Skip to main content
Erschienen in: Cancer Cell International 1/2018

Open Access 01.12.2018 | Primary research

Polymorphisms of TGFBR1, TLR4 are associated with prognosis of gastric cancer in a Chinese population

verfasst von: Bangshun He, Tao Xu, Bei Pan, Yuqin Pan, Xuhong Wang, Jingwu Dong, Huiling Sun, Xueni Xu, Xiangxiang Liu, Shukui Wang

Erschienen in: Cancer Cell International | Ausgabe 1/2018

Abstract

Background

Helicobacter pylori (H. pylori)-induced gastric cancer is an intricate progression of immune response against H. pylori infection. IL-16, TGF-β1 and TLR4 pathways were the mediators involved in the immune response. We hypothesized that genetic variations in genes of these pathways have potential susceptibility to gastric cancer risk, and predict clinical outcomes of patients.

Methods

To investigate the susceptibility and prognostic value of genetic variations of IL-16, TGFBR1 and TLR4 pathways to gastric cancer, we performed a case–control study combined a retrospective study in a Chinese population. Genotyping for all polymorphisms was based on the Sequenom’s MassARRAY platform, and H. pylori infection was determined by using an immunogold testing kit.

Results

We found rs10512263 CC genotype was found to be a decreased risk of gastric cancer (CC vs. TT: adjusted OR = 0.54, 95% CI 0.31–0.97); however, rs334348 GG genotype was associated with increased risk of gastric cancer (GG vs. AA: adjusted OR = 1.51, 95% CI 1.05–2.18). We found that carriers harboring rs1927911 A allele (GA/AA) or rs10512263 C allele (CT/CC) have unfavorable survival time than none carriers (rs1927911: GA/AA vs. GG: adjusted HR = 1.27, 95% CI 1.00–1.63; rs10512263: CT/CC vs. TT: adjusted HR = 1.29, 95% CI 1.02–1.63) and that individuals harboring both two minor alleles (rs1927911 GA/AA and rs10512263 CT/CC) suffered a significant unfavorable survival (adjusted HR = 1.64, 95% CI 1.17–2.31).

Conclusion

In short, we concluded that two polymorphisms (rs334348, rs10512263) in TGFBR1 were associated with risk of gastric cancer, and that TLR4 rs1927911 and TGFBR1 rs10512263 were associated with clinical outcomes of gastric cancer patients.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12935-018-0682-0) contains supplementary material, which is available to authorized users.
Bangshun He and Tao Xu contributed equally to this study
Abkürzungen
TGF-β1
transforming growth factor beta-1
TGFBR1
TGF-β receptor 1
H. pylori
Helicobacter pylori
TLRs
toll-like receptors
LPS
lipopolysaccharide
MAF
minor allele frequency
5′ UTR
5′ untranslated regions
OR
odds ratios
CI
confidence intervals
HR
hazard ratios
HWE
Hardy–Weinberg equilibrium
NCGC
non-cardiac gastric cancer

Background

Gastric cancer is the fifth most common cancer worldwide and ranks third cause of cancer related mortality [1]. Almost over half of new diagnosed cases are from eastern Asian, predominantly in China [2]. Gastric cancer is a multifactorial disease with multistep etiology. Epidemiological studies have demonstrated that interaction of environmental factors, such as Helicobacter pylori (H. pylori) infection, excessive salt intake, alcohol drinking and tobacco smoking, and genetic background was regarded as risk of gastric cancer.
For environmental factors, H. pylori causing chronic inflammation has been verified as a key factor involved in gastric carcinogenesis. Moreover, for genetic background, polymorphisms in immune-related genes, such as IL-1B, IL-1RN, IL-10, could affect their expression and were suggested as risk factors of gastric cancer [3, 4]. In addition, we previously reported genetic polymorphisms in the promoter of IL-1B/IL-1RN were the risk of gastric cancer [5, 6]. Of immune-related genes, IL-16 is a pro-inflammatory cytokine that has a variety of biological functions, playing role in the development and homeostasis of the immune system [7], and stimulating the secretion of tumor-associated inflammatory cytokines including TNF-α, IL-1β, IL-6, and IL-15 [8]. In addition, polymorphisms in IL-16 were investigated to be risk of various cancers, including gastric cancer, and the diagnostic and prognostic value of serum IL-16 levels for patients with gastric cancer was also reported [9]. Transforming growth factor beta-1 (TGF-β1), a multifunctional cytokine, combined it’s receptor (TGFBR1) plays biphasic role in carcinogenesis that, in early stages of cancer, it acts as a tumor suppressor by inhibiting cellular proliferation or by promoting cellular differentiation and apoptosis; in later stages of cancer, however, it turns to be a tumor promoter by stimulating angiogenesis and cell motility, suppressing immune response, and increasing progressive invasion and metastasis [1012]. Moreover, serum TGF-β1 levels implicating a predictive and prognostic value for patients with gastric cancer [13, 14] may indicate polymorphisms in genes of TGF-β1 pathway including TGFBR1 could influence the risk and clinical progression of gastric cancer [1517]. In the progression of H. pylori infection, toll-like receptors (TLRs), a group of membrane-bound receptors proteins, play a pivotal role in innate immune response and provide first line of host defense. Among TLRs, TLR-4 is the main receptor of lipopolysaccharide (LPS) and plays a role in initiating the inflammatory response of H. pylori infection. After binding of microbial ligands, a dysregulation of TLR signalling may contribute to an unbalanced ratio between pro- and anti-inflammatory cytokines, resulting in increasing higher risk of developing gastric cancer [18]. Similarly, polymorphisms in TLR4 has been implicated as risk factors for gastric cancer [18]; however, the conclusion of susceptibility of these polymorphisms to gastric cancer risk remains elusive [1921].
Immune response triggered by H. pylori infection, including host adaptive immune response (such as IL-1b, TNF-a, IL-10, IL-16) and innate immune response (such as TLR4), is an intricate progression, which is responsible for clinical outcomes of individuals with H. pylori infection. Thus, polymorphisms occurring in immune genes could serves as possible susceptibility factors to the development of gastric cancer and have a predictive value for gastric cancer clinical outcome. Here, we conducted a case–control study to assess the susceptibility of polymorphisms in IL-16, TGFBR1 and TLR4 to risk of gastric cancer in a Chinese population, and the prognostic value of the polymorphisms was also evaluated by a retrospective study.

Materials and methods

Study population

For the case–controls study, we recruited 479 patents histologically diagnosed as gastric cancer and 483 age- and sex-matched healthy controls who came to the hospital for routine physical examination. The demographic features of participants were collected via a questionnaire or by reviewing patients’ medical records. The TNM stages were classified according to American Joint Commission for Cancer Staging in 2002 (the sixth edition). For retrospective study, we traced survival state of all patients through on-site interview, direct calling or medical chart review, and finally, a total of 460 patients were followed up to 5 years. The protocol of this study was approved by the Institutional Review Board of the Nanjing First Hospital, and written informed consents were obtained from all participants.

DNA extraction and genotyping

We retrieved the potential genetic variations in IL-16, TGF-BR1 and TLR4 from the National Center for Biotechnology Information dbSNP database (http://​www.​ncbi.​nlm.​nih.​gov/​projects/​SNP), and then the genetic variations were selected followed the following criteria: (1) the minor allele frequency (MAF) is not less than 5% in Han Chinese population; (2) with position in exons, promoter region, 5′ untranslated regions (UTR) or 3′ UTR; and (3) published results shown to be associated with any cancer risk. For those polymorphisms in intron if meet the criterion (3) were also included. Finally, a total of 11 polymorphisms were selected (Additional file 1: Table S1).
The DNA extraction and genotyping was performed as we previously described [22]. A GoldMag-Mini Whole Blood Genomic DNA Purification Kit (GoldMag Co. Ltd. Xi’an, China) was used for DNA extraction, and then the genotyping was performed on the SequenomMassARRAY platform.

H. pylori infection detection

To identify the H. pylori infection, the serum of all participants were collected to detect H. pylori antibody by using a H. pylori immunogold testing kit (KangmeiTianhong Biotech Co., Ltd, Beijing, China).

Statistical analysis

The difference of demographic features of the two groups was assessed by t test or χ2 test. For the distribution of genotypes, a goodness of fit Chi square test was adopted to test the Hardy–Weinberg equilibrium (HWE) in the control group, and then, the susceptibility of polymorphisms to gastric cancer risk was expressed with odds ratios (ORs) and 95% confidence intervals (CIs). Subgroups analyze was conducted if there was a significant association of the polymorphism to gastric cancer risk. The risk of polymorphisms was calculated by using a logistic regression model based on SAS v9.1 (SAS Institute, Cary, NC, USA). The hazard ratios (HRs) of genotypes to survival time of patients were calculated by Cox regression analysis with SPSS 11.0 (SPSS, Chicago, IL, USA). The p value < 0.05 was considered statistically significant difference.

Result

Characteristics of the study population

The health controls and patients were matched for age (p = 0.748) and gender (p = 0.881). There were significant differences between the two groups with respect to the frequency of H. pylori infection (p = 0.039), cigarette smoking (p < 0.001) and alcohol consumption (p < 0.001), summarized in Additional file 1: Table S2. The observed frequencies of all tested genotypes in controls did not deviate from HWE (shown in Additional file 1: Table S1).

Association between polymorphisms and risk of gastric cancer

Two polymorphisms in TGFBR1 were observed to be potentially associated with risk of gastric cancer. rs10512263 CC genotype was found to be a decreased risk of gastric cancer (CC vs. TT: adjusted OR = 0.54, 95% CI 0.31–0.97, p = 0.039); however, rs334348 GG genotype was associated with increased risk of gastric cancer (GG vs. AA: adjusted OR = 1.51, 95% CI 1.05–2.18, p = 0.028), shown in Table 1.
Table 1
Association between polymorphisms and risk of gastric cancer
Polymorphism
Genotype
Cases, n (%)
Controls, n (%)
OR (95% CI)
OR (95% CI)a
p value
IL-16 rs4072111
CC
334 (69.73)
345 (71.43)
Reference
Reference
 
TC
126 (26.30)
122 (25.26)
1.07 (0.80,1.43)
1.01 (0.75,1.36)
0.970
TT
19 (3.97)
16 (3.31)
1.23 (0.62,2.43)
1.20 (0.60,2.41)
0.600
TC/TT
145 (30.27)
138 (28.57)
1.09 (0.82,1.43)
1.02 (0.77,1.36)
0.870
Additive model
  
1.08 (0.86,1.37)
1.04 (0.82,1.32)
0.755
rs4778889
TT
267 (55.74)
266 (55.07)
Reference
Reference
 
CT
182 (38.00)
192 (39.75)
0.94 (0.73,1.23)
0.92 (0.70,1.20)
0.524
CC
30 (6.26)
25 (5.18)
1.20 (0.68,2.09)
1.19 (0.68,2.10)
0.542
CT/CC
212 (44.26)
217 (44.93)
0.97 (0.76,1.26)
0.95 (0.73,0.23)
0.688
Additive model
  
1.01 (0.82,1.25)
1.00 (0.81,1.23)
0.965
rs859
TT
129 (26.93)
124 (25.67)
Reference
Reference
 
CT
235 (49.06)
248 (51.35)
0.91 (0.67,1.24)
0.88 (0.64,1.20)
0.406
CC
115 (24.01)
111 (22.98)
1.00 (0.70,1.43)
0.98 (0.68,1.41)
0.899
CT/CC
350 (73.07)
359 (74.33)
0.94 (0.70,1.25)
0.92 (0.68,1.23)
0.551
Additive model
  
1.00 (0.83,1.19)
0.98 (0.82,1.18)
0.859
rs11556218
TT
306 (63.88)
308 (63.77)
Reference
Reference
 
GT
151 (31.52)
157 (32.51)
1.97 (0.74,1.27)
0.93 (0.71,1.23)
0.628
GG
22 (4.59)
18 (3.73)
1.23 (0.65,2.34)
1.29 (0.68,2.48)
0.439
GT/GG
173 (36.12)
175 (36.23)
1.00 (0.77,1.29)
0.97 (0.94,1.27)
0.820
Additive model
  
1.02 (0.82,1.28)
1.01 (0.81,1.27)
0.913
rs1131445
TT
221 (46.14)
210 (43.48)
Reference
Reference
 
CT
211 (44.05)
222 (45.96)
0.90 (0.69,1.18)
0.94 (0.72,1.23)
0.655
CC
47 (9.81)
51 (10.56)
0.88 (0.57,1.36)
0.93 (0.59,1.46)
0.748
CT/CC
258 (53.86)
273 (56.52)
0.90 (0.70,1.16)
0.93 (0.72,1.21)
0.592
Additive model
  
0.92 (0.76,1.12)
0.95 (0.78,1.16)
0.618
TLR4 rs10759932
TT
240 (50.10)
251 (51.97)
Reference
Reference
 
TC
191 (39.87)
196 (40.58)
1.02 (0.78,1.33)
1.05 (0.80,1.38)
0.733
CC
48 (10.02)
36 (7.45)
1.39 (0.87,2.22)
1.38 (0.86,2.23)
0.184
TC/CC
239 (49.90)
232 (48.03)
1.08 (0.84,1.39)
1.10 (0.85,1.42)
0.481
Additive model
  
1.11 (0.91,1.35)
1.12 (0.92,1.36)
0.275
rs1927911
GG
171 (35.70)
175 (36.23)
Reference
Reference
 
GA
226 (47.18)
226 (46.79)
1.02 (0.77,1.35)
1.04 (0.78,1.38)
0.801
AA
82 (17.12)
82 (16.98)
1.02 (0.71,1.48)
0.99 (0.68,1.45)
0.967
GA/AA
308 (64.30)
308 (63.77)
1.02 (0.79,1.33)
1.03 (0.79,1.34)
0.844
Additive model
  
1.01 (0.85,1.21)
1.01 (0.84,1.21)
0.930
rs11536889
GG
303 (63.26)
293 (60.66)
Reference
Reference
 
CG
156 (32.57)
166 (34.37)
0.91 (0.69,1.19)
0.91 (0.69,1.19)
0.477
CC
20 (4.18)
24 (4.97)
0.81 (0.44,1.49)
0.76 (0.40,1.43)
0.392
CG/CC
176 (36.74)
190 (39.34)
0.90 (0.69,1.16)
0.89 (0.69,1.16)
0.402
Additive model
  
0.90 (0.73,1.13)
0.90 (0.72,1.12)
0.355
TGF-BR1 rs6478974
TT
219 (45.72)
194 (40.17)
Reference
Reference
 
AT
204 (42.59)
220 (45.55)
0.82 (0.63,1.08)
0.80 (0.61,1.06)
0.118
AA
56 (11.69)
69 (14.29)
0.72 (0.48,1.08)
0.68 (0.45,1.02)
0.063
AT/AA
260 (54.28)
289 (59.83)
0.80 (0.62,1.03)
0.78 (0.60,1.01)
0.055
Additive model
  
0.84 (0.70,1.01)
0.82 (0.68,0.99)
0.038
rs334348
AA
143 (29.85)
158 (32.71)
Reference
Reference
 
AG
221 (46.14)
240 (49.69)
1.02 (0.76,1.36)
1.05 (0.78,1.42)
0.730
GG
115 (24.01)
85 (17.60)
1.50 (1.04,2.14)
1.51 (1.05,2.18)
0.028
AG/GG
336 (70.15)
325 (69.29)
1.14 (0.87,1.50)
1.17 (0.89,1.55)
0.263
Additive model
  
1.20 (1.01,1.43)
1.22 (1.02,1.46)
0.032
rs10512263
TT
279 (58.25)
262 (54.24)
Reference
Reference
 
CT
178 (37.16)
187 (38.72)
0.89 (0.69,1.17)
0.87 (0.66,1.14)
0.297
CC
22 (4.59)
34 (7.04)
0.61 (0.35,1.07)
0.54 (0.31,0.97)
0.039
CT/CC
200 (41.75)
221 (45.76)
0.85 (0.66,1.10)
0.82 (0.63,1.06)
0.127
Additive model
  
0.84 (0.68,1.03)
0.81 (0.65,1.00)
0.047
Italic represents any values with p < 0.05
OR odds ratio
aAdjusted for age, gender, smoking, drinking, and H. pylori infection status
Stratified analysis by age, gender, H. pylori infection status, tumor stage and tumor site revealed that the significant association of rs10512263 to risk of gastric cancer was maintained in the subgroup of male, and subgroup of individuals with older age, shown in Table 2. In the stratification analysis by pathologic characteristics, we observed that the significant association of rs334348 to risk of gastric cancer was maintained in the subgroup of patients with clinical stage T1–T2. In addition, although no significant association was found, aboundary significant of two polymorphisms to risk of gastric cancer was observed in subgroup of clinical stage T1–T2 and in subgroup of non-cardiac, shown in Table 3.
Table 2
Stratification analyses the association between polymorphisms in TGF-BR1 and gastric cancer risk
Genotype
Age
Gender
H. pylori infection
≤ 64
> 64
Male
Female
Positive
Negative
Ca/Co
OR (95% CI)a
p value
Ca/Co
OR (95% CI)a
p value
Ca/Co
OR (95% CI)a
p value
Ca/Co
OR (95% CI)a
p value
Ca/Co
OR (95% CI)a
p value
Ca/Co
OR (95% CI)a
p value
rs6478974
 TT
100/87
Reference
 
119/107
Reference
 
156/134
Reference
 
63/60
Reference
 
120/91
Reference
 
99/103
Reference
 
 AT
101/113
0.76 (0.51,1.13)
0.175
103/107
0.85 (0.58,1.25)
0.412
159/170
0.80 (0.58,1.10)
0.164
45/50
0.81 (0.46,.41)
0.452
108/107
0.76 (0.52,1.12)
0.160
96/113
0.86 (0.58,1.28)
0.457
 AA
28/30
0.76 (0.41,1.40)
0.374
28/39
0.61 (0.34,1.07)
0.082
38/54
0.55 (0.33,0.90)
0.017
18/15
1.19 (0.55,2.58)
0.663
33/33
0.73 (0.41,1.28)
0.272
23/36
0.62 (0.34,1.14)
0.127
 AT/AA
129/143
0.76 (0.52,1.11)
0.156
131/146
0.80 (0.56,1.14)
0.206
197/224
0.74 (0.54,1.00)
0.049
63/65
0.89 (0.54,1.48)
0.658
141/140
0.75 (0.52,1.08)
0.125
119/149
0.81 (0.55,1.17)
0.260
 Additive model
 
0.83 (0.63,1.10)
0.199
 
0.81 (0.62,1.04)
0.100
 
0.76 (0.61,0.95)
0.016
 
1.01 (0.70,1.44)
0.975
 
0.83 (0.64,1.08)
0.157
 
0.81 (0.62,1.07)
0.135
rs334348
 AA
72/74
Reference
 
71/84
Reference
 
108/121
Reference
 
35/37
Reference
 
77/83
Reference
 
66/75
Reference
 
 AG
105/122
0.95 (0.62,1.47)
0.832
116/118
1.16 (0.77,1.76)
0.478
168/173
1.15 (0.81,1.62)
0.143
53/67
0.85 (0.46,1.54)
0.583
124/100
1.43 (0.94,2.17)
0.093
97/140
0.79 (0.51,1.21)
0.278
 GG
52/34
1.57 (0.90,2.73)
0.114
63/51
1.44 (0.88,2.37)
0.146
77/64
1.37 (0.89,2.11)
0.149
38/21
1.97 (0.94,4.11)
0.072
60/48
0.37 (0.83,2.25)
0.223
55/37
1.71 (0.99,2.53)
0.054
 AG/GG
157/156
1.09 (0.72,1.63)
0.694
179/169
1.26 (0.86,1.85)
0.245
245/237
1.20 (0.87,1.66)
0.269
91/88
1.10 (0.63,1.92)
0.745
184/148
1.40 (0.95,2.06)
0.088
152/177
0.98 (0.66,1.47)
0.927
 Additive model
 
1.21 (0.93,1.59)
0.161
 
1.22 (0.95,1.56)
0.113
 
1.17 (0.95,1.45)
0.141
 
1.36 (0.95,1.94)
0.096
 
1.19 (0.93,1.53)
0.166
 
1.25 (0.96,1.64)
0.097
rs10512263
 TT
130/129
Reference
 
149/133
Reference
 
202/189
Reference
 
77/73
Reference
 
150/127
Reference
 
129/135
Reference
 
 CT
88/88
0.95 (0.64,1.41)
0.814
90/99
0.80 (0.55,1.16)
0.230
136/140
0.89 (0.65,1.22)
0.470
42/47
0.82 (0.48,1.41)
0.471
98/87
0.91 (0.62,1.33)
0.610
80/100
0.83 (0.56,1.22)
0.335
 CC
11/13
0.70 (0.29,1.67)
0.417
11/21
0.44 (0.20,0.96)
0.040
15/29
0.41 (0.21,0.81)
0.010
7/5
1.40 (0.42,4.63)
0.585
13/17
0.64 (0.29,1.39)
0.258
9/17
0.45 (0.19,1.10)
0.080
 CT/CC
99/101
0.91 (0.62,1.33)
0.621
101/120
0.74 (0.52,1.06)
0.101
151/169
0.80 (0.59,1.09)
0.159
49/52
0.87 (0.52,1.46)
0.604
111/104
0.86 (0.60,1.23)
0.405
89/117
0.77 (0.53,1.13)
0.180
 Additive model
 
0.88 (0.64,1.21)
0.441
 
0.74 (0.55,0.99)
0.040
 
0.76 (0.60,0.98)
0.033
 
0.96 (0.63,1.48)
0.864
 
0.85 (0.63,1.14)
0.272
 
0.76 (0.56,1.04)
0.083
Italic represents any values with p < 0.05
OR odds ratio, Ca case, Co control
aAdjusted for age, gender, smoking, drinking, and H. pylori infection status
Table 3
Stratification analyses the association between polymorphisms in TGF-BR1 and gastric cancer by pathologic characteristics
Genotype
Co
Clinical stage
Tumor site
T1–T2
T3–T4
Cardiac
Non-cardiac
Ca
OR (95% CI)a
p value
Ca
OR (95% CI)a
p value
Ca
OR (95% CI)a
p value
Ca
OR (95% CI)a
p value
rs6478974
 TT
194
75
Reference
 
144
Reference
 
62
Reference
 
157
Reference
 
 AT
220
69
0.82 (0.55,1.21)
0.311
135
0.79 (0.58,1.08)
0.140
62
0.87 (0.58,1.31)
0.497
142
0.78 (0.57,1.05)
0.105
 AA
69
15
0.54 (0.28,1.04)
0.065
41
0.76 (0.49,1.20)
0.238
14
0.61 (0.32,1.17)
0.139
42
0.72 (0.45,1.13)
0.147
 AT/AA
289
84
0.75 (0.52,1.09)
0.134
176
0.79 (0.59,1.05)
0.109
76
0.80 (0.54,1.18)
0.268
184
0.77 (0.58,1.02)
0.069
 Additive model
  
0.76 (0.57,1.01)
0.054
 
0.85 (0.69,1.05)
0.140
 
0.80 (0.60,1.07)
0.135
 
0.83 (0.67,1.02)
0.077
rs334348
 AA
158
45
Reference
 
98
Reference
 
40
Reference
 
103
Reference
 
 AG
240
73
1.19 (0.76,1.87)
0.442
148
1.01 (0.73,1.41)
0.937
64
1.14 (0.73,1.80)
0.565
157
1.04 (0.75,1.45)
0.825
 GG
85
41
1.73 (1.03,2.90)
0.039
74
1.42 (0.94,2.13)
0.092
34
1.56 (0.91,2.68)
0.103
81
1.48 (0.99,2.21)
0.055
 AG/GG
325
114
1.31 (0.87,1.99)
0.196
222
1.12 (0.82,1.52)
0.487
 
1.24 (0.81,1.89)
0.323
 
1.15 (0.85,1.57)
0.373
 Additive model
  
1.33 (1.02,1.73)
0.035
 
1.17 (0.96,1.43)
0.125
 
1.25 (0.95,1.64)
0.108
 
1.21 (0.99,1.47)
0.069
rs10512263
 TT
262
99
Reference
 
180
Reference
 
82
Reference
 
197
Reference
 
 CT
187
53
0.75 (0.51,1.12)
0.161
125
0.93 (0.69,1.25)
0.620
50
0.81 (0.54,1.21)
0.300
128
0.90 (0.67,1.21)
0.479
 CC
34
7
0.51 (0.21,1.24)
0.137
15
0.59 (0.31,1.13)
0.113
6
0.51 (0.20,1.28)
0.149
16
0.56 (0.29,1.07)
0.080
 CT/CC
221
60
0.71 (0.49,1.05)
0.084
140
0.88 (0.66,1.17)
0.366
56
0.76 (0.51,1.12)
0.162
144
0.85 (0.64,1.13)
0.256
 Additive model
  
0.73 (0.53,1.01)
0.054
 
0.85 (0.67,1.08)
0.184
 
0.76 (0.55,1.05)
0.093
 
0.83 (0.66,1.05)
0.120
Italic represents any values with p < 0.05
OR odds ratio, Ca case, Co control
aAdjusted for age, gender, smoking, drinking, and H. pylori infection status

Association between polymorphisms and clinical outcome

A retrospective study was conducted based on 460 patients with follow-up information on survival period of 5 years. We found that carriers harboring rs1927911 A allele (GA/AA) or rs10512263 C allele (CT/CC) have unfavorable survival time than none carriers (rs1927911: GA/AA vs. GG: adjusted HR = 1.27, 95% CI 1.00–1.63, p = 0.054; rs10512263: CT/CC vs. TT: adjusted HR = 1.29, 95% CI 1.02–1.63, p = 0.031), shown in Table 4.
Table 4
Association between polymorphism and overall survival of gastric cancer patients in co-dominant model
Genotype
Cases, n
Death, n (%)
Log-rank p-value
HR
HR (95% CI)a
p-value
rs4072111
 CC
322
205 (0.64)
 
Reference
Reference
 
 TC/TT
138
81 (0.59)
0.344
0.88 (0.68,1.14)
1.12 (0.86,1.45)
0.408
rs4778889
 TT
256
172 (0.67)
 
Reference
Reference
 
 CT/CC
204
114 (0.56)
0.028
0.77 (0.61,0.97)
0.84 (0.66,1.06)
0.146
rs11556218
 TT
293
192 (0.66)
 
Reference
Reference
 
 GT/GG
167
94 (0.56)
0.110
0.82 (0.64,1.05)
0.94 (0.73,1.20)
0.607
rs859
 AA
109
68 (0.62)
 
Reference
Reference
 
 GA/GG
351
218 (0.62)
0.633
1.07 (0.81,1.40)
1.03 (0.79,1.36)
0.814
rs1131445
 TT
211
127 (0.60)
 
Reference
Reference
 
 CT/CC
249
159 (0.64)
0.150
1.18 (0.94,1.50)
1.06 (0.84,1.35)
0.617
rs10759932
 TT
231
141 (0.61)
 
Reference
Reference
 
 TC/CC
229
145 (0.63)
0.563
1.07 (0.85,1.35)
1.07 (0.84,1.35)
0.588
rs1927911
 GG
165
95 (0.58)
 
Reference
Reference
 
 GA/AA
295
191 (0.65)
0.113
1.22 (0.95,1.56)
1.27 (1.00,1.63)
0.054
rs11536889
 GG
293
181 (0.62)
 
Reference
Reference
 
 CG/CC
167
105 (0.63)
0.957
1.01 (0.79,1.28)
0.99 (0.77,1.26)
0.924
rs6478974
 TT
212
126 (0.59)
 
Reference
Reference
 
 TA/AA
248
160 (0.65)
0.224
1.16 (0.92,1.46)
1.23 (0.98,1.56)
0.079
rs334348
 GG
110
64 (0.58)
 
Reference
Reference
 
 AG/AA
350
222 (0.63)
0.491
1.10 (0.84,1.46)
1.04 (0.79,1.38)
0.787
rs10512263
 TT
269
157 (0.58)
 
Reference
Reference
 
 CT/CC
191
129 (0.68)
0.031
1.29 (1.02,1.63)
1.29 (1.02,1.63)
0.031
Italic represents any values with p < 0.05
aAdjusted for age, sex, tumor site and TNM stage
The stratified analysis based on the age, gender, tumor site or clinical stage was also performed for the significant polymorphisms, and the result revealed that carriers with rs1927911 A allele have poor survival in subgroup of patients with age younger than 64 years old (GA/AA vs. GG: adjusted HR = 1.64, 95% CI 1.13–2.38), male (GA/AA vs. GG: adjusted HR = 1.36, 95% CI 1.03–1.81), and non-cardiac gastric cancer (GA/AA vs. GG: adjusted HR = 1.34, 95% CI 1.00–1.80), and that rs1927911 A allele carriers have poor survival in the subgroup of male (CT/CC vs. TT: adjusted HR = 1.43, 95% CI 1.09–1.87), patients in clinical stage T1–T2 (CT/CC vs. TT: adjusted HR = 2.54, 95% CI 1.38–4.69), and non-cardiac gastric cancer (NCGC) (CT/CC vs. TT: adjusted HR = 1.36, 95% CI 1.02–1.80), shown in Table 5.
Table 5
Subgroup analyses of association between polymorphisms and survival in co-dominant model
Group
Case, n
Death, n (%)
rs1927911
rs10512263
GA/AA: GG
HR (95% CI)a
p-value
CT/CC: TT
HR (95% CI)a
P-value
Age
 < 64
224
130 (0.58)
142/82
1.64 (1.13,2.38)
0.009
97/127
1.34 (0.95,1.90)
0.099
 ≥ 64
236
156 (0.66)
153/83
1.04 (0.75,1.45)
0.817
94/142
1.19 (0.86,1.64)
0.286
Gender
 Male
338
214 (0.63)
216/122
1.36 (1.03,1.81)
0.033
145/193
1.43 (1.09,1.87)
0.010
 Female
122
72 (0.64)
79/43
1.08 (0.65,1.80)
0.754
46/76
1.26 (0.76,2.08)
0.365
Clinical stage
 T1–T2
159
42 (0.26)
102/57
1.36 (0.70,2.66)
0.367
60/99
2.61 (1.40,4.86)
0.003
 T3–T4
301
244 (0.81)
193/108
1.21 (0.93,1.58)
0.160
131/170
1.04 (0.80,1.34)
0.784
Tumor site
 Cardiac
132
87 (0.66)
91/41
1.07 (0.67,1.71)
0.768
54/78
1.47 (0.94,2.31)
0.094
 Non-cardiac
328
199 (0.61)
204/124
1.34 (1.00,1.80)
0.050
137/191
1.36 (1.02,1.80)
0.034
Italic represents any values with p < 0.05
aAdjusted for age, sex, tumor site and TNM stage
To identify the impact of the co-occurrence of rs1927911 and rs10512263 on overall survival, we analyzed the association between locus–locus interaction and overall survival, and the result shown that individuals harboring both two minor alleles (rs1927911GA/AA and rs10512263CT/CC) suffered a significant unfavorable survival (adjusted HR = 1.64, 95% CI 1.17–2.31), shown in Table 6.
Table 6
Locus–locus interactions between rs1927911 and rs10512263 and survival
rs1927911
rs10512263
Cases, n
Death, n (%)
Log-rank p value
HR (95% CI)a
p-value
GG
TT
100
53 (53.00)
0.018
Reference
 
GG
CT/CC
65
42 (64.42)
1.18 (0.79,1.03)
0.421
GA/AA
TT
169
104 (61.54)
1.20 (0.86,1.67)
0.279
GA/AA
CT/CC
126
87 (69.05)
1.64 (1.17,2.31)
0.005
Italic represents any values with p < 0.05
aAdjusted for age, sex, tumor site and TNM stage

Discussion

This case–control study combined retrospective study observed that two polymorphisms (rs334348, rs10512263) in TGFBR1 were associated with risk of gastric cancer, and that rs1927911and rs10512263 were associated with survival of gastric cancer patients.
TGFBR1 rs6478974 is a genetic variation in intron 1, it was previously reported to be associated with microRNAs expression and involved in carcinogenesis [23]. In addition, the significant association of rs6478974 to gastric cancer risk was also reported [15]; however, in this study, we observed such a significant association in the subgroup of male but for all participants, indicating male carrying rs6478974 polymorphisms have higher gastric cancer risk than female. Another polymorphism rs10512263 locating intron 1 of TGBR1 was observed as a susceptibility of gastric cancer in this study; however, an opposite result was also reported [15]. It is noted that, in the subgroup analysis, we observed that the decreased risk of the polymorphism to gastric cancer was maintained in the subgroup of male, and those with age older than 64 years, suggesting the susceptibility of the polymorphism to gastric cancer risk could be effected by demographic characteristics of participants. Due to the limited sample sized of this study, the significant should be verified by further study. TGFBR1 rs334348 located in the 3′ UTR region, and it was suggested with location in miRNA-628-5p binding site, resulting in GG genotype turn to be associated with lower TGFBR1 expression [24]. In addition, previous study has also reported that it could confer an increased risk of colorectal cancer by affecting TGFBR1 expression [25].
In the retrospective study, we observed TLR4 rs1927911 and TGFBR1 rs10512263 were associated with clinical outcomes of gastric cancer patients. TLR4 rs1927911 is an intron variation that was previously reported as a protective factor for gastric cancer [26, 27]; however, we failed to find such a significant association but we observed it was associated with unfavorable OS of gastric cancer patients, especially for male, patients with age younger than 64 years old, or patients with NCGC. To date, the function of rs1927911 remains unclear, we speculated that such a significant association was related the microenvironment of cancer by that TLR4 signaling was involved in drug resistant by inducing the M1 phenotype macrophages [28] and by that TLR4/NF-κB signal pathway mediated uncontrolled inflammation [29]. Moreover, this study observed TGFBR1 rs10512263 has a predictive value for clinical outcomes of gastric cancer patients. Although the function of rs10512263 remains unclear, TGF-β signaling has been suggested to promote gastric cancer progression by enhancing motility and inducing invasiveness of gastric cancer cell [11], or by promoting tumor vasculature conformation [30], which could be partly explained for the predictive role of TGFBR1 rs10512263 in gastric cancer patients.
Polymorphisms in three immune related genes was discussed for their susceptibility and predictive role in gastric cancer. Here, some limitations of this study should be noted. Firstly, the function of these polymorphisms is largely unclear, and we failed to assess the association of polymorphism and TGFBR1, TLR4 expression in patients. Instead of that, to perform functional candidate polymorphism and expression quantitative trait locus (eQTL) analyses on the promising genes, we mined the data from the following databases: GTExPortal (https://​www.​gtexportal.​org/​home/​) and Haploreg (http://​www.​broadinstitute.​org/​mammals/​haploreg/​haploreg.​php), and the results shown that TLR4 rs1927911, TGF-BR1 rs6478974 and rs334348 could affect their corresponding gene expression, and that TGF-BR1 rs10512263 could regulate certain motifs, which were consistent to our results, see Additional file 2: Figures S1 and S2. Secondly, the sample size of this study was not large enough, which may weaken the statistical power. Thirdly, environmental factors, such as diet, physical exercises, gastric diseases history, and subtype of H. pylori were not included in this study, which may influence the conclusion. Finally, there are number of polymorphisms in the immune related genes, here we selected three of them and some more immune related genes required to be discussed.

Conclusion

We concluded that two polymorphisms (rs334348, rs10512263) in TGF-BR1 were associated with risk of gastric cancer, and that TLR4 rs1927911 and TGFBR1 rs10512263 were associated with clinical outcomes of gastric cancer patients. This is a study firstly discussed the relation of polymorphisms in genes of IL-16, TGFBR1 and TLR4 pathways and survival time of gastric cancer patients in Chinese population and our study could provide epidemiology data for further study.

Authors’ contributions

BH and SW designed this study; TX, BP and YP collected the sample and data; XW, JD analyzed the date; TX, BP and XL conducted the experiments. BH, SW wrote the paper. All authors have reviewed the final version of the manuscript and approved to submit to your journal. All authors read and approved the final manuscript.

Acknowledgements

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

The data of the study are available from the corresponding author on reasonable request.
Not applicable.
The study has acquired approval of the Institutional Review Board of the Nanjing First Hospital, and all enrolled participants or their representatives signed the informed consent according to relevant regulations. All participants signed informed consent in the study.

Funding

This study was supported by grants from Jiangsu 333 High-level Talents Cultivating Project to B. H (No. BRA201702), Jiangsu Provincial Medical Youth Talent to B.H (QNRC2016066) and Y.P (QNRC2016074), Innovation team of Jiangsu provincial health-strengthening engineering by science and education (CXTDB2017008), and Nanjing Medical University Science and Technique Development Foundation Project to HL.S (No. 2015NJMUZD049).

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 Global Burden of Disease Cancer C, Fitzmaurice C, Dicker D, Pain A, Hamavid H, Moradi-Lakeh M, MacIntyre MF, Allen C, Hansen G, Woodbrook R, et al. The Global Burden of Cancer 2013. JAMA Oncol. 2015;1(4):505–27.CrossRef Global Burden of Disease Cancer C, Fitzmaurice C, Dicker D, Pain A, Hamavid H, Moradi-Lakeh M, MacIntyre MF, Allen C, Hansen G, Woodbrook R, et al. The Global Burden of Cancer 2013. JAMA Oncol. 2015;1(4):505–27.CrossRef
2.
Zurück zum Zitat Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–32.CrossRef Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–32.CrossRef
3.
Zurück zum Zitat Zabaleta J. Multifactorial etiology of gastric cancer. Methods Mol Biol. 2012;863:411–35.CrossRef Zabaleta J. Multifactorial etiology of gastric cancer. Methods Mol Biol. 2012;863:411–35.CrossRef
4.
Zurück zum Zitat Persson C, Canedo P, Machado JC, El-Omar EM, Forman D. Polymorphisms in inflammatory response genes and their association with gastric cancer: a HuGE systematic review and meta-analyses. Am J Epidemiol. 2011;173(3):259–70.CrossRef Persson C, Canedo P, Machado JC, El-Omar EM, Forman D. Polymorphisms in inflammatory response genes and their association with gastric cancer: a HuGE systematic review and meta-analyses. Am J Epidemiol. 2011;173(3):259–70.CrossRef
5.
Zurück zum Zitat He BS, Pan YQ, Xu YF, Zhu C, Qu LL, Wang SK. Polymorphisms in interleukin-1B (IL-1B) and interleukin 1 receptor antagonist (IL-1RN) genes associate with gastric cancer risk in the Chinese population. Dig Dis Sci. 2011;56(7):2017–23.CrossRef He BS, Pan YQ, Xu YF, Zhu C, Qu LL, Wang SK. Polymorphisms in interleukin-1B (IL-1B) and interleukin 1 receptor antagonist (IL-1RN) genes associate with gastric cancer risk in the Chinese population. Dig Dis Sci. 2011;56(7):2017–23.CrossRef
6.
Zurück zum Zitat He B, Zhang Y, Pan Y, Xu Y, Gu L, Chen L, Wang S. Interleukin 1 beta (IL1B) promoter polymorphism and cancer risk: evidence from 47 published studies. Mutagenesis. 2011;26(5):637–42.CrossRef He B, Zhang Y, Pan Y, Xu Y, Gu L, Chen L, Wang S. Interleukin 1 beta (IL1B) promoter polymorphism and cancer risk: evidence from 47 published studies. Mutagenesis. 2011;26(5):637–42.CrossRef
7.
Zurück zum Zitat Yellapa A, Bahr JM, Bitterman P, Abramowicz JS, Edassery SL, Penumatsa K, Basu S, Rotmensch J, Barua A. Association of interleukin 16 with the development of ovarian tumor and tumor-associated neoangiogenesis in laying hen model of spontaneous ovarian cancer. Int J Gynecol Cancer. 2012;22(2):199–207.CrossRef Yellapa A, Bahr JM, Bitterman P, Abramowicz JS, Edassery SL, Penumatsa K, Basu S, Rotmensch J, Barua A. Association of interleukin 16 with the development of ovarian tumor and tumor-associated neoangiogenesis in laying hen model of spontaneous ovarian cancer. Int J Gynecol Cancer. 2012;22(2):199–207.CrossRef
8.
Zurück zum Zitat Mathy NL, Scheuer W, Lanzendorfer M, Honold K, Ambrosius D, Norley S, Kurth R. Interleukin-16 stimulates the expression and production of pro-inflammatory cytokines by human monocytes. Immunology. 2000;100(1):63–9.CrossRef Mathy NL, Scheuer W, Lanzendorfer M, Honold K, Ambrosius D, Norley S, Kurth R. Interleukin-16 stimulates the expression and production of pro-inflammatory cytokines by human monocytes. Immunology. 2000;100(1):63–9.CrossRef
9.
Zurück zum Zitat Yang H, Han Y, Wu L, Wu C. Diagnostic and prognostic value of serum interleukin16 in patients with gastric cancer. Mol Med Rep. 2017;16(6):9143–8.CrossRef Yang H, Han Y, Wu L, Wu C. Diagnostic and prognostic value of serum interleukin16 in patients with gastric cancer. Mol Med Rep. 2017;16(6):9143–8.CrossRef
10.
Zurück zum Zitat Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human disease. N Engl J Med. 2000;342(18):1350–8.CrossRef Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human disease. N Engl J Med. 2000;342(18):1350–8.CrossRef
11.
Zurück zum Zitat Ishimoto T, Miyake K, Nandi T, Yashiro M, Onishi N, Huang KK, Lin SJ, Kalpana R, Tay ST, Suzuki Y, et al. Activation of transforming growth factor beta 1 signaling in gastric cancer-associated fibroblasts increases their motility, via expression of rhomboid 5 homolog 2, and ability to induce invasiveness of gastric cancer cells. Gastroenterology. 2017;153(1):191 e116–204 e116.CrossRef Ishimoto T, Miyake K, Nandi T, Yashiro M, Onishi N, Huang KK, Lin SJ, Kalpana R, Tay ST, Suzuki Y, et al. Activation of transforming growth factor beta 1 signaling in gastric cancer-associated fibroblasts increases their motility, via expression of rhomboid 5 homolog 2, and ability to induce invasiveness of gastric cancer cells. Gastroenterology. 2017;153(1):191 e116–204 e116.CrossRef
12.
Zurück zum Zitat Ota M, Horiguchi M, Fang V, Shibahara K, Kadota K, Loomis C, Cammer M, Rifkin DB. Genetic suppression of inflammation blocks the tumor-promoting effects of TGF-beta in gastric tissue. Can Res. 2014;74(9):2642–51.CrossRef Ota M, Horiguchi M, Fang V, Shibahara K, Kadota K, Loomis C, Cammer M, Rifkin DB. Genetic suppression of inflammation blocks the tumor-promoting effects of TGF-beta in gastric tissue. Can Res. 2014;74(9):2642–51.CrossRef
13.
Zurück zum Zitat Tas F, Yasasever CT, Karabulut S, Tastekin D, Duranyildiz D. Serum transforming growth factor-beta1 levels may have predictive and prognostic roles in patients with gastric cancer. Tumour Biol. 2015;36(3):2097–103.CrossRef Tas F, Yasasever CT, Karabulut S, Tastekin D, Duranyildiz D. Serum transforming growth factor-beta1 levels may have predictive and prognostic roles in patients with gastric cancer. Tumour Biol. 2015;36(3):2097–103.CrossRef
14.
Zurück zum Zitat Hu WQ, Wang LW, Yuan JP, Yan SG, Li JD, Zhao HL, Peng CW, Yang GF, Li Y. High expression of transform growth factor beta 1 in gastric cancer confers worse outcome: results of a cohort study on 184 patients. Hepatogastroenterology. 2014;61(129):245–50.PubMed Hu WQ, Wang LW, Yuan JP, Yan SG, Li JD, Zhao HL, Peng CW, Yang GF, Li Y. High expression of transform growth factor beta 1 in gastric cancer confers worse outcome: results of a cohort study on 184 patients. Hepatogastroenterology. 2014;61(129):245–50.PubMed
15.
Zurück zum Zitat Chen J, Miao L, Jin G, Ren C, Ke Q, Qian Y, Dong M, Li H, Zhang Q, Ding Y, et al. TGFBR1 tagging SNPs and gastric cancer susceptibility: a two-stage case-control study in Chinese population. Mol Carcinog. 2014;53(2):109–16.CrossRef Chen J, Miao L, Jin G, Ren C, Ke Q, Qian Y, Dong M, Li H, Zhang Q, Ding Y, et al. TGFBR1 tagging SNPs and gastric cancer susceptibility: a two-stage case-control study in Chinese population. Mol Carcinog. 2014;53(2):109–16.CrossRef
16.
Zurück zum Zitat Chang WW, Zhang L, Su H, Yao YS. An updated meta-analysis of transforming growth factor-beta1 gene: three polymorphisms with gastric cancer. Tumour Biol. 2014;35(4):2837–44.CrossRef Chang WW, Zhang L, Su H, Yao YS. An updated meta-analysis of transforming growth factor-beta1 gene: three polymorphisms with gastric cancer. Tumour Biol. 2014;35(4):2837–44.CrossRef
17.
Zurück zum Zitat Guo W, Dong Z, Guo Y, Chen Z, Yang Z, Kuang G. Association of polymorphisms in transforming growth factor-beta receptors with susceptibility to gastric cardia adenocarcinoma. Mol Biol Rep. 2012;39(4):4301–9.CrossRef Guo W, Dong Z, Guo Y, Chen Z, Yang Z, Kuang G. Association of polymorphisms in transforming growth factor-beta receptors with susceptibility to gastric cardia adenocarcinoma. Mol Biol Rep. 2012;39(4):4301–9.CrossRef
18.
Zurück zum Zitat Kutikhin AG. Impact of Toll-like receptor 4 polymorphisms on risk of cancer. Hum Immunol. 2011;72(2):193–206.CrossRef Kutikhin AG. Impact of Toll-like receptor 4 polymorphisms on risk of cancer. Hum Immunol. 2011;72(2):193–206.CrossRef
19.
Zurück zum Zitat Stubljar D, Jeverica S, Jukic T, Skvarc M, Pintar T, Tepes B, Kavalar R, Stabuc B, Peterlin B, Ihan A. The influence of cytokine gene polymorphisms on the risk of developing gastric cancer in patients with Helicobacter pylori infection. Radiol Oncol. 2015;49(3):256–64.CrossRef Stubljar D, Jeverica S, Jukic T, Skvarc M, Pintar T, Tepes B, Kavalar R, Stabuc B, Peterlin B, Ihan A. The influence of cytokine gene polymorphisms on the risk of developing gastric cancer in patients with Helicobacter pylori infection. Radiol Oncol. 2015;49(3):256–64.CrossRef
20.
Zurück zum Zitat Qadri Q, Rasool R, Afroze D, Naqash S, Gulzar GM, Yousuf A, Siddiqi MA, Shah ZA. Study of TLR4 and IL-8 gene polymorphisms in H. pylori-induced inflammation in gastric cancer in an ethnic Kashmiri population. Immunol Invest. 2014;43(4):324–36.CrossRef Qadri Q, Rasool R, Afroze D, Naqash S, Gulzar GM, Yousuf A, Siddiqi MA, Shah ZA. Study of TLR4 and IL-8 gene polymorphisms in H. pylori-induced inflammation in gastric cancer in an ethnic Kashmiri population. Immunol Invest. 2014;43(4):324–36.CrossRef
21.
Zurück zum Zitat Santini D, Angeletti S, Ruzzo A, Dicuonzo G, Galluzzo S, Vincenzi B, Calvieri A, Pizzagalli F, Graziano N, Ferraro E, 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.CrossRef Santini D, Angeletti S, Ruzzo A, Dicuonzo G, Galluzzo S, Vincenzi B, Calvieri A, Pizzagalli F, Graziano N, Ferraro E, 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.CrossRef
22.
Zurück zum Zitat He BS, Sun HL, Xu T, Pan YQ, Lin K, Gao TY, Zhang ZY, Wang SK. Association of genetic polymorphisms in the LncRNAs with gastric cancer risk in a chinese population. J Cancer. 2017;8(4):531–6.CrossRef He BS, Sun HL, Xu T, Pan YQ, Lin K, Gao TY, Zhang ZY, Wang SK. Association of genetic polymorphisms in the LncRNAs with gastric cancer risk in a chinese population. J Cancer. 2017;8(4):531–6.CrossRef
23.
Zurück zum Zitat Slattery ML, Trivellas A, Pellatt AJ, Mullany LE, Stevens JR, Wolff RK, Herrick JS. Genetic variants in the TGFbeta-signaling pathway influence expression of miRNAs in colon and rectal normal mucosa and tumor tissue. Oncotarget. 2017;8(10):16765–83.CrossRef Slattery ML, Trivellas A, Pellatt AJ, Mullany LE, Stevens JR, Wolff RK, Herrick JS. Genetic variants in the TGFbeta-signaling pathway influence expression of miRNAs in colon and rectal normal mucosa and tumor tissue. Oncotarget. 2017;8(10):16765–83.CrossRef
24.
Zurück zum Zitat Nicoloso MS, Sun H, Spizzo R, Kim H, Wickramasinghe P, Shimizu M, Wojcik SE, Ferdin J, Kunej T, Xiao L, et al. Single-nucleotide polymorphisms inside microRNA target sites influence tumor susceptibility. Can Res. 2010;70(7):2789–98.CrossRef Nicoloso MS, Sun H, Spizzo R, Kim H, Wickramasinghe P, Shimizu M, Wojcik SE, Ferdin J, Kunej T, Xiao L, et al. Single-nucleotide polymorphisms inside microRNA target sites influence tumor susceptibility. Can Res. 2010;70(7):2789–98.CrossRef
25.
Zurück zum Zitat Valle L, Serena-Acedo T, Liyanarachchi S, Hampel H, Comeras I, Li Z, Zeng Q, Zhang HT, Pennison MJ, Sadim M, et al. Germline allele-specific expression of TGFBR1 confers an increased risk of colorectal cancer. Science. 2008;321(5894):1361–5.CrossRef Valle L, Serena-Acedo T, Liyanarachchi S, Hampel H, Comeras I, Li Z, Zeng Q, Zhang HT, Pennison MJ, Sadim M, et al. Germline allele-specific expression of TGFBR1 confers an increased risk of colorectal cancer. Science. 2008;321(5894):1361–5.CrossRef
26.
Zurück zum Zitat Castano-Rodriguez N, Kaakoush NO, Pardo AL, Goh KL, Fock KM, Mitchell HM. Genetic polymorphisms in the Toll-like receptor signalling pathway in Helicobacter pylori infection and related gastric cancer. Hum Immunol. 2014;75(8):808–15.CrossRef Castano-Rodriguez N, Kaakoush NO, Pardo AL, Goh KL, Fock KM, Mitchell HM. Genetic polymorphisms in the Toll-like receptor signalling pathway in Helicobacter pylori infection and related gastric cancer. Hum Immunol. 2014;75(8):808–15.CrossRef
27.
Zurück zum Zitat Huang L, Yuan K, Liu J, Ren X, Dong X, Tian W, Jia Y. Polymorphisms of the TLR4 gene and risk of gastric cancer. Gene. 2014;537(1):46–50.CrossRef Huang L, Yuan K, Liu J, Ren X, Dong X, Tian W, Jia Y. Polymorphisms of the TLR4 gene and risk of gastric cancer. Gene. 2014;537(1):46–50.CrossRef
28.
Zurück zum Zitat Yamaguchi T, Fushida S, Yamamoto Y, Tsukada T, Kinoshita J, Oyama K, Miyashita T, Tajima H, Ninomiya I, Munesue S, et al. Low-dose paclitaxel suppresses the induction of M2 macrophages in gastric cancer. Oncol Rep. 2017;37(6):3341–50.CrossRef Yamaguchi T, Fushida S, Yamamoto Y, Tsukada T, Kinoshita J, Oyama K, Miyashita T, Tajima H, Ninomiya I, Munesue S, et al. Low-dose paclitaxel suppresses the induction of M2 macrophages in gastric cancer. Oncol Rep. 2017;37(6):3341–50.CrossRef
29.
Zurück zum Zitat Li G, Wang Z, Ye J, Zhang X, Wu H, Peng J, Song W, Chen C, Cai S, He Y, et al. Uncontrolled inflammation induced by AEG-1 promotes gastric cancer and poor prognosis. Can Res. 2014;74(19):5541–52.CrossRef Li G, Wang Z, Ye J, Zhang X, Wu H, Peng J, Song W, Chen C, Cai S, He Y, et al. Uncontrolled inflammation induced by AEG-1 promotes gastric cancer and poor prognosis. Can Res. 2014;74(19):5541–52.CrossRef
30.
Zurück zum Zitat Zonneville J, Safina A, Truskinovsky AM, Arteaga CL, Bakin AV. TGF-beta signaling promotes tumor vasculature by enhancing the pericyte-endothelium association. BMC cancer. 2018;18(1):670.CrossRef Zonneville J, Safina A, Truskinovsky AM, Arteaga CL, Bakin AV. TGF-beta signaling promotes tumor vasculature by enhancing the pericyte-endothelium association. BMC cancer. 2018;18(1):670.CrossRef
Metadaten
Titel
Polymorphisms of TGFBR1, TLR4 are associated with prognosis of gastric cancer in a Chinese population
verfasst von
Bangshun He
Tao Xu
Bei Pan
Yuqin Pan
Xuhong Wang
Jingwu Dong
Huiling Sun
Xueni Xu
Xiangxiang Liu
Shukui Wang
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2018
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-018-0682-0

Weitere Artikel der Ausgabe 1/2018

Cancer Cell International 1/2018 Zur Ausgabe

Update Onkologie

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