Oncotarget

Research Papers:

Association between SNPs in microRNA machinery genes and gastric cancer susceptibility, invasion, and metastasis in Chinese Han population

PDF |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:86435-86446. https://doi.org/10.18632/oncotarget.21199

Metrics: PDF 1183 views  |   HTML 2302 views  |   ?  

Xingbo Song, Huiyu Zhong, Qian Wu, Minjin Wang, Juan Zhou, Yi Zhou, Xiaojun Lu and Binwu Ying _

Abstract

Xingbo Song1,*, Huiyu Zhong1,*, Qian Wu1,*, Minjin Wang1, Juan Zhou1, Yi Zhou1, Xiaojun Lu1 and Binwu Ying1

1Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China

*These authors have contributed equally to this work

Correspondence to:

Binwu Ying, email: [email protected]

Keywords: microRNA, single nucleotide polymorphism, gastric cancer, tumor invasion and metastasis

Received: June 16, 2017     Accepted: July 30, 2017     Published: September 23, 2017

ABSTRACT

Objective: The present study investigates the influence of genetic variants in miRNA machinery genes (DROSHA, DICER, AGO1, and GEMIN4) on gastric cancer in Chinese Han population, further revealing the genetic mechanisms of gastric cancer occurrence and development.

Methods: Genotyping of single nucleotide polymorphisms (SNPs) was performed in 628 patients with GC and 502 frequency-matched (age and gender) controls by the high resolution melting (HRM) method.

Results: The SNPs rs3742330 (DICER) and rs7813 (GEMIN4) were associated with susceptibility to gastric cancer (P = 0.002 and 0.010, respectively). Stratified analysis showed that the G allele of rs3742330 and genotype TT as well as T allele of rs7813 were associated with a later stage of gastric cancer (P=0.027, 0.032 and 0.018, respectively). Furthermore, the genotype TT and T allele of rs7813 appeared to be associated with a higher level of lymphatic metastasis of gastric cancer (P=0.021 and 0.030, respectively), while the genotype AA and A allele of rs636832 (AGO1) were correlated with a lower level of lymphatic metastasis of gastric cancer (P=0.016 and 0.041, respectively). There was no significant association between rs10719 (DROSHA) and gastric cancer.

Conclusion: The present data demonstrated that genetic variants in miRNA machinery genes had a significant association with GC susceptibility (DICER and GEMIN4) and malignant behavior such as tumor stage (DICER and GEMIN4) and lymphatic metastasis of GC (GEMIN4 and AGO1) in Chinese Han population.


INTRODUCTION

Gastric cancer (GC) is the most common malignant tumor of the digestive system and the third leading cause of cancer mortality worldwide [1]. The development and progression of GC are affected by the interaction between environmental factors and individual genetic factors. Factors including Helicobacter pylori infection, salted food, drinking, smoking and so on were proved by classic epidemiological studies to be the main risk factors of GC [2], but there are differences in GC susceptibility and tumor progression between individuals. These differences are associated with gene polymorphisms.

MicroRNAs (miRNAs) are small single-stranded RNA molecules of about 21-23 nucleotides (nt) in length. Recently, miRNA is widely recognized as regulators of gene expression and regulate about 30% genes in humans [36]. The process of miRNA synthesis begins within the nucleus where RNA polymerase II converts miRNA into pri-miRNA. The pri-miRNA is then processed into a precursor of ~70 nt in length with a hairpin structure by a DNA endonuclease enzyme named DROSHA (RNase III) as well as its cofactor DGCR8; this precursor is called pre-miRNA. At the same time, DROSHA and DGCR8 protein constitute a microprocessor complex in the formation of pre-miRNA. Next, Exportin-5/Ran-GTP complex transfers pre-miRNA to the cytoplasm, and pre-miRNA is then cut into miRNA duplexes (about 20 bp) by the TAR RNA binding protein (TRBP)-related DICER [711]. One strand of the miRNA duplexes integrates into miRNA-induced silencing complex (miRISC) and becomes mature miRNA. The miRISC contains proteins including AGO1-4, GEMIN3, and GEMIN4 that participate in mRNA inhibition or shearing of target mRNA [1215]. Therefore, genetic polymorphisms in microRNA machinery genes could lead to abnormal expression of miRNAs and in turn affect the expression level of target genes, thus becoming the risk factor of disease such as tumor. Currently, there is little research exploring the influence of single nucleotide polymorphisms (SNPs) in miRNA machinery genes on GC susceptibility, invasion, and metastasis.

This research chose SNP loci in classical miRNA machinery genes (rs3742330 in DICER, rs3744741 and rs7813 in GEMIN4, rs10719 in DROSHA, and rs636832 in AGO1) by using a candidate gene-based approach to genetically explore the effect of variants in miRNA machinery genes on GC susceptibility, invasion, and metastasis in Chinese Han population. In addition, the findings of this study might provide the basis for further revealing the specific mechanisms by which genetic variants of these genes participate in the occurrence and development of GC. Additional in-silico studies were used to assess the possible functional significance and miRNA-binding of the positive polymorphisms.

RESULTS

Demographic characteristics of the study participants

The demographic characteristics of 628 cases and 502 controls are presented in Table 1. The average age and sex had no significant differences between the patient group and control group (P=0.727, 0.577 respectively) and for smoking status and drinking status (P=0.297, 0.631 respectively). All the participants were from Chinese population.

Table 1: Basic demographic data of subjects and clinical characteristics of GC cases

Parameters

Case

Control

P

n

Frequencies (%)

n

Frequencies (%)

Age (year, mean ± SD)

56.5±12.1

56.2±12.2

0.727

Gender

 Male

418

66.6

323

64.3

0.577

 Female

210

33.4

179

35.7

Smoking Status

 Non-smokers

365

58.1

297

59.2

0.297

 Former Smokers

139

22.1

125

24.9

 Current Smokers

124

19.7

80

15.9

Drinking status

 Non-drinker

437

69.6

348

69.3

0.631

 Light Drinkers

93

14.8

66

13.2

 Heavy Drinkers

98

15.6

88

17.5

Tumor size (diameter)

 <5 cm

207

33.0

 5-10 cm

204

32.5

 ≥10 cm

56

8.9

 N.A.

161

25.6

Tumor stages

 1a

64

10.2

 1b

36

5.7

 2a

41

6.5

 2b

72

11.5

 3a

55

8.8

 3b

71

11.3

 3c

109

17.4

 4

180

28.7

Degree of differentiation

 Low

433

68.9

 Medium

185

29.5

 High

10

1.6

Lymphatic metastasis

 0

151

24.0

 1

103

16.4

 2

109

17.4

 3a

124

19.7

 3b

61

9.7

 N.A.

80

12.7

N.A. data not available.

The relationship between SNPs in miRNA machinery genes and GC susceptibility

Genotyping of five SNPs was successfully completed for the cases and controls. Genotype distribution of rs3744741 in patient group was not in accordance with the Hardy-Weinberg equilibrium (HWE) (χ2=10.18, P=0.001), while the other 4 SNPs in either patient or control groups met HWE (P > 0.05 for all loci). Thus, the SNP rs3744741 was excluded from further analysis (data not shown). Table 2 shows the genotype distributions and allele frequencies of the 4 SNPs in miRNA machinery genes between cases and controls.

Table 2: Comparisons of gene polymorphisms between the case and control groups

SNP

Cases

Controls

OR (95% C.I.)*

P*

N

%

N

%

rs 3742330

Genotype

 AA

273

43.5

177

35.3

1.00 (Reference)

 AG

284

45.2

246

49.0

0.75 (0.58-0.97)

0.026

 GG

71

11.3

79

15.7

0.58 (0.39-0.86)

0.004

Allele

 A

830

66.1

600

59.8

1.00 (Reference)

 G

426

33.9

404

40.2

0.76 (0.64-0.91)

0.002

 rs7813

Genotype

 TT

261

41.6

241

48.0

1.00 (Reference)

 CT

294

46.8

222

44.2

1.22 (0.95-1.58)

0.110

 CC

73

11.6

39

7.8

1.73 (1.11-2.71)

0.011

Allele

 T

816

65.0

704

70.1

1.00 (Reference)

 C

440

35.0

300

29.9

1.27 (1.05-1.52)

0.010

 rs10719

Genotype

 TT

314

50.0

248

49.4

1.00 (Reference)

 CT

257

40.9

205

40.8

0.99 (0.77-1.28)

0.938

 CC

57

9.1

49

9.8

0.92 (0.59-1.42)

0.690

Allele

 T

885

70.5

701

69.8

1.00 (Reference)

 C

371

29.5

303

30.2

0.97 (0.81-1.17)

0.741

 rs 636832

Genotype

 AA

321

51.1

268

53.4

1.00 (Reference)

 AG

261

41.6

198

39.4

1.10 (0.85-1.42)

0.445

 GG

46

7.3

36

7.2

1.07 (0.65-1.74)

0.785

Allele

 A

903

71.9

734

73.1

1.00 (Reference)

 G

353

28.1

270

26.9

1.06 (0.88-1.29)

0.521

* Adjusted by sex, age, smoking status, and drinking status.

As shown in Table 2, the minor allele (G allele) frequency of rs3742330 was 33.9% in cases and 40.2% in controls and was significantly different (OR= 0.76, 95% CI= 0.64-0.91), the p-value was 0.002; this indicated that the G allele could be a protective element for GC susceptibility. As expected, genotype GG and AG of rs3742330 had a significantly decreased risk of GC compared with AA genotype (P=0.004, OR= 0.58, 95% CI= 0.39-0.86 for GG versus AA, and P=0.026, OR= 0.75, 95% CI= 0.58-0.97 for AG versus AA).

Conversely, subjects carrying a CC genotype in rs7813 showed a significant increase in risk for GC than those carrying the TT genotype (P=0.011, OR = 1.73, 95% CI = 1.11-2.71), and it was suggested that the C allele of rs7813 may be associated with a higher risk of GC than T allele (P=0.010, OR=1.27, 95%CI=1.05-1.52). However, no significant differences in genotype distributions or allelic frequencies of rs10719 and rs636832 were demonstrated between the cases and controls. All the above data were adjusted by sex, age, smoking status, and drinking status.

Stratified analysis for the SNP genotypes and clinicopathologic characteristics of GC patients

To demonstrate the association between SNP genotypes and clinicopathologic characteristics of GC, the cases were stratified into subgroups according to tumor size, tumor stages, degree of differentiation, and lymphatic metastasis. The results for each SNP are summarized in Tables 3-1, 3-2, 3-3 and 3-4, respectively.

Table 3-1: Stratified analysis for the association between rs3742330 and GC clinical characteristics

Clinical characteristics

Genotype

OR (95% C.I.)*

P

Allele

OR (95% C.I.)

P

AA

AG

GG

A

G

Tumor size

 <5 cm

100

86

21

1.00 (Reference)

286

128

1.00 (Reference)

 5-10 cm

83

94

27

0.73 (0.49-1.11)

0.120

260

148

0.79 (0.58-1.06)

0.104

 ≥10 cm

26

26

4

0.93 (0.49-1.75)

0.803

78

34

1.03 (0.64-1.66)

0.909

Tumor stages

 1a

29

30

5

1.00 (Reference)

88

40

1.00 (Reference)

 1b

14

18

4

0.77 (0.31-1.91)

0.533

46

26

0.80 (0.42-1.55)

0.483

 2a

13

18

10

0.56 (0.23-1.37)

0.165

44

38

0.53 (0.28-0.97)

0.027

 2b

38

29

5

1.35 (0.65-2.81)

0.385

74

39

0.86 (0.49-1.53)

0.590

 3a

25

24

6

1.01 (0.46-2.21)

0.988

74

36

0.93 (0.52-1.67)

0.807

 3b

29

32

10

0.83 (0.40-1.75)

0.601

90

52

0.79 (0.46-1.35)

0.353

 3c

46

51

12

0.88 (0.45-1.72)

0.690

143

75

0.87 (0.53-1.42)

0.548

 4

79

80

21

0.94 (0.51-1.74)

0.844

238

122

0.89 (0.56-1.40)

0.586

Degree of differentiation

 Low

187

198

47

1.00 (Reference)

572

292

1.00 (Reference)

 Medium

82

82

21

1.04 (0.73-1.50)

0.812

246

124

1.01 (0.78-1.32)

0.923

 High

4

4

2

12

8

Lymphatic metastasis

 0

66

72

13

1.00 (Reference)

204

98

1.00 (Reference)

 1

42

42

19

0.89 (0.52-1.52)

0.643

126

80

0.76 (0.51-1.11)

0.139

 2

49

51

9

1.05 (0.62-1.78)

0.842

149

69

1.04 (0.70-1.53)

0.847

 3a

62

50

12

1.29 (0.78-2.13)

0.298

174

74

1.13 (0.77-1.65)

0.511

 3b

19

33

9

0.58 (0.30-1.14)

0.091

71

51

0.67 (0.42-1.06)

0.068

Table 3-2: Stratified analysis for the association between rs7813 and GC clinical characteristics

Clinical characteristics

Genotype

OR (95% C.I.)*

P

Allele

OR (95% C.I.)

P

TT

CT

CC

T

C

Tumor size

 <5 cm

81

105

21

1.00 (Reference)

267

147

1.00 (Reference)

 5-10 cm

88

94

22

1.18 (0.78-1.78)

0.409

270

138

1.08 (0.80-1.45)

0.612

 ≥10 cm

26

26

4

1.35 (0.71-2.55)

0.324

78

34

1.26 (0.79-2.03)

0.309

Tumor stages

 1a

20

37

7

1.00 (Reference)

77

51

1.00 (Reference)

 1b

15

13

8

1.57 (0.62-4.00)

0.295

43

29

0.98 (0.52-1.85)

0.952

 2a

13

24

4

1.02 (0.40-2.58)

0.961

50

32

1.03 (0.56-1.90)

0.906

 2b

33

30

9

1.86 (0.87-4.00)

0.082

96

48

1.32 (0.78-2.24)

0.265

 3a

20

28

7

1.26 (0.55-2.89)

0.556

68

42

1.07 (0.61-1.87)

0.793

 3b

24

38

9

1.12 (0.51-2.46)

0.601

86

56

1.02 (0.61-1.71)

0.946

 3c

54

50

5

2.16 (1.08-4.36)

0.019

158

60

1.74 (1.07-2.84)

0.018

 4

84

74

22

1.92 (1.01-3.69)

0.032

242

118

1.36 (0.88-2.10)

0.149

Degree of differentiation

 Low

178

203

52

1.00 (Reference)

559

307

1.00 (Reference)

 Medium

79

88

18

1.07 (0.74-1.54)

0.713

246

124

1.09 (0.84-1.42)

0.513

 High

4

3

3

11

9

Lymphatic metastasis

 0

50

80

21

1.00 (Reference)

180

122

1.00 (Reference)

 1

48

45

10

1.76 (1.02-3.05)

0.030

141

65

1.47 (1.00-2.17)

0.042

 2

43

52

14

1.36 (0.76-2.27)

0.293

138

80

1.17 (0.80-1.70)

0.393

 3a

58

54

12

1.78 (1.06-2.98)

0.021

170

78

1.48 (1.02-2.14)

0.030

 3b

28

28

5

1.71 (0.89-3.29)

0.080

84

38

1.50 (0.94-2.40)

0.075

Table 3-3: Stratified analysis for the association between rs10719 and GC clinical characteristics

Clinical characteristics

Genotype

OR (95% C.I.)*

P

Allele

OR (95% C.I.)

P

TT

CT

CC

T

C

Tumor size

 <5 cm

106

83

18

1.00 (Reference)

295

119

1.00 (Reference)

 5-10 cm

104

79

21

0.99 (0.66-1.49)

0.963

287

121

0.96 (0.70-1.31)

0.773

 ≥10 cm

30

21

5

1.10 (0.58-2.07)

0.753

81

31

1.05 (0.65-1.73)

0.825

Tumor stages

 1a

26

32

6

1.00 (Reference)

84

44

1.00 (Reference)

 1b

20

14

2

1.83 (0.74-4.54)

0.150

54

18

1.57 (0.79-3.16)

0.169

 2a

23

14

4

1.87 (0.78-4.47)

0.121

60

22

1.43 (0.74-2.75)

0.251

 2b

34

33

5

1.31 (0.63-2.74)

0.439

101

43

1.23 (0.72-2.12)

0.426

 3a

30

19

6

1.75 (0.79-3.88)

0.129

79

31

1.33 (0.74-2.41)

0.305

 3b

37

24

10

1.59 (0.76-3.34)

0.182

98

44

1.17 (0.68-2.00)

0.553

 3c

59

41

9

1.72 (0.88-3.38)

0.086

159

59

1.41 (0.86-2.32)

0.151

 4

85

80

15

1.31 (0.73-2.43)

0.363

250

110

1.19 (0.76-1.87)

0.425

Degree of differentiation

 Low

215

179

39

1.00 (Reference)

609

257

1.00 (Reference)

 Medium

94

74

17

1.05 (0.73-1.50)

0.792

262

108

1.02 (0.73-1.35)

0.863

 High

5

4

1

14

6

Lymphatic metastasis

 0

69

69

13

1.00 (Reference)

207

95

1.00 (Reference)

 1

55

39

9

1.36 (0.80-2.32)

0.228

149

57

1.20 (0.80-1.81)

0.360

 2

60

39

10

1.46 (0.86-2.46)

0.137

159

59

1.24 (0.83-1.85)

0.279

 3a

67

46

11

1.40 (0.80-2.42)

0.169

180

68

1.21 (0.83-1.79)

0.302

 3b

26

26

9

0.88 (0.46-1.68)

0.684

78

44

0.81 (0.51-1.44)

0.360

Table 3-4: Stratified analysis for the association between rs636832 and GC clinical characteristics

Clinical characteristics

Genotype

OR (95% C.I.)*

P

Allele

OR (95% C.I.)

P

AA

AG

GG

A

G

Tumor size

 <5 cm

96

94

17

1.00 (Reference)

286

128

1.00 (Reference)

 5-10 cm

106

86

12

1.25 (0.83-1.88)

0.258

298

110

1.21 (0.89-1.66)

0.211

 ≥10 cm

27

24

5

1.08 (0.57-2.03)

0.807

78

34

1.03 (0.64-1.66)

0.909

Tumor stages

 1a

35

26

3

1.00 (Reference)

96

32

1.00 (Reference)

 1b

23

12

1

1.47 (0.58-3.70)

0.371

58

14

1.38 (0.65-2.98)

0.370

 2a

25

12

4

1.29 (0.54-3.11)

0.525

62

20

1.03 (0.52-2.07)

0.920

 2b

36

30

6

1.83 (0.40-1.72)

0.585

102

42

0.81 (0.46-1.43)

0.441

 3a

34

20

1

1.34 (0.60-2.99)

0.432

88

22

1.33 (0.69-2.58)

0.358

 3b

31

36

4

0.64 (0.31-1.34)

0.201

98

44

0.74 (0.42-1.31)

0.275

 3c

46

51

12

0.60 (0.31-1.18)

0.112

143

75

0.64 (0.38-1.06)

0.068

 4

91

74

15

0.85 (0.46-1.56)

0.570

256

104

0.82 (0.50-1.33)

0.399

Degree of differentiation

 Low

211

191

31

1.00 (Reference)

613

253

1.00 (Reference)

 Medium

103

67

15

1.32 (0.92-1.90)

0.114

273

97

1.16 (0.88-1.54)

0.284

 High

7

3

0

17

3

Lymphatic metastasis

 0

88

56

7

1.00 (Reference)

232

70

1.00 (Reference)

 1

59

36

8

0.96 (0.56-1.65)

0.875

154

52

0.89 (0.58-1.38)

0.593

 2

47

56

6

0.54 (0.32-0.92)

0.016

150

68

0.67 (0.44-1.00)

0.041

 3a

61

47

16

0.69 (0.42-1.15)

0.132

169

79

0.65 (0.43-0.96)

0.023

 3b

27

31

3

0.57 (0.30-1.08)

0.064

85

37

0.69 (0.42-1.14)

0.125

As shown in Table 3-1, the A allele of rs3742330 may decrease the risk of GC in stage 1b rather than 1a (P=0.027, OR=0.53, 95%CI=0.28-0.97). However, there was no significant difference found in the frequency of AA genotype. According to Table 3-2, individuals carrying TT genotype and T allele of rs7813 had an increased risk of GC in tumor stage 3c than stage 1a (P=0.019, OR=2.16, 95%CI=1.08-4.36; P=0.018, OR=1.74, 95%CI=1.07-2.84, respectively). In terms of the data, the TT genotype of rs7813 also increased the risk of GC in stage 4 than stage 1a (P=0.032, OR=1.92, 95%CI=1.01-3.69). For GC invasion and metastasis, the data in Table 3-2 indicated that the TT genotype and the T allele of rs7813 had a higher risk of lymphatic metastasis stage 1 or 3a than stage 0 (P=0.030, OR=1.76, 95%CI=1.02-3.05; P=0.042, OR=1.47, 95%CI=1.00-2.17 and P=0.021, OR=1.78, 95%CI=1.06-2.98; P=0.030, OR=1.48, 95%CI=1.02-2.14). With regard to rs636832, as shown in Table 3-4, it is suggested that the AA genotype and A allele had an association with a lower risk of lymphatic metastasis stage 2 compared with stage 0 (P=0.016, OR=0.54, 95%CI=0.32-0.92 and P=0.041, OR=0.67, 95%CI=0.44-1.00 respectively), similar to the A allele which had a lower risk of lymphatic metastasis stage 3a (P=0.023, OR=0.65, 95%CI=0.43-0.96). Stratified analysis of rs10719 showed no significant differences in tumor size, tumor stages, degree of differentiation, or lymphatic metastasis of GC (Table 3-3). To demonstrate the mechanisms of these associations, further study is urgently needed.

In-silico analysis of microRNA-binding and function prediction

As for rs3742330, computational modeling suggested that this polymorphism was located in the potential target sequence of hsa-miR-632 in DICER 3'UTR region (Supplementary Figure 1). The G allele could reduce the affinity of microRNA-mRNA binding by disrupting the local structure of DICER mRNA, possibly leading an increased DICER expression. In addition rs7813(C>T, R1033C) was a missense variant in exon region of GEMIN4, which could alter the structure of GEMIN4 protein by turning Arginine into Cysteine (Supplementary Figure 2), thus reducing GEMIN4 expression. There was no function results for rs636832 obtained from the software.

DISCUSSION

Individual genetic factors play an important role in susceptibility and progression of GC. miRNA is a small single-stranded RNA of 21-23 nt in length and is widely recognized as regulators of gene expression. miRNAs participate in a variety of important biological processes including cell cycle, cell differentiation, and cell proliferation and apoptosis [16]. Previous studies have confirmed that miRNAs play an important role in a wide variety of tumor biological behaviors such as tumor cell proliferation and apoptosis. Clinically, there is abnormal expression of different levels of miRNAs in cancer patients, indicating that miRNA has a large influence on the development of tumor [1719]. Ahn DH [20] chose four SNPs in miRNA and analyzed the genotypes and allele frequencies of these SNPs in 461 Korean GC patients. The study found that these polymorphisms in miRNA were associated with the risk of GC; in addition, genotypes rs2292832 and rs3746444 were associated with survival rates of GC patients. Xiong XD [21] found that rs895819 in pre-miR-27a could alter the expression level of the miRNA and thus was correlated with the incidence of cervical cancer. A previous study showed correlations between genetic variants in miRNA and gastric lesions or even GC. One study investigated rs112310158 in hsa-miR-449a in Chinese population and revealed that GG genotype of rs112310158 had a higher risk of GC than other genotypes [22]. Jin X [23] analyzed genotypes of SNPs in mir-421 and found it to be significantly associated with GC susceptibility, lymphatic metastasis, and prognosis.

The expression level and regulatory function of miRNA depend on the orderly division of function of genes in miRNA biogenesis pathways. Proteins such as GEMIN4, AGO1, DROSHA, DICER, and their complex regulating miRNA biogenesis pathways are key components of miRNA maturation, transfer, and function. Proper cooperation of these proteins enables the expression of genes that regulate miRNA. Genetic variants in miRNA machinery genes could affect the maturation and regulatory function of miRNA by influencing the transcription ability of genes (UTR region) or protein function (exon region), thus manifesting as a change in tumor susceptibility and malignant behavior [8, 24]. Recent studies have already revealed a relationship between SNPs in miRNA machinery genes and several tumors including GC [13, 2527], and investigation of variants in miRNA machinery genes could clarify the mechanism of the occurrence and development of GC and provide new basis for its clinical diagnosis and management. Our group speculates that genetic polymorphisms of the important miRNA machinery genes (DICER, GEMIN4, DROSHA and AGO1) could play a role in GC susceptibility and malignant behavior by affecting the maturity and functioning of miRNA.

This study analyzed the genotype and allele frequencies of four SNPs in miRNA machinery genes (GEMIN4, DROSHA, DICER and AGO1) in GC patients and healthy controls in Chinese Han population to investigate whether the genetic polymorphisms in these genes can affect the susceptibility, invasion and metastasis of GC. We found that among the chosen SNPs, the distribution of genotype and allele frequencies of rs3742330 in DICER and rs7813 in GEMIN4 were significantly different between GC patients and healthy controls, indicating that genetic variants in DICER and GEMIN4 were correlated with GC susceptibility in Chinese Han population. Tchernitsa O [28] analyzed the expression of DICER in adjacent normal and tumor samples of patients with GC by using immunohistochemistry and detected an elevated DICER level in GC tissues. However, another study using the same sample type and analytical method demonstrated a down regulation of DICER in GC tissues in both mRNA and protein levels [29]. There is no study demonstrating a definite association between GEMIN4 and GC. Xie Y [26] investigated SNPs in miRNA machinery genes including GEMIN4 (rs2740348) but found no significant correlation between this SNP in GEMIN4 and GC pathogenesis. Despite the controversial results reported, it is clear that DICER and GEMIN4 participated in the pathogenesis of tumors including GC, and polymorphisms in these genes could affect tumor susceptibility. Thus far, the influence of SNPs that we investigated in DICER and GEMIN4 on GC susceptibility had rarely been reported. Rs3742330 in DICER had been reported to be associated with the risk of larynx cancer in Polish population [30]. Another study in Korean population showed a significantly increased risk of colon cancer in individuals with AG genotype of rs3742330 [31]. The location of rs3742330 in the 3’-UTR region of DICER may potentially influence the stability and expression of DICER through changing the binding capacity of regulatory miRNAs [32, 33]. But the mechanism underlying how rs3742330 modified GC susceptibility remains unclear. Our group conducted the in-silico analysis and found that rs3742330 was located in the hsa-miR-632 potential target sequence in DICER 3'UTR region, which might probably upregulate the expression of DICER. Rs7813 in GEMIN4 was reported to be evidently associated with the risk of lung cancer [34], but another study showed no significant association of rs7813 with the risk of esophageal squamous cell carcinoma [35]. Our predicted analyses showed that rs7813 could alter the structure of GEMIN4 protein by turning Arginine into Cysteine and the alteration might reduce GEMIN4 expression. It was reported that rs7813 in GEMIN4 could induce Arg to Cys substitution at the 1033 amino acid position through the C to T transition [34], which could then affect the function of miRNAs. Our study found a correlation between the two polymorphisms in DICER and GEMIN4 and GC susceptibility, suggesting a predictive role of these SNPs in gastric carcinogenesis.

Furthermore we established a database of all the GC patients, including enormous clinical information such as tumor size, tumor stage, degree of differentiation, lymphatic metastasis and so on. Stratified analysis with all the clinical features revealed a notable correlation between rs3742330 (DICER) and rs7813 (GEMIN4) and the stage of GC, providing molecular markers of prognosis at an early stage. In addition, the TT genotype and the T allele of rs7813 (GEMIN4), and the AA genotype and A allele of rs636832 (AGO1) were related to lymphatic metastasis of GC. These three SNPs could be potential biomarkers for predicting the invasion and metastasis of GC. Previously, several researchers have reported the dysregulation and potential role of DICER, GEMIN4 and AGO1 in tumor progression, including GC. Down regulation of DICER has been reported to be highly correlated with tumor differentiation and lymph node invasion in GC tissues, while decrease of DICER was more common in GC cases with low tumor differentiation and lymph node metastasis [29]. Shi Z [36] further demonstrated the mechanism that DICER could process pre-miR-21 to mature miR-21, while the inhibitor of DICER (AC1MMYR2) blocked its ability for miRNA maturation and further suppressed proliferation, survival, and invasion in glioblastoma, breast cancer, and gastric cancer cells in vivo. According to an in vitro experiment, DEAD-box RNA helicase 6 (DDX6), which directly interacts with AGO1 in RNA-induced silencing complexes (RISC), was reported to down regulate miR-143/145 expression by prompting the degradation of its host gene product [37]. Thus far, no association has been found between GEMIN4 and GC progression. Consistent with our study, rs3742330 in DICER and rs7813 in GEMIN4 were found to participate in tumor progression. Mi Na Kim [38] demonstrated that rs3742330 was associated with the survival of hepatocellular carcinoma patients, while another study reported that the G allele of rs3742330 was associated with lower aggressiveness of prostate cancer [39]. Yang PW [40] showed a borderline significant association between rs7813 in GEMIN4 and the prognosis of esophageal squamous cell carcinoma (ESCC). In addition, AGO1 is located at chromosome 1p35-p34 and frequently lost in human malignant tumors, and rs636832 is located in the intron of AGO1, which might influence the conformation and function of proteins or the splicing of precursor miRNA [41], but no study reported the effect of rs636832 in AGO1 on tumor development, while current studies have not yet demonstrated a definite correlation between rs3742330 as well as rs7813 and GC invasion and metastasis. Our present study is the first to revealed an influence of the three SNPs in miRNA machinery genes on GC progression.

The results from this study demonstrated that genetic polymorphisms in miRNA machinery genes (DICER, GEMIN4 and AGO1) affected the susceptibility and the invasion and metastasis of GC in Chinese Han population, extremely probably by affecting maturing and functioning of relevant miRNAs. We confirmed in a relatively large sample size that these polymorphisms participated in the development of GC and its malignant behavior, genetically proving the essential roles of these genes in tumorigenesis and progression of tumor. Follow-up studies with larger sample size are required to further verify the results and design innovative experiments and functional verification to investigate the specific mechanism by which polymorphisms in these miRNA machinery genes influence the maturation of miRNA and then participate in the genesis and development of GC. The subsequent research could further reveal the molecular mechanism of GC and provide new molecular markers for GC diagnosis and treatment.

MATERIALS AND METHODS

Study populations

The study involved 628 cases and 502 controls. The cases were from West China Hospital outpatient or inpatients with GC between July 2010 and July 2016. The diagnosis of GC was based on both clinical criteria and pathological confirmation. The controls included unrelated healthy individuals screened from the physical examination center of West China Hospital of Sichuan University. All the controls had no significant history of disease. The controls were matched with the cases in the age and gender and came from the same region and same period as the cases. All participants provided informed consent to participate in the study, and this study was approved by the ethical committee of West China Hospital of Sichuan University.

Genomic DNA was extracted from the peripheral blood of the participants by using QIAamp® DNA Blood mini kit (Qiagen, Düsseldorf, Germany) following the manufacturer’s instructions. The samples were selected from patients with GC who had not been treated with chemotherapy but had been pathologically confirmed. Each sample used in the experiment had detailed clinical information and DNA met the requirements of concentration and purity.

SNP selection and genotyping

Based on the data from the International HapMap Project (http://www.hapmap.org), dbSNP (http://www.ncbi.nlm.nih.gov/projects/SNP/), and miRBase registry (http://microrna.sanger.ac.uk), we identified 20 potential polymorphisms in the miRNA biogenesis pathway (Supplementary Table 1) that met the criteria of minor allele frequency (MAF) > 0.01 in Chinese population. Thirty subjects including 15 healthy individuals and 15 patients with GC were randomly involved in SNP screening by high resolution melting (HRM). Finally, only five GC-associated SNPs with a high frequency (>0.1) of the minor allele were selected (rs3742330 in DICER, rs3744741 in GEMIN4, rs7813 in GEMIN4, rs10719 in DROSHA, and rs636832 in AGO1).

The isolated DNA was stored in a freezer at -80°C. Genotyping of the SNPs was performed by the HRM method. The data were analyzed using the LightCycler®480 Gene Scanning software (v1.2, Roche Diagnostics, Bavaria, Germany). Polymerase chain reaction (PCR) amplifications were conducted in the LightCycler® 480 (Roche Diagnostics). The PCR reaction mixture (20 μL) included the following: 0.5 μL forward primer (10 μM), 0.5 μL reverse primer (10 μM), 0.2 μL Hot Star Taq® Plus DNA Polymerase (5 U/μL), 1 μL 20×EVA-GREEN, 2 μL dNTP (10 mM), 1 μL genomic DNA (10 ng/μL), 2 μL MgCl2 (25 mM), 2 μL 10×buffer, and 10.8 μL H2O. Real-time PCR was performed with the following conditions: an initial denaturation at 95°C for 15 min, followed by 50 cycles of denaturation at 95°C for 10 s, annealing at 60°C for 15 s, extension at 72°C for 25 s. Following the completion of the cycle program, PCR products were denatured at 95°C for 1 min and cooled to 40°C for 1 min to form double-stranded DNA. The HRM analysis was then performed by gradually increasing the temperature from 65°C to 95°C at a rate of 0.01°C/s. Three DNA samples with known genotypes were run simultaneously in each experiment as a reference, and 10% of the samples were randomly selected to genotype twice; all results were identical.

DNA sequencing

PCR products were purified using shrimp alkaline phosphatase (SAP). Sequencing primers for the five SNPs were the same as primers in PCR. Nucleotide sequencing was detected by BigDye Terminator v3.1 Cycle Sequencing Kit and ABI 3130 genetic analyzer (Applied Biosystems, California, USA).

In-silico analysis of microRNA-binding and function prediction

The mature human microRNA sequences were obtained from the microRNA database (miRBase) (http://microrna.sanger.ac.uk). A region comprising the rs3742330 plus 15 bp 5' and 3' was included for analyzing hybridization of putative microRNAs using miRanda software with default parameters. The predicted analysis for rs7813 and rs636832 was conducted using Polyphen2 online software (http://genetics.bwh.harvard.edu/pph2/).

Statistical analysis

The Goodness-of-fit chi-square test (χ2) was used for testing Hardy-Weinberg Equilibrium (HWE) with cases and controls. Differences in demographic characteristics were assessed by Student’s t-test (for continuous variables) or χ2 test (for categorical variables). Logistic regression was used to analyze the associations between SNPs and susceptibility of GC, adjusted by sex, age, smoking status, and drinking status. All the statistical analyses were two-sided and P < 0.05 was set as a criterion for statistical significance. All statistical analyses were performed using SPSS statistical software (version 20.0, SPSS Inc., USA).

ACKNOWLEDGMENTS

This work was supported by grants from the National Natural Science Foundation of China [81672096] and the Projects in the Science and Technology Department of Sichuan Province pillar program [2017FZ0065].

CONFLICTS OF INTEREST

The authors declare that they have no conflicts of interest.

REFERENCES

1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015; 65:87–108.

2. Stadtländer CT, Waterbor JW. Molecular epidemiology, pathogenesis and prevention of gastric cancer. Carcinogenesis. 1999; 20:2195–208.

3. Ueda T, Volinia S, Okumura H, Shimizu M, Taccioli C, Rossi S, Alder H, Liu CG, Oue N, Yasui W, Yoshida K, Sasaki H, Nomura S, et al. Relation between microRNA expression and progression and prognosis of gastric cancer: a microRNA expression analysis. Lancet Oncol. 2010; 11:136–46.

4. Zhao XD, Lu YY, Guo H, Xie HH, He LJ, Shen GF, Zhou JF, Li T, Hu SJ, Zhou L, Han YN, Liang SL, Wang X, et al. MicroRNA-7/NF-κB signaling regulatory feedback circuit regulates gastric carcinogenesis. J Cell Biol. 2015; 210:613–27.

5. Chen J, Sun D, Chu H, Gong Z, Zhang C, Gong B, Li Y, Li N, Jiang L. Screening of differential microRNA expression in gastric signet ring cell carcinoma and gastric adenocarcinoma and target gene prediction. Oncol Rep. 2015;33:2963-71. https://doi.org/10.3892/or.2015.3935.

6. Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA Targets Cell. 2005; 120:15-20. https://doi.org/10.1016/j.cell.2004.12.035.

7. Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH, Kim VN. MicroRNA genes are transcribed by RNA polymerase II. EMBO J. 2004; 23: 4051-60. https://doi.org/10.1038/sj.emboj.7600385.

8. Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Rådmark O, Kim S, Kim VN. The nuclear RNase III Drosha initiates microRNA processing. Nature. 2003; 425:415–19.

9. Han J, Lee Y, Yeom KH, Nam JW, Heo I, Rhee JK, Sohn SY, Cho Y, Zhang BT, Kim VN. Molecular basis for the recognition of primary microRNAs by the Drosha-DGCR8 complex. Cell. 2006; 125:887–901.

10. McCarthy N. Cancer: small losses, big gains with microRNAs. Nat Rev Genet. 2010; 11:8.

11. Chendrimada TP, Gregory RI, Kumaraswamy E, Norman J, Cooch N, Nishikura K, Shiekhattar R. TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing. Nature. 2005; 436:740–44.

12. Arribas-Hernández L, Kielpinski LJ, Brodersen P. mRNA decay of most arabidopsis miRNA targets requires slicer activity of AGO1. Plant Physiol. 2016; 171:2620–32.

13. Slaby O, Bienertova-Vasku J, Svoboda M, Vyzula R. Genetic polymorphisms and microRNAs: new direction in molecular epidemiology of solid cancer. J Cell Mol Med. 2012; 16:8–21.

14. Tolia NH, Joshua-Tor L. Slicer and the argonautes. Nat Chem Biol. 2007; 3:36–43.

15. Carthew RW, Sontheimer EJ. Origins and Mechanisms of miRNAs and siRNAs. Cell. 2009; 136:642–55.

16. Farazi TA, Spitzer JI, Morozov P, Tuschl T. miRNAs in human cancer. J Pathol. 2011; 223:102–15.

17. Fernández C, Bellosillo B, Ferraro M, Seoane A, Sánchez-González B, Pairet S, Pons A, Barranco L, Vela MC, Gimeno E, Colomo L, Besses C, Navarro A, Salar A. MicroRNAs 142-3p, miR-155 and miR-203 are deregulated in gastric MALT lymphomas compared to chronic gastritis. Cancer Genomics Proteomics. 2017; 14:75–82.

18. Segura MF, Belitskaya-Lévy I, Rose AE, Zakrzewski J, Gaziel A, Hanniford D, Darvishian F, Berman RS, Shapiro RL, Pavlick AC, Osman I, Hernando E. Melanoma MicroRNA signature predicts post-recurrence survival. Clin Cancer Res. 2010; 16:1577-86. https://doi.org/10.1158/1078-0432.CCR-09-2721.

19. Nagano Y, Toiyama Y, Okugawa Y, Imaoka H, Fujikawa H, Yasuda H, Yoshiyama S, Hiro J, Kobayashi M, Ohi M, Araki T, Inoue Y, Mohri Y, Kusunoki M. MicroRNA-7 is associated with malignant potential and poor prognosis in human colorectal cancer. Anticancer Res. 2016; 36:6521–26.

20. Ahn DH, Rah H, Choi YK, Jeon YJ, Min KT, Kwack K, Hong SP, Hwang SG, Kim NK. Association of the miR-146aC>G, miR-149T>C, miR-196a2T>C, and miR-499A>G polymorphisms with gastric cancer risk and survival in the Korean population. Mol Carcinog. 2013 (Suppl 1); 52:E39–51.

21. Xiong XD, Luo XP, Cheng J, Liu X, Li EM, Zeng LQ. A genetic variant in pre-miR-27a is associated with a reduced cervical cancer risk in southern Chinese women. Gynecol Oncol. 2014; 132:450–54.

22. Shi J, Liu Y, Liu J, Zhou J. Hsa-miR-449a genetic variant is associated with risk of gastric cancer in a Chinese population. Int J Clin Exp Pathol. 2015; 8:13387–92.

23. Jin X, Yu N. MicroRNA-421 Gene Polymorphism in Gastric Carcinoma. Med Sci Monit. 2016; 22:1467–71.

24. Kumar MS, Lu J, Mercer KL, Golub TR, Jacks T. Impaired microRNA processing enhances cellular transformation and tumorigenesis. Nat Genet. 2007; 39:673–77.

25. Jiang Y, Chen J, Wu J, Hu Z, Qin Z, Liu X, Guan X, Wang Y, Han J, Jiang T, Jin G, Zhang M, Ma H, et al. Evaluation of genetic variants in microRNA biosynthesis genes and risk of breast cancer in Chinese women. Int J Cancer. 2013; 133:2216–24.

26. Xie Y, Wang Y, Zhao Y, Guo Z. Single-nucleotide polymorphisms of microRNA processing machinery genes are associated with risk for gastric cancer. Onco Targets Ther. 2015; 8:567–71.

27. Zhao Y, Du Y, Zhao S, Guo Z. Single-nucleotide polymorphisms of microRNA processing machinery genes and risk of colorectal cancer. Onco Targets Ther. 2015; 8:421–25.

28. Tchernitsa O, Kasajima A, Schäfer R, Kuban RJ, Ungethüm U, Györffy B, Neumann U, Simon E, Weichert W, Ebert MP, Röcken C. Systematic evaluation of the miRNA-ome and its downstream effects on mRNA expression identifies gastric cancer progression. J Pathol. 2010; 222:310–19.

29. Zhang J, Zhang XH, Wang CX, Liu B, Fan XS, Wen JJ, Shi QL, Zhou XJ. Dysregulation of microRNA biosynthesis enzyme Dicer plays an important role in gastric cancer progression. Int J Clin Exp Pathol. 2014; 7:1702–07.

30. Osuch-Wojcikiewicz E, Bruzgielewicz A, Niemczyk K, Sieniawska-Buccella O, Nowak A, Walczak A, Majsterek I. Association of Polymorphic Variants of miRNA Processing Genes with Larynx Cancer Risk in a Polish Population. BioMed Res Int. 2015; 2015:298378.

31. Cho SH, Ko JJ, Kim JO, Jeon YJ, Yoo JK, Oh J, Oh D, Kim JW, Kim NK. 3′-UTR Polymorphisms in the MiRNA Machinery Genes DROSHA, DICER1, RAN, and XPO5 Are Associated with Colorectal Cancer Risk in a Korean Population. PLoS One. 2015; 10:e0131125.

32. Persson H, Kvist A, Rego N, Staaf J, Vallon-Christersson J, Luts L, Loman N, Jonsson G, Naya H, Hoglund M, Borg A, Rovira C. Identification of new microRNAs in paired normal and tumor breast tissue suggests a dual role for the ERBB2/Her2 gene. Cancer Res. 2011; 71:78–86.

33. Friedländer MR, Mackowiak SD, Li N, Chen W, Rajewsky N. miRDeep2 accurately identifies known and hundreds of novel microRNA genes in seven animal clades. Nucleic Acids Res. 2012; 40:37–52.

34. Fang X, Yin Z, Li X, Xia L, Zhou B. Polymorphisms in GEMIN4 and AGO1 Genes Are Associated with the Risk of Lung Cancer: A Case-Control Study in Chinese Female Non-Smokers. Int J Environ Res Public Health. 2016; 13:939.

35. Qu Y, Qu H, Luo M, Wang P, Song C, Wang K, Zhang J, Dai L. MicroRNAs related polymorphisms and genetic susceptibility to esophageal squamous cell carcinoma. Mol Genet Genomics. 2014; 289:1123–30.

36. Shi Z, Zhang J, Qian X, Han L, Zhang K, Chen L, Liu J, Ren Y, Yang M, Zhang A, Pu P, Kang C. AC1MMYR2, an inhibitor of dicer-mediated biogenesis of Oncomir miR-21, reverses epithelial-mesenchymal transition and suppresses tumor growth and progression. Cancer Res. 2013; 73:5519–31.

37. Iio A, Takagi T, Miki K, Naoe T, Nakayama A, Akao Y. DDX6 post-transcriptionally down-regulates miR-143/145 expression through host gene NCR143/145 in cancer cells. Biochim Biophys Acta. 2013; 1829:1102–10.

38. Kim MN, Kim JO, Lee SM, Park H, Lee JH, Rim KS, Hwang SG, Kim NK. Variation in the Dicer and RAN genes are associated with survival in patients with hepatocellular carcinoma. PLoS One. 2016; 11:e0162279.

39. Nikolić Z, Savić Pavićević D, Vučić N, Cerović S, Vukotić V, Brajušković G. Genetic variants in RNA-induced silencing complex genes and prostate cancer. World J Urol. 2017; 35:613–24.

40. Yang PW, Huang YC, Hsieh CY, Hua KT, Huang YT, Chiang TH, Chen JS, Huang PM, Hsu HH, Kuo SW, Kuo ML, Lee JM. Association of miRNA-related genetic polymorphisms and prognosis in patients with esophageal squamous cell carcinoma. Ann Surg Oncol. 2014 (Suppl 4); 21:S601–09.

41. Koesters R, Adams V, Betts D, Moos R, Schmid M, Siermann A, Hassam S, Weitz S, Lichter P, Heitz PU, von Knebel Doeberitz M, Briner J. Human eukaryotic initiation factor EIF2C1 gene: cDNA sequence, genomic organization, localization to chromosomal bands 1p34-p35, and expression. Genomics. 1999; 61:210–18.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 21199