori infection-positive if both tests were positive. A subject was defined as H. pylori infection-negative if both tests were negative. Subjects with only one positive test were excluded from this study.

Histologic examination of chronic gastritis

One pathologist histologically evaluated chronic gastritis status in biopsy specimens. AG was graded based on the presence and proportion of glandular loss (mild, moderate, and severe) according to the updated Sydney system [42].

Genotyping of MBL2 exon 1 codons 52, 54 and 57, and of IL-8 -251

Genomic DNA was extracted from peripheral venous blood using a genomic DNA purification method. Polymerase chain reaction (PCR) amplification, restriction fragment length polymorphism (RFLP) analysis, and gel electrophoresis were performed for MBL2 (codons 52, 54, and 57 in exon 1) and IL-8 (−251 promoter region) as described previously [7, 34]. The PCR product involving codon 52 was digested by incubation with MluI at 37 °C for 3 h, resulting in two bands of 204 and 94 bp for the T/T genotype (mutant), three bands of 298, 204, and 94 bp for the A/T genotype (heterozygote), and one band of 298 bp for the A/A genotype (wild type). The PCR product involving codon 54 was digested by BanI at 50 °C for 3 h, resulting in two bands of 195 and 103 bp for the G/G genotype (wild type), three bands of 298, 195, and 103 bp for the G/A genotype (heterozygote), and one band of 298 bp for the A/A genotype (mutant). The PCR product involving codon 57 was digested with MboI at 37 °C for 3 h, resulting in two bands of 190 and 108 bp for the A/A genotype (mutant), three bands of 298, 190, and 108 bp for the G/A genotype (heterozygote), and one band of 298 bp for the G/G genotype (wild type). For genotyping of the IL-8 -251 T > A polymorphism, PCR products were digested with MfeI at 37 °C for 3 h, resulting in two bands of 449 and 92 bp for the A/A genotype (mutant), three bands of 541, 449, and 92 bp for the T/A genotype (heterozygote), and one band of 541 bp for the T/T genotype (wild type).

Measurement of serum MBL levels

MBL is a serum protein produced mainly by hepatocytes, and expressed in immune cells, but not in epithelial cells [43]. Circulatory MBL levels were taken as an indicator of the functional activity of MBL protein. Serum MBL levels were measured after overnight fasting by enzyme-linked immunosorbent assay (ELISA; MBL Oligomer ELISA kit; BioProto Diagnostics, Denmark).

Measurement of IL-8 levels in gastric mucosal tissues

Although measurement of serum IL-8 levels is straightforward, serum IL-8 levels do not reflect the severity of H. pylori-associated gastritis [44]. Therefore, we measured IL-8 levels in gastric mucosal tissues rather than serum IL-8 levels.
Three biopsy specimens were taken from the greater curvature side of the proximal antrum during endoscopic procedures. The specimens were put into a tube with 2.0 mL phosphate-buffered saline (pH 7.4), frozen on dry ice, and stored at −70 °C. Samples were homogenized and centrifuged, and the supernatants were aliquoted. Total protein was measured using the bicinchoninic acid assay (Thermo Scientific, Rockford, IL, USA). Gastric mucosal IL-8 levels were measured by ELISA (R&D Systems Inc., Minneapolis, MN, USA). The mucosal level of IL-8 was expressed as picograms per milligram of gastric biopsy protein.

Analysis of global raw data regarding IL-8 -251 T > A polymorphism and GC risk

The results obtained regarding the association of GC risk with IL-8 -251 T > A genotype was not consistent with previous epidemiological results [1824]. Therefore, we collected large-scale raw data of GC patients (n = 3217) and controls (n = 3810) from Asian (Korea, Japan, and China), and Caucasian (Poland, Finland, and Portugal) populations [1323], and analyzed GC risk according to IL-8 -251 T > A genotype.

Statistical analysis

Data are expressed as mean values ± standard deviations or as frequencies and percentages. Chi-squared and Kruskal–Wallis tests were performed to compare clinical parameters between the control and disease groups. Hardy–Weinberg equilibrium for polymorphisms in MBL2 and IL-8 was tested using R version 3.1.0 (R Development Core Team). Biases caused by differences in clinical parameters were adjusted using the chi-squared and Kruskal–Wallis tests. Multiple logistic regression analysis was performed to evaluate the associations of the genetic polymorphisms with susceptibility to H. pylori infection and the risk of gastroduodenal diseases using the SAS statistical software package version 9.4 (SAS Institute Inc.). All clinical parameters with a p value <0.20 in the univariate analysis were included in the full logistic regression model. The odds ratios (ORs) and their 95% confidence intervals (CIs) were used to compare the risks between the control and disease groups. P values <0.05 were considered statistically significant.

Results

Table 1 shows clinical features of the control and disease groups. Age, sex ratio, and alcohol consumption were similar among all groups. Risk factors for GC, such as smoking, high salt intake, and family history of GC, were more frequently observed in the H. pylori-positive GC group than in the control group. The differences were statistically significant.
Table 1
Basic clinical features of the control and disease groups
 
Hp (−)
Control
(n = 176)
Hp (+)
NAG
(n = 108)
Hp (+)
Mild AG
(n = 52)
Hp (+)
Severe AG
(n = 61)
DU
(n = 175)
GC
(n = 283)
p
Sex (M:F)
87:89
41:67
23:29
37:24
90:85
151:132
0.1832a
Mean age (years)
52.5 ± 6.7
51.58 ± 6.65
59.52 ± 8.30
60.89 ± 8.55
53.2 ± 12.6
55.5 ± 8.8
0.1431b
Alcohol consumption (%)
52.27%
51.85%
48.46%
47.54%
57.71%
55.48%
0.1751a
Smoking habits (%)
19.89%
18.52%
30.77%
26.23%
58.86%
40.99%
<0.0001a
High salt intake (%)
25.00%
26.85%
16.92%
49.18
39.43%
48.41%
<0.0001a
FHx of GC (%)
10.23%
15.52%
13.79%
16.39%
10.86%
20.85%
0.0043a
Hp positivity (%)
0%
100%
100%
100%
96.00%
73.14%
<0.0001a
a: Chi square test for comparison of percentage (GC vs control)
b: Kruskal-Wallis test for comparison of mean ± SD (GC vs control)
The frequencies of the MBL2 codon 54 and IL-8 -251 polymorphisms in the control group did not deviate significantly from those expected under Hardy–Weinberg equilibrium (p = 1.000 and p = 0.184, respectively). In this study population, MBL exon 1 polymorphic variants were found only in codon 54. There were no variants at codons 52 or 57; only the wild type was observed.

The frequencies of MBL2 codon 54 and IL-8 -251 genotypes in the control and disease groups

The frequencies of MBL2 codon 54 genotypes were similar among the control and disease groups (Table 2). The frequency of IL-8 -251 A allele carriers was higher in the H. pylori-positive severe AG and H. pylori-positive GC groups than in the control group, but the differences did not reach statistical significance (Table 2).
Table 2
The frequencies of MBL2 codon 54 and IL-8 -251 genotypes in the control and disease groups
Genotype
Control
n = 176 (%)
Hp (+)
NAG
n = 108 (%)
Hp (+)
Mild AG
n = 52 (%)
Hp (+)
Severe AG
n = 61 (%)
Hp (−)
DU
n = 7 (%)
Hp (+)
DU
n = 168 (%)
Hp (−)
GC
n = 76 (%)
Hp (+)
GC
n = 207 (%)
MBL2 codon 54
G/G (wild type)
103 (58.52)
65 (60.19)
29 (55.77)
32 (52.46)
4 (57.14)
95 (56.55)
41 (53.95)
129 (62.32)
G/A(heterozygote)
63 (35.80)
37 (34.26)
22 (43.31)
27 (44.26)
3 (42.86)
69 (41.07)
29 (32.37)
67 (32.37)
A/A (mutant)
10 (5.68)
6 (5.56)
1 (1.92)
2 (3.28)
0 (0.00)
4 (2.38)
6 (5.31)
11 (5.31)
G/A + A/A (A carrier)
73 (41.48)
43 (39.81)
23 (44.23)
29 (47.54)
4 (57.14)
73 (43.45)
35 (46.05)
78 (37.68)
IL-8 -251
T/T (wild type)
70 (39.77)
52 (48.15)
20 (38.46)
15 (24.59)
3 (42.86)
81 (48.21)
23 (30.26)
58 (28.02)
T/A (heterozygote)
89 (50.57)
49 (45.37)
27 (51.92)
37 (60.66)
3 (42.86)
73 (43.45)
46 (60.53)
122 (58.94)
A/A (mutant)
17 (9.66)
7 (6,48)
5 (9.62)
9 (16.75)
1 (14.29)
14 (8.33)
7 (9.21)
27 (13.04)
T/A + A/A (A carrier)
106 (60.23)
56 (51.85)
32 (61.54)
46 (75.41)
4 (57.14)
87 (51.79)
53 (69.74)
149 (71.99)
The frequencies of MBL2 codon 54 and IL-8 -251 genotypes were not significantly different between the control and disease groups

Association between MBL2 codon 54 G > A polymorphism and the risk of gastroduodenal diseases

We examined the association between the MBL2 codon 54 G > A polymorphism and the risk of gastroduodenal disease using univariate and multivariate logistic regression analysis. We regarded the control group as the reference subject group, and considered G/G (wild type) as the reference genotype. The MBL2 codon 54 G > A polymorphism did not increase susceptibility to H. pylori-positive NAG, mild AG, or severe AG, and also was not associated with the risk of DU and GC (Table 3).
Table 3
MBL codon 54 G > A polymorphism and the risk of gastroduodenal diseases
Genotype
Disease group
Unadjusted OR (95% CI) by univariate analysis
p
Adjusted OR (95% CI) by multivariate analysis
p
G/A (heterozygote)
Hp(+) NAG (n = 49)
0.93 (0.56–1.55)
0.7828
1.02 (0.59–1.75)
0.9466
Hp(+) mild AG (n = 27)
1.24 (0.66–2.34)
0.5074
1.16 (0.60–2.26)
0.6527
Hp(+) severe AG (n = 37)
1.38 (0.76–2.52)
0.2937
1.16 (0.61–2.18)
0.6498
Hp(−) DU(n = 3)
1.23 (0.27–5.66)
0.7938
1.21 (0.26–5.71)
0.8082
Hp(+) DU (n = 73)
1.19 (0.76–1.85)
0.4450
1.24 (0.78–1.99)
0.3605
Hp(−) GC(n = 46)
1.16 (0.65–2.04)
0.6170
1.11 (0.61–2.00)
0.7356
Hp(+) GC (n = 122)
0.85 (0.55–1.31)
0.4566
0.85 (0.54–1.34)
0.7356
A/A (mutant)
Hp(+) NAG (n = 7)
0.95 (0.33–2.74)
0.9255
1.19 (0.39–3.61)
0.7561
Hp(+) mild AG (n = 5)
0.36 (0.04–2.89)
0.3332
0.33 (0.04–2.81)
0.3126
Hp(+) severe AG (n = 9)
0.64 (0.13–3.09)
0.5822
0.61 (0.12–3.12)
0.5560
Hp(−) DU (n = 1)
1.25(0.32–5.88)
0.9521
1.24(0.31–5.77)
0.9434
Hp(+) DU (n = 14)
0.43 (0.13–1.43)
0.1697
0.45 (0.13–1.55)
0.2031
Hp(−) GC (n = 7)
1.51 (0.52–4.42)
0.4543
1.93 (0.63–5.89)
0.2484
Hp(+) GC (n = 27)
0.88 (0.36–2.15)
0.7762
1.10 (0.43–2.79)
0.8496
G/A + A/A (A carrier)
Hp(+) NAG (n = 56)
0.93 (0.57–1.52)
0.7820
1.04 (0.62–1.75)
0.8774
Hp(+) mild AG (n = 32)
1.12 (0.60–2.09)
0.7239
1.05 (0.55–2.00)
0.8893
Hp(+) severe AG (n = 46)
1.28 (0.71–2.29)
0.4103
1.09 (0.59–2.01)
0.7929
Hp(−) DU (n = 4)
1.06 (0.23–4.87)
0.9421
1.06 (0.23–4.98)
0.9386
Hp(+) DU (n = 87)
1.08 (0.71–1.66)
0.7110
1.13 (0.72–1.78)
0.5868
Hp(−) GC (n = 53)
1.20 (0.70–2.07)
0.5008
1.19 (0.68–2.09)
0.5480
Hp(+) GC (n = 149)
0.85 (0.57–1.29)
0.4488
0.88 (0.57–1.35)
0.5528
The control was regarded as the reference subject group, and the G/G wild type was considered as the reference genotype

Serum MBL levels

Serum levels of MBL were high in carriers of the G/G (wild type) genotype, intermediate in those with the G/A heterozygous genotype, and low in those with the A/A (mutant) genotype in all subjects (n = 855, Fig. 1). The differences between the three genotypes were highly statistically significant (p < 0.0001). However, there were no significant differences in serum MBL levels between the control (139.9 ± 83.2 ng/mL), H. pylori-positive NAG (149.3 ± 81.2 ng/mL), mild AG (146.9 ± 81.8 ng/mL), severe AG (140.2 ± 87.3 ng/mL), DU (143.8 ± 82.5 ng/mL), and GC (149.8 ± 82.6 ng/mL) groups.

Association between IL-8 -251 T > A polymorphism and the risk of disease development

Because the IL-8 cytokine response is mainly dependent on the H. pylori-associated inflammatory severity, we sub-classified the H. pylori-positive chronic gastritis group (n = 221) into H. pylori-positive NAG, mild AG, and severe AG. We regarded the control group as the reference subject group, and considered T/T (wild type) as the reference genotype. The IL-8 -251 A allele significantly increased the risk of severe AG and GC, as determined by both univariate and multivariate logistic regression analysis (Table 4).
Table 4
IL-8 -251 T > A polymorphism and the risk of gastroduodenal diseases
Genotype
Disease group
Unadjusted OR (95% CI) by univariate analysis
p
Adjusted OR (95% CI) by multivariate analysis
p
T/A (heterozygote)
Hp(+) NAG (n = 49)
0.74 (0.45–1.22)
0.2406
0.76 (0.46–1.29)
0.3018
Hp(+) mild AG (n = 27)
1.06 (0.55–2.05)
0.8581
1.09 (0.55–2.16)
0.8052
Hp(+) severe AG (n = 37)
1.94 (0.99–3.82)
0.0549
2.06 (1.01–4.21)
0.0471
Hp(−) DU(n = 3)
0.79 (0.15–4.02)
0.7729
0.84 (0.16–4.36)
0.8342
Hp(+) DU (n = 73)
0.71 (0.45–1.11)
0.1297
0.80 (0.50–1.29)
0.3616
Hp(−) GC(n = 46)
1.57 (0.87–2.84)
0.1326
1.66 (0.89–3.07)
0.1058
Hp(+) GC (n = 122)
1.65 (1.06–2.58)
0.0257
1.71 (1.07–2.72)
0.0252
A/A (mutant)
Hp(+) NAG (n = 7)
0.55 (0.21–1.43)
0.2237
0.59 (0.21–1.59)
0.2952
Hp(+) mild AG (n = 5)
1.03 (0.34–3.14)
0.9593
1.00 (0.31–3.17)
0.9941
Hp(+) severe AG (n = 9)
2.47 (0.93–6.59)
0.0710
2.42 (0.85–6.84)
0.0966
Hp(−) DU (n = 1)
1.37 (0.13–14.03)
0.7895
1.66 (0.16–17.71)
0.6743
Hp(+) DU (n = 14)
0.71 (0.33–1.55)
0.3905
0.72 (0.32–1.63)
0.4289
Hp(−) GC (n = 7)
1.25 (0.46–3.40)
0.6577
1.19 (0.43–3.34)
0.7403
Hp(+) GC (n = 27)
1.92 (0.95–3.86)
0.0683
1.94 (0.93–4.06)
0.0744
T/A + A/A
(A carrier)
Hp(+) NAG (n = 56)
0.71 (0.44–1.15)
0.1669
0.73 (0.44–1.22)
0.2282
Hp(+) mild AG (n = 32)
1.06 (0.56–1.99)
0.8651
1.07 (0.55–2.08)
0.8336
Hp(+) severe AG (n = 46)
2.03 (1.05–3.90)
0.0351
2.12 (1.06–4.25)
0.0335
Hp(−) DU (n = 4)
0.88 (0.19–4.05)
0.8703
0.96 (0.20–4.48)
0.3066
Hp(+) DU (n = 87)
0.71 (0.46–1.09)
0.1152
0.79 (0.50–1.24)
0.3035
Hp(−) GC (n = 53)
1.52 (0.86–2.70)
0.1524
1.58 (0.87–2.87)
0.1346
Hp(+) GC (n = 149)
1.70 (1.11–2.60)
0.0155
1.74 (1.11–2.74)
0.0161
The control was regarded as the reference subject group, and the T/T wild type was considered as the reference genotype

Comparison of IL-8 levels according to disease phenotypes and IL-8 -251 each genotype

IL-8 levels were low in subjects with the T/T (wild type) genotype, intermediate in those with the T/A heterozygous genotype, and high in those with the A/A (mutant) genotype. The differences between the three genotypes were statistically significant (p = 0.0262, Fig. 2).
IL-8 levels were markedly low in H. pylori-negative control (n = 176, 4.43 ± 3.30 pg/mg protein) and H. pylori-negative subjects regardless of any disease phenotypes (n = 83, 5.02 ± 3.80 pg/mg protein) compared with H. pylori-positive subjects (n = 596, 154.05 ± 56.26 pg/mg protein; p < 0.0001). IL-8 levels were very low in the H. pylori-negative DU and H. pylori-negative GC groups as well as in the control group. Therefore, we regarded the H. pylori-positive NAG group as the reference subject group instead of the control group. We selected five H. pylori-positive disease groups to evaluate the gastric precancerous cascade: NAG, mild AG, severe AG, DU, and GC (Fig. 3). The five disease groups did not show any significant differences in IL-8 levels for the T/T (p = 0.7979), T/A (p = 0.2200), or A/A (p = 0.1000) genotypes, or A allele carriers (p = 0.0550), as analyzed by a multiple group comparison test. However, H. pylori-positive GC A allele carriers (n = 207, 172.3 ± 65.4 pg/mg protein) showed significantly higher IL-8 levels than NAG A allele carriers (n = 108, 148.3 ± 42.9 pg/mg protein, p = 0.0229), as determined by the two-group comparison test (Fig. 3).

Analysis of global results of IL-8 -251 T > A polymorphism and GC risk

The Korean population, including the subjects of this study, showed a significant positive association between the IL-8 -251 T > A polymorphism and GC risk. On the contrary, the Chinese and Caucasian populations showed a negative association. The Japanese population was similar to the Korean population. The combined Korean and Japanese populations showed significantly increased GC risk for the IL-8 -251 T/A and A/A genotypes compared to the T/T genotype, and for A allele carriers compared to non-carriers (Table 5).
Table 5
The association of IL-8 -251 T > A polymorphism with the risk of GC in different ethnicities
Ethnicity
TT / total GC (%)
TT / total controls (%)
OR [95% CI]
TA / total GC (%)
TA / total controls (%)
OR [95% CI]
AA / total GC (%)
AA / total controls (%)
OR [95% CI]
A* / total GC (%)
A* / total controls (%)
OR [95% CI]
This study
(Korean)
81/283 (28.62%)
70/176 (39.77%)
0.60 [0.40, 0.90]
168/283(59.36%)
89/176 (50.57%)
1.43 [0.98, 2.09]
34/283(12.01%)
17/176(9.66%)
1.27 [0.70, 2.31]
202/283(71.38%)
106/176 (60.23%)
1.66 [1.11, 2.47]
Korean
[16, 17]
180/487(36.96%)
244/528(46.21%)
0.68 [0.53, 0.88]
241/487(49.49%)
234/528(44.32%)
1.23 [0.96, 1.57]
66/487(13.55%)
50/528(9.47%)
1.50 [1.02, 2.20]
307/487(63.04%)
284/528 (53.79%)
1.46 [1.14, 1.88]
Japanese
[1315]
337/789(42.71%)
485/964(50.31%)
0.74 [0.61, 0.89]
375/789(47.53%)
397/964(41.18%)
1.29 [1.07, 1.56]
77/789(9.76%)
82/964(8.51%)
1.16 [0.84, 1.62]
452/789(57.29%)
479/964(49.69%)
1.36 [1.12, 1.64]
Chinese
[1820]
329/926(35.53%)
270/814(33.17%)
1.11 [0.91, 1.35]
425/926(45.90%)
406/814(49.88%)
0.85 [0.71, 1.03]
172/926(18.57%)
138/814(16.95%)
1.12 [0.87, 1.43]
597/926(64.47%)
544/814(66.83%)
0.90 [0.74, 1.10]
Caucasian
[2123]
224/732(30.60%)
381/1328(28.69%)
1.10 [0.90, 1.34]
365/732(49.86%)
669/1328(50.38%)
0.98 [0.82, 1.17]
143/732(19.54%)
278/1328(20.93%)
0.92 [0.73, 1.15]
508/732(69.40%)
947/1328(71.31%)
0.91 [0.75, 1.11]
Combined
Korean and Japanese
598/1559(38.36%)
799/1668(47.90%)
0.70 [0.61, 0.81]
784/1559(50.29%)
720/1668(43.17%)
1.29 [1.12, 1.48]
177/1559(11.35%)
149/1668(8.93%)
1.29 [1.02, 1.62]
961/1559(61.64%)
869/1668(52.10%)
1.42 [1.24, 1.64]
A*: A allele carriers (T/A + A/A)
The combined Korean and Japanese populations showed significant GC risk in IL-8 -251 T/A, A/A, and A allele carriers

Discussion

The innate immune response is the first line of defense against H. pylori infection in the human stomach. TLR and MBL are recognized as important proteins in innate immunity. Several studies have demonstrated that TLR4 and TLR2 polymorphisms are associated with the risk of GC [4547]. However, some of the associations are controversial, and there are discrepancies between the results for Asian and Western populations [48]. A recent study in the Netherlands found that only the TLR1 polymorphism is associated with the prevalence of H. pylori seropositivity [49]. Further studies are needed in other populations worldwide to confirm these associations.
MBL binds to bacteria, yeasts, and viruses via specific repeated oligosaccharide moieties on the cell surface. MBL activates the complement-lectin pathway, facilitates opsonization and phagocytosis, and induces direct cellular lysis. MBL deficiency or a low serum MBL level has been associated with several infectious and autoimmune diseases, including meningococcal meningitis, pneumonia, arterial thrombosis, systemic lupus erythematosus, and celiac disease [50, 51].
At the time of its discovery, H. pylori was considered an extracellular bacterium that mainly colonized the gastric mucus layer or attached to gastric epithelial cells. However, it has since been demonstrated that H. pylori invades the lamina propria and gastric epithelial cells [52]. Therefore, H. pylori might be a target of phagocytosis by MBL activation. There have been few clinical studies regarding the role of MBL in H. pylori infection. Various microorganisms such as H. pylori, Neisseria meningitidis groups B and C, Nocardia farcinica, and Legionella pneumophila induce MBL activity in vitro [28]. Activated complements are found in the epithelium of patients with H. pylori-associated gastritis [29]. One pediatric study reported that MBL2 mRNA expression in gastric biopsy specimens was higher in H. pylori-positive chronic gastritis than in H. pylori-negative chronic gastritis patients [53]. However, the study had two weaknesses in terms of its ability to reach conclusions regarding the role of MBL2 expression in the development of H. pylori-infected chronic gastritis. The first weakness is the small number of biopsy specimens that were obtained, with only five H. pylori-positive children and four control children included. The second weakness is that they could not find any association between MBL2 genotype and the risk of H. pylori-infected chronic gastritis.
The association between the MBL2 haplotype and the risk of GC has been studied previously [30, 31]. A study conducted in Southern Italy found that the HYP + D haplotype (H/Y promoter region mutation + P untranslated region mutation + codon 52 mutation) may be a genetic marker for H. pylori-positive GC risk [30]. Another study performed in Warsaw, Poland found that the HY + D haplotype (H/Y promoter region mutation + codon 52 mutation) was related to an increased risk of GC compared with the HY+ A haplotype (H/Y mutation + codon 52 wild type) [31]. Therefore, the codon 52 D variant (cysteine > arginine) was specifically related to the risk of GC in two populations. In contrast to the above studies, which reported positive associations, Australian researchers evaluated healthy individuals for H. pylori infection, MBL2 genotype, mannan binding level, and complement 4 level in plasma, and found that MBL deficiency, defined by either genotype or plasma activity, was not associated with higher susceptibility to H. pylori infection [33]. In a Japanese study, they could no significant differences were found in MBL2 genotypes between GC patients and healthy controls [32]. Instead, the investigators found that the MBL2 codon 54 polymorphism was weakly associated with severe AG and advanced GC [32, 54]. In the present study, we first demonstrated that the codon 54 polymorphism did not increase susceptibility to H. pylori infection in a Korean population. Secondly, we did not find any evidence of a role for MBL2 in the development of gastroduodenal diseases. Thirdly, we did not find a higher risk of advanced GC or severe AG compared to early GC or mild AG, respectively, associated with MBL2 genotype.
With regard to interracial differences, the Korean population differs from the European (Italian and Polish) and African populations reported previously. However, the results for the Korean population are very similar to those reported for the Chinese and Japanese populations [35, 3840]. The frequencies of point mutations in European populations are in between those of the East Asian and African populations.
In the present study, serum levels of MBL, an indicator of the functional activity of MBL, differed significantly according to the genotype. However, serum MBL levels were not significantly different between the control and disease groups, because the frequency of each genotype was similar in these groups.
H. pylori infection stimulates IL-8 gene expression and increases the IL-8 cytokine level in gastric epithelial cells. A significant correlation between a high level of IL-8 in the gastric mucosa and the risk of GC has been reported [13]. Our previous study found that the IL-8 level in gastric mucosal tissues was significantly higher in H. pylori-infected subjects compared with that in H. pylori non-infected subjects, irrespective of their gastroduodenal disease phenotype. After H. pylori eradication, the IL-8 level decreased dramatically, to the same level observed in non-infected subjects [55]. In this study, we confirmed once again that the IL-8 level in gastric mucosal tissues is mainly dependent on H. pylori-positive status.
It has been reported that the IL-8 -251 T > A polymorphism is related to higher levels of IL-8 and to an increased risk of AG, gastric ulcer, and GC [13, 14]. In this study, we also demonstrated that the IL-8 -251 T > A polymorphism increased IL-8 production, and was significantly associated with the risk of GC and severe AG. However, many other epidemiological studies have reported negative associations between the IL-8 -251 polymorphism and GC risk (18–23), and a meta-analysis revealed no overall association (24). In this study, we analyzed large-scale raw data from controls and GC patients from Korean, Japanese, Chinese, and Caucasian (Poland, Finland, and Portugal) populations (13–23). Korean results, including ours, were consistent with Japanese results, but not with Chinese or Caucasian results. The concordance between the Korean and Japanese results might be explained by genetic similarities. In a large study of single nucleotide polymorphism (SNP) maps covering the human genome performed in African Americans, Asians (Japanese-Chinese-Korean), and European Americans (Caucasians) [56], SNP differences in autosomes were only 5.86% between Korean and Japanese populations. Therefore, the Korean population is very similar to the Japanese population with respect to the pattern of SNPs [56].

Conclusions

The MBL2 codon 54 G > A polymorphism does not influence susceptibility to H. pylori infection and does not increase the risk of gastroduodenal diseases. We suggest that a combination of the IL-8 -251 T > A polymorphism and increased IL-8 production in response to H. pylori infection may be a risk factor for severe AG and GC development in a Korean population.

Acknowledgements

We thank Ms. Eunhee Lee for encouragement and scientific discussions.

Funding

The work was not granted.

Availability of data and materials

The datasets of the current study available from the corresponding author on reasonable request.

Authors’ contributions

YWC, CHO, and JWK participated in study design and the conception. YWC wrote the manuscript. JWL and MJP participated in the design of the study and performed the statistical analysis. JJS and CKL are responsible for acquisition of data. JYJ, SHD and HJK carried out quality control of data and algorithms. SSK, BHK and JWK are responsible for critical revision of the work. All authors reviewed and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
The Institutional Review Board of the Kyung Hee University Hospital approved this study. Written informed consent was obtained from the all included subjects.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat McColl KE. Clinical practice. Helicobacter pylori infection. N Engl J Med. 2010;362(17):1597–604.CrossRefPubMed McColl KE. Clinical practice. Helicobacter pylori infection. N Engl J Med. 2010;362(17):1597–604.CrossRefPubMed
2.
Zurück zum Zitat Lim SH, Kwon JW, Kim N, Kim GH, Kang JM, Park MJ, et al. Prevalence and risk factors of Helicobacter pylori infection in Korea: nationwide multicenter study over 13 years. BMC Gastroenterol. 2013;13:104–13. Lim SH, Kwon JW, Kim N, Kim GH, Kang JM, Park MJ, et al. Prevalence and risk factors of Helicobacter pylori infection in Korea: nationwide multicenter study over 13 years. BMC Gastroenterol. 2013;13:104–13.
4.
Zurück zum Zitat Miehlke S, Kibler K, Kim JG, Figura N, Small SM, Graham DY, et al. Allelic variation in the cagA gene of Helicobacter pylori obtained from Korea compared to the United States. Am J Gastroenterol. 1996;91(7):1322–5. Miehlke S, Kibler K, Kim JG, Figura N, Small SM, Graham DY, et al. Allelic variation in the cagA gene of Helicobacter pylori obtained from Korea compared to the United States. Am J Gastroenterol. 1996;91(7):1322–5.
5.
Zurück zum Zitat Park SK, Kim GH, Jeong EJ, Bae YM, Heo J, Chu HJ, et al. Clinical relevance between the cagA, vacA, and iceA status of Helicobacter pylori and benign gastroduodenal diseases. Korean J Gastroenterol. 2002;40(1):23–31. Park SK, Kim GH, Jeong EJ, Bae YM, Heo J, Chu HJ, et al. Clinical relevance between the cagA, vacA, and iceA status of Helicobacter pylori and benign gastroduodenal diseases. Korean J Gastroenterol. 2002;40(1):23–31.
6.
Zurück zum Zitat Chang YW, Han YS, Lee DK, Kim HJ, Lim HS, Moon JS, et al. Role of Helicobacter pylori infection among offspring or siblings of gastric cancer patients. Int J Cancer. 2002;101(5):469–74. Chang YW, Han YS, Lee DK, Kim HJ, Lim HS, Moon JS, et al. Role of Helicobacter pylori infection among offspring or siblings of gastric cancer patients. Int J Cancer. 2002;101(5):469–74.
7.
Zurück zum Zitat Ferrero RL. Innate immune recognition of the extracellular mucosal pathogen, Helicobacter pylori. Mol Immunol. 2005;42(8):879–85.CrossRefPubMed Ferrero RL. Innate immune recognition of the extracellular mucosal pathogen, Helicobacter pylori. Mol Immunol. 2005;42(8):879–85.CrossRefPubMed
8.
Zurück zum Zitat Rad R, Dossumbekova A, Neu B, Lang R, Bauer S, Saur D, et al. Cytokine gene polymorphisms influence mucosal cytokine expression, gastric inflammation, and host specific colonisation during Helicobacter pylori infection. Gut. 2004;53(8):1082–9. Rad R, Dossumbekova A, Neu B, Lang R, Bauer S, Saur D, et al. Cytokine gene polymorphisms influence mucosal cytokine expression, gastric inflammation, and host specific colonisation during Helicobacter pylori infection. Gut. 2004;53(8):1082–9.
9.
Zurück zum Zitat Yamaoka Y, Kita M, Kodama T, Sawai N, Kashima K, Imanishi J. Induction of various cytokines and development of severe mucosal inflammation by cagA gene positive Helicobacter pylori strains. Gut. 1997;41(4):442–51.CrossRefPubMedPubMedCentral Yamaoka Y, Kita M, Kodama T, Sawai N, Kashima K, Imanishi J. Induction of various cytokines and development of severe mucosal inflammation by cagA gene positive Helicobacter pylori strains. Gut. 1997;41(4):442–51.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Hwang IR, Kodama T, Kikuchi S, Sakai K, Peterson LE, Graham DY, et al. Effect of interleukin 1 polymorphisms on gastric mucosal interleukin 1beta production in Helicobacter pylori infection. Gastroenterology. 2002;123(6):1793–803. Hwang IR, Kodama T, Kikuchi S, Sakai K, Peterson LE, Graham DY, et al. Effect of interleukin 1 polymorphisms on gastric mucosal interleukin 1beta production in Helicobacter pylori infection. Gastroenterology. 2002;123(6):1793–803.
11.
Zurück zum Zitat Bodger K, Wyatt JI, Heatley RV. Gastric mucosal secretion of interleukin-10: relations to histopathology, Helicobacter pylori status, and tumour necrosis factor-alpha secretion. Gut. 1997;40(6):739–44.CrossRefPubMedPubMedCentral Bodger K, Wyatt JI, Heatley RV. Gastric mucosal secretion of interleukin-10: relations to histopathology, Helicobacter pylori status, and tumour necrosis factor-alpha secretion. Gut. 1997;40(6):739–44.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Yamaoka Y, Kodama T, Kita M, Imanishi J, Kashima K, Graham DY. Relation between cytokines and Helicobacter pylori in gastric cancer. Helicobacter. 2001;6(2):116–24.CrossRefPubMed Yamaoka Y, Kodama T, Kita M, Imanishi J, Kashima K, Graham DY. Relation between cytokines and Helicobacter pylori in gastric cancer. Helicobacter. 2001;6(2):116–24.CrossRefPubMed
13.
Zurück zum Zitat Taguchi A, Ohmiya N, Shirai K, Mabuchi N, Itoh A, Hirooka Y, et al. Interleukin-8 promoter polymorphism increases the risk of atrophic gastritis and gastric cancer in Japan. Cancer Epidemiol Biomark Prev. 2005;14(11):2487–93. Taguchi A, Ohmiya N, Shirai K, Mabuchi N, Itoh A, Hirooka Y, et al. Interleukin-8 promoter polymorphism increases the risk of atrophic gastritis and gastric cancer in Japan. Cancer Epidemiol Biomark Prev. 2005;14(11):2487–93.
14.
Zurück zum Zitat Ohyauchi M, Imatani A, Yonechi M, Asano N, Miura A, Iijima K, et al. The polymorphism interleukin 8–251 A/T influences the susceptibility of Helicobacter pylori related gastric diseases in the Japanese population. Gut. 2005;54(3):330–5. Ohyauchi M, Imatani A, Yonechi M, Asano N, Miura A, Iijima K, et al. The polymorphism interleukin 8–251 A/T influences the susceptibility of Helicobacter pylori related gastric diseases in the Japanese population. Gut. 2005;54(3):330–5.
15.
Zurück zum Zitat Shirai K, Ohmiya N, Taguchi A, Mabuchi N, Yatsuya H, Itoh A, et al. Interleukin-8 gene polymorphism associated with susceptibility to non-cardia gastric carcinoma with microsatellite instability. J Gastroenterol Hepatol. 2006;21:1129–35.CrossRefPubMed Shirai K, Ohmiya N, Taguchi A, Mabuchi N, Yatsuya H, Itoh A, et al. Interleukin-8 gene polymorphism associated with susceptibility to non-cardia gastric carcinoma with microsatellite instability. J Gastroenterol Hepatol. 2006;21:1129–35.CrossRefPubMed
16.
Zurück zum Zitat Ye BD, Kim SG, Park JH, Kim JS, Jung HC, Song IS. The interleukin-8 -251 a allele is associated with increased risk of noncardia gastric adenocarcinoma in Helicobacter pylori-infected Koreans. J Clin Gastroenterol. 2009;43:233–9.CrossRefPubMed Ye BD, Kim SG, Park JH, Kim JS, Jung HC, Song IS. The interleukin-8 -251 a allele is associated with increased risk of noncardia gastric adenocarcinoma in Helicobacter pylori-infected Koreans. J Clin Gastroenterol. 2009;43:233–9.CrossRefPubMed
17.
Zurück zum Zitat Kang JM, Kim N, Lee DH, Park JH, Lee MK, Kim JS, et al. The effects of genetic polymorphisms of IL-6, IL-8, and IL-10 on Helicobacter pylori induced gastroduodenal diseases in Korea. J Clin Gastroenterol. 2009;43:420–8.CrossRefPubMed Kang JM, Kim N, Lee DH, Park JH, Lee MK, Kim JS, et al. The effects of genetic polymorphisms of IL-6, IL-8, and IL-10 on Helicobacter pylori induced gastroduodenal diseases in Korea. J Clin Gastroenterol. 2009;43:420–8.CrossRefPubMed
18.
Zurück zum Zitat Lee WP, Tai DI, Lan KH, Li AF, Hsu HC, Lin EJ, et al. The −251T allele of the interleukin-8 promoter is associated with increased risk of gastric carcinoma featuring diffuse-type histopathology in Chinese population. Clin Cancer Res. 2005;11:6431–41.CrossRefPubMed Lee WP, Tai DI, Lan KH, Li AF, Hsu HC, Lin EJ, et al. The −251T allele of the interleukin-8 promoter is associated with increased risk of gastric carcinoma featuring diffuse-type histopathology in Chinese population. Clin Cancer Res. 2005;11:6431–41.CrossRefPubMed
19.
Zurück zum Zitat Lu W, Pan K, Zhang L, Lin D, Miao X, You W. Genetic polymorphisms of interleukin (IL)-1B, IL-1RN, IL-8, IL-10 and tumor necrosis factor a and risk of gastric cancer in a Chinese population. Carcinogenesis. 2005;26:631–6.CrossRefPubMed Lu W, Pan K, Zhang L, Lin D, Miao X, You W. Genetic polymorphisms of interleukin (IL)-1B, IL-1RN, IL-8, IL-10 and tumor necrosis factor a and risk of gastric cancer in a Chinese population. Carcinogenesis. 2005;26:631–6.CrossRefPubMed
20.
Zurück zum Zitat Zeng ZR, Zhou SZ, Liao SY, Chen B, Li CJ, Chen MH, et al. Correlation of polymorphism of interleukin 8 gene -251 locus and gastric cancer in high and low prevalence regions in China. J Sun Yat-Sen Univ (Med Sci) 2005;26:537–540. Zeng ZR, Zhou SZ, Liao SY, Chen B, Li CJ, Chen MH, et al. Correlation of polymorphism of interleukin 8 gene -251 locus and gastric cancer in high and low prevalence regions in China. J Sun Yat-Sen Univ (Med Sci) 2005;26:537–540.
21.
Zurück zum Zitat Savage SA, Hou L, Lissowska J, Chow WH, Zatonski W, Chanock SJ, et al. Interleukin-8 polymorphisms are not associated with gastric cancer risk in a polish population. Cancer Epidemiol Biomark Prev. 2006;15:589–91.CrossRef Savage SA, Hou L, Lissowska J, Chow WH, Zatonski W, Chanock SJ, et al. Interleukin-8 polymorphisms are not associated with gastric cancer risk in a polish population. Cancer Epidemiol Biomark Prev. 2006;15:589–91.CrossRef
22.
Zurück zum Zitat Kamangar F, Abnet CC, Hutchinson AA, Newschaffer CJ, Helzlsouer K, Shugart YY, et al. Polymorphisms in inflammation-related genes and risk of gastric cancer (Finland). Cancer Causes Control. 2006;17:117–25.CrossRefPubMed Kamangar F, Abnet CC, Hutchinson AA, Newschaffer CJ, Helzlsouer K, Shugart YY, et al. Polymorphisms in inflammation-related genes and risk of gastric cancer (Finland). Cancer Causes Control. 2006;17:117–25.CrossRefPubMed
23.
Zurück zum Zitat Canedo P, Castanheira-Vale AJ, Lunet N, Pereira F, Figueiredo C, Gioia-Patricola L, et al. The interleukin-8 -251 T/a polymorphism is not associated with risk for gastric carcinoma development in a Portuguese population. Eur J Cancer Prev. 2008;17:28–32.CrossRefPubMed Canedo P, Castanheira-Vale AJ, Lunet N, Pereira F, Figueiredo C, Gioia-Patricola L, et al. The interleukin-8 -251 T/a polymorphism is not associated with risk for gastric carcinoma development in a Portuguese population. Eur J Cancer Prev. 2008;17:28–32.CrossRefPubMed
24.
Zurück zum Zitat Liu L, Zhuang W, Wang C, Chen Z, Wu XT, Zhou Y. Interleukin-8 -251 a/T gene polymorphism and gastric cancer susceptibility: a meta-analysis of epidemiological studies. Cytokine. 2010;50(3):328–34.CrossRefPubMed Liu L, Zhuang W, Wang C, Chen Z, Wu XT, Zhou Y. Interleukin-8 -251 a/T gene polymorphism and gastric cancer susceptibility: a meta-analysis of epidemiological studies. Cytokine. 2010;50(3):328–34.CrossRefPubMed
25.
Zurück zum Zitat Kutikhin AG. Association of polymorphisms in TLR genes and in genes of the toll-like receptor signaling pathway with cancer risk. Hum Immunol. 2011;72(11):1095–116.CrossRefPubMed Kutikhin AG. Association of polymorphisms in TLR genes and in genes of the toll-like receptor signaling pathway with cancer risk. Hum Immunol. 2011;72(11):1095–116.CrossRefPubMed
26.
Zurück zum Zitat Jack DL, Klein NJ, Turner MW. Mannose-binding lectin: targeting the microbial world for complement attack and opsonophagocytosis. Immunol Rev. 2001;180:86–99.CrossRefPubMed Jack DL, Klein NJ, Turner MW. Mannose-binding lectin: targeting the microbial world for complement attack and opsonophagocytosis. Immunol Rev. 2001;180:86–99.CrossRefPubMed
27.
Zurück zum Zitat Kuhlman M, Joiner K, Ezekowitz RA. The human mannose-binding protein functions as an opsonin. J Exp Med. 1989;169(5):1733–45.CrossRefPubMed Kuhlman M, Joiner K, Ezekowitz RA. The human mannose-binding protein functions as an opsonin. J Exp Med. 1989;169(5):1733–45.CrossRefPubMed
28.
Zurück zum Zitat Kuipers S, Aerts PC, van Dijk H. Differential microorganism-induced mannose-binding lectin activation. FEMS Immunol Med Microbiol. 2003;36(1–2):33–9.CrossRefPubMed Kuipers S, Aerts PC, van Dijk H. Differential microorganism-induced mannose-binding lectin activation. FEMS Immunol Med Microbiol. 2003;36(1–2):33–9.CrossRefPubMed
29.
Zurück zum Zitat Berstad AE, Brandtzaeg P, Stave R, Halstensen TS. Epithelium related deposition of activated complement in Helicobacter pylori associated gastritis. Gut. 1997;40(2):196–203.CrossRefPubMedPubMedCentral Berstad AE, Brandtzaeg P, Stave R, Halstensen TS. Epithelium related deposition of activated complement in Helicobacter pylori associated gastritis. Gut. 1997;40(2):196–203.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Scudiero O, Nardone G, Omodei D, Tatangelo F, Vitale DF, Salvatore F, et al. A mannose-binding lectin-defective haplotype is a risk factor for gastric cancer. Clin Chem. 2006;52(8):1625–7. Scudiero O, Nardone G, Omodei D, Tatangelo F, Vitale DF, Salvatore F, et al. A mannose-binding lectin-defective haplotype is a risk factor for gastric cancer. Clin Chem. 2006;52(8):1625–7.
31.
Zurück zum Zitat Baccarelli A, Hou L, Chen J, Lissowska J, El-Omar EM, Grillo P, et al. Mannose-binding lectin-2 genetic variation and stomach cancer risk. Int J Cancer. 2006;119(8):1970–5. Baccarelli A, Hou L, Chen J, Lissowska J, El-Omar EM, Grillo P, et al. Mannose-binding lectin-2 genetic variation and stomach cancer risk. Int J Cancer. 2006;119(8):1970–5.
32.
Zurück zum Zitat Wang FY, Tahara T, Arisawa T, Shibata T, Yamashita H, Nakamura M, et al. Mannan-binding lectin (MBL) polymorphism and gastric cancer risk in Japanese population. Dig Dis Sci. 2008;53(11):2904–8. Wang FY, Tahara T, Arisawa T, Shibata T, Yamashita H, Nakamura M, et al. Mannan-binding lectin (MBL) polymorphism and gastric cancer risk in Japanese population. Dig Dis Sci. 2008;53(11):2904–8.
33.
Zurück zum Zitat Worthley DL, Mullighan CG, Dean MM, Gordon DL, Phillips P, Heatley S, et al. Mannose-binding lectin deficiency does not increase the prevalence of Helicobacter pylori seropositivity. Eur J Gastroenterol Hepatol. 2007;19(2):147–52. Worthley DL, Mullighan CG, Dean MM, Gordon DL, Phillips P, Heatley S, et al. Mannose-binding lectin deficiency does not increase the prevalence of Helicobacter pylori seropositivity. Eur J Gastroenterol Hepatol. 2007;19(2):147–52.
34.
Zurück zum Zitat Madsen HO, Garred P, Thiel S, Kurtzhals JA, Lamm LU, Ryder LP, et al. Interplay between promoter and structural gene variants control basal serum level of mannan-binding protein. J Immunol. 1995;155(6):3013–20. Madsen HO, Garred P, Thiel S, Kurtzhals JA, Lamm LU, Ryder LP, et al. Interplay between promoter and structural gene variants control basal serum level of mannan-binding protein. J Immunol. 1995;155(6):3013–20.
35.
Zurück zum Zitat Madsen HO, Satz ML, Hogh B, Svejgaard A, Garred P. Different molecular events result in low protein levels of mannan-binding lectin in populations from southeast Africa and South America. J Immunol. 1998;161(6):3169–75.PubMed Madsen HO, Satz ML, Hogh B, Svejgaard A, Garred P. Different molecular events result in low protein levels of mannan-binding lectin in populations from southeast Africa and South America. J Immunol. 1998;161(6):3169–75.PubMed
36.
Zurück zum Zitat Madsen HO, Garred P, Kurtzhals JA, Lamm LU, Ryder LP, Thiel S, et al. A new frequent allele is the missing link in the structural polymorphism of the human mannan-binding protein. Immunogenetics. 1994;40(1):37–44. Madsen HO, Garred P, Kurtzhals JA, Lamm LU, Ryder LP, Thiel S, et al. A new frequent allele is the missing link in the structural polymorphism of the human mannan-binding protein. Immunogenetics. 1994;40(1):37–44.
37.
Zurück zum Zitat Sumiya M, Super M, Tabona P, Levinsky RJ, Arai T, Turner MW, et al. Molecular basis of opsonic defect in immunodeficient children. Lancet. 1991;337(8757):1569–70. Sumiya M, Super M, Tabona P, Levinsky RJ, Arai T, Turner MW, et al. Molecular basis of opsonic defect in immunodeficient children. Lancet. 1991;337(8757):1569–70.
38.
Zurück zum Zitat Lipscombe RJ, Sumiya M, Hill AV, Lau YL, Levinsky RJ, Summerfield JA, et al. High frequencies in African and non-African populations of independent mutations in the mannose binding protein gene. Hum Mol Genet. 1992;1(9):709–15. Lipscombe RJ, Sumiya M, Hill AV, Lau YL, Levinsky RJ, Summerfield JA, et al. High frequencies in African and non-African populations of independent mutations in the mannose binding protein gene. Hum Mol Genet. 1992;1(9):709–15.
39.
Zurück zum Zitat Shi H, Wang F, Jin L, Liu M, Hong W, Du Q, et al. Genotype polymorphism and its implications of mannose-binding protein allele in 5 Chinese nationalities. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2001;18(3):202–5. Shi H, Wang F, Jin L, Liu M, Hong W, Du Q, et al. Genotype polymorphism and its implications of mannose-binding protein allele in 5 Chinese nationalities. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2001;18(3):202–5.
40.
Zurück zum Zitat Gomi K, Tokue Y, Kobayashi T, Takahashi H, Watanabe A, Fujita T, et al. Mannose-binding lectin gene polymorphism is a modulating factor in repeated respiratory infections. Chest. 2004;126(1):95–9. Gomi K, Tokue Y, Kobayashi T, Takahashi H, Watanabe A, Fujita T, et al. Mannose-binding lectin gene polymorphism is a modulating factor in repeated respiratory infections. Chest. 2004;126(1):95–9.
41.
Zurück zum Zitat Lee SG, Yum JS, Moon HM, Kim HJ, Yang YJ, Kim HL, et al. Analysis of mannose-binding lectin2 (MBL2) genotype and the serum protein levels in the Korean population. Mol Immunol. 2005;42(8):969–77. Lee SG, Yum JS, Moon HM, Kim HJ, Yang YJ, Kim HL, et al. Analysis of mannose-binding lectin2 (MBL2) genotype and the serum protein levels in the Korean population. Mol Immunol. 2005;42(8):969–77.
42.
Zurück zum Zitat Rugge M, Genta RM. Staging and grading of chronic gastritis. Hum Pathol. 2005;36(3):228–33.CrossRefPubMed Rugge M, Genta RM. Staging and grading of chronic gastritis. Hum Pathol. 2005;36(3):228–33.CrossRefPubMed
43.
Zurück zum Zitat Boniotto M, Radillo O, Braida L, Pirulli D, Città A, Not T, et al. Detection of MBL-2 gene expression in intestinal biopsies of celiac patients by in situ reverse transcription polymerase chain reaction. Eur J Histochem. 2003;47:177–80. Boniotto M, Radillo O, Braida L, Pirulli D, Città A, Not T, et al. Detection of MBL-2 gene expression in intestinal biopsies of celiac patients by in situ reverse transcription polymerase chain reaction. Eur J Histochem. 2003;47:177–80.
44.
Zurück zum Zitat Bayraktaroğlu T, Aras AS, Aydemir S, Davutoğlu C, Ustündağ Y, Atmaca H, et al. Serum levels of tumor necrosis factor-alpha, interleukin-6 and interleukin-8 are not increased in dyspeptic patients with Helicobacter pylori-associated gastritis. Mediat Inflamm. 2004;13:25–8. Bayraktaroğlu T, Aras AS, Aydemir S, Davutoğlu C, Ustündağ Y, Atmaca H, et al. Serum levels of tumor necrosis factor-alpha, interleukin-6 and interleukin-8 are not increased in dyspeptic patients with Helicobacter pylori-associated gastritis. Mediat Inflamm. 2004;13:25–8.
45.
Zurück zum Zitat Hold GL, Rabkin CS, Chow WH, et al. A functional polymorphism of toll-like receptor 4 gene increases risk of gastric carcinoma and its precursors. Gastroenterology. 2007;132(3):905–12.CrossRefPubMed Hold GL, Rabkin CS, Chow WH, et al. A functional polymorphism of toll-like receptor 4 gene increases risk of gastric carcinoma and its precursors. Gastroenterology. 2007;132(3):905–12.CrossRefPubMed
46.
Zurück zum Zitat Santini D, Angeletti S, Ruzzo A, et al. Toll-like receptor 4 Asp299Gly and Thr399Ile polymorphisms in gastric cancer of intestinal and diffuse histotypes. Clin Exp Immunol. 2008;154(3):360–4.CrossRefPubMedPubMedCentral Santini D, Angeletti S, Ruzzo A, et al. Toll-like receptor 4 Asp299Gly and Thr399Ile polymorphisms in gastric cancer of intestinal and diffuse histotypes. Clin Exp Immunol. 2008;154(3):360–4.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Tahara T, Arisawa T, Wang F, et al. Toll-like receptor 2–196 to 174del polymorphism influences the susceptibility of Japanese people to gastric cancer. Cancer Sci. 2007;98(11):1790–4.CrossRefPubMed Tahara T, Arisawa T, Wang F, et al. Toll-like receptor 2–196 to 174del polymorphism influences the susceptibility of Japanese people to gastric cancer. Cancer Sci. 2007;98(11):1790–4.CrossRefPubMed
48.
Zurück zum Zitat Castaño-Rodríguez N, Kaakoush NO, Goh KL, Fock KM, Mitchell HM. The role of TLR2, TLR4 and CD14 genetic polymorphisms in gastric carcinogenesis: a case-control study and meta-analysis. PLoS One. 2013;8(4):e60327.CrossRefPubMedPubMedCentral Castaño-Rodríguez N, Kaakoush NO, Goh KL, Fock KM, Mitchell HM. The role of TLR2, TLR4 and CD14 genetic polymorphisms in gastric carcinogenesis: a case-control study and meta-analysis. PLoS One. 2013;8(4):e60327.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Mayerle J, den Hoed CM, Schurmann C, Stolk L, Homuth G, Peters MJ, et al. Identification of genetic loci associated with Helicobacter pylori serologic status. JAMA. 2013;309(18):1912–20. Mayerle J, den Hoed CM, Schurmann C, Stolk L, Homuth G, Peters MJ, et al. Identification of genetic loci associated with Helicobacter pylori serologic status. JAMA. 2013;309(18):1912–20.
50.
Zurück zum Zitat Turner MW. The role of mannose-binding lectin in health and disease. Mol Immunol. 2003;40(7):423–9.CrossRefPubMed Turner MW. The role of mannose-binding lectin in health and disease. Mol Immunol. 2003;40(7):423–9.CrossRefPubMed
51.
Zurück zum Zitat Øhlenschlaeger T, Garred P, Madsen HO, Jacobsen S. Mannose-binding lectin variant alleles and the risk of arterial thrombosis in systemic lupus erythematosus. N Engl J Med. 2004;351(3):260–7.CrossRefPubMed Øhlenschlaeger T, Garred P, Madsen HO, Jacobsen S. Mannose-binding lectin variant alleles and the risk of arterial thrombosis in systemic lupus erythematosus. N Engl J Med. 2004;351(3):260–7.CrossRefPubMed
52.
Zurück zum Zitat Ko GH, Kang SM, Kim YK, Lee JH, Park CK, Youn HS, et al. Invasiveness of Helicobacter pylori into human gastric mucosa. Helicobacter. 1999;4(2):77–81. Ko GH, Kang SM, Kim YK, Lee JH, Park CK, Youn HS, et al. Invasiveness of Helicobacter pylori into human gastric mucosa. Helicobacter. 1999;4(2):77–81.
53.
Zurück zum Zitat Bak-Romaniszyn L, Cedzyński M, Szemraj J, St Swierzko A, Zeman K, Kałuzyński A, et al. Mannan-binding lectin in children with chronic gastritis. Scand J Immunol. 2006;63(2):131–5. Bak-Romaniszyn L, Cedzyński M, Szemraj J, St Swierzko A, Zeman K, Kałuzyński A, et al. Mannan-binding lectin in children with chronic gastritis. Scand J Immunol. 2006;63(2):131–5.
54.
Zurück zum Zitat Tahara T, Shibata T, Wang FY, Nakamura M, Yamashita H, Yoshioka D, et al. Mannan-binding lectin B allele is associated with a risk of developing more severe gastric mucosal atrophy in Helicobacter pylori-infected Japanese patients. Eur J Gastroenterol Hepatol. 2009;21(7):781–6. Tahara T, Shibata T, Wang FY, Nakamura M, Yamashita H, Yoshioka D, et al. Mannan-binding lectin B allele is associated with a risk of developing more severe gastric mucosal atrophy in Helicobacter pylori-infected Japanese patients. Eur J Gastroenterol Hepatol. 2009;21(7):781–6.
55.
Zurück zum Zitat Chang YW, Oh HC, Jang JY, Hwangbo Y, Lee JW, Lee HJ, et al. IL-1beta and IL-8, matrix metalloproteinase 3, and pepsinogen secretion before and after H. pylori eradication in gastroduodenal phenotypes. Scand J Gastroenterol. 2008;43(10):1184–93. Chang YW, Oh HC, Jang JY, Hwangbo Y, Lee JW, Lee HJ, et al. IL-1beta and IL-8, matrix metalloproteinase 3, and pepsinogen secretion before and after H. pylori eradication in gastroduodenal phenotypes. Scand J Gastroenterol. 2008;43(10):1184–93.
56.
Zurück zum Zitat Miller RD, Phillips MS, Jo I, Donaldson MA, Studebaker JF, Addleman N, et al. The SNP consortium allele frequency project. High-density single-nucleotide polymorphism maps of the human genome. Genomics. 2005;86:117–26.CrossRefPubMedPubMedCentral Miller RD, Phillips MS, Jo I, Donaldson MA, Studebaker JF, Addleman N, et al. The SNP consortium allele frequency project. High-density single-nucleotide polymorphism maps of the human genome. Genomics. 2005;86:117–26.CrossRefPubMedPubMedCentral
Metadaten
Titel
Combination of Helicobacter pylori infection and the interleukin 8 –251 T > A polymorphism, but not the mannose-binding lectin 2 codon 54 G > A polymorphism, might be a risk factor of gastric cancer
verfasst von
Young Woon Chang
Chi Hyuk Oh
Jung-Wook Kim
Jae Won Lee
Mi Ju Park
Jae-Jun Shim
Chang Kyun Lee
Jae-Young Jang
Seok Ho Dong
Hyo Jong Kim
Sung Soo Kim
Byung-Ho Kim
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2017
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-017-3378-2

Weitere Artikel der Ausgabe 1/2017

BMC Cancer 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.