Skip to main content
Erschienen in: BMC Cancer 1/2023

Open Access 01.12.2023 | Research

Association study for the role of MMP8 gene polymorphisms in Colorectal cancer susceptibility

verfasst von: Shuyong Yu, Jiajia Cheng, Ping Li, Le Tian, Zhuang Chen, Zhaowei Chen, Yongyu Li, Jian Song

Erschienen in: BMC Cancer | Ausgabe 1/2023

Abstract

Background

Colorectal cancer (CRC) is one of the most common malignant tumors, influenced by several genetic loci in its clinical phenotypes. The aim of this study was to determine the relationship between the MMP8 gene polymorphism and CRC risk in the Chinese Han population.

Method

This study recruited 688 CRC patients and 690 healthy controls. The relationship between MMP8 polymorphism and CRC susceptibility was assessed by calculating the odds ratio (OR) and 95% confidence interval (CI) after stratifying by age, gender, body mass index (BMI), smoking, and alcohol consumption under a multi-genetic model.

Results

MMP8 rs3740938 was associated with increased CRC predisposition (p = 0.016, OR = 1.24, 95% CI: 1.04–1.48), and this association was detected particularly in subjects aged > 60 years, females, people with BMI > 24 kg/m2, smokers, and drinkers. Moreover, rs3740938 was found to be associated with the pathological type of rectal cancer.

Conclusions

Our results first displayed that rs3740938 in MMP8 was a risk factor for CRC predisposition. This finding may provide a new biological perspective for understanding the role of the MMP8 gene in CRC pathogenesis.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12885-023-11662-z.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Colorectal cancer (CRC) is the third most diagnosed cancer in the world and the second leading cause of cancer death, with a 10.0% incidence and 9.4% mortality [1]. According to Global Cancer Statistics 2020, more than 1.9 million new CRC (including anus) cases and 935,000 deaths were estimated to occur in 2020, representing about one in 10 cancer cases and deaths [1]. According to the National Cancer Center (NCC) of China, CRC is one of the most prevalent cancers, affecting around 408,000 individuals, making up 10% of all cancer cases in the country [2]. In recent decades, the incidence and mortality rates of CRC have been on the rise in recent decades in China [3], and its underlying pathogenesis of CRC remains unclear. While genetic and environmental factors are believed to play a vital role in CRC development [4], several demographic and lifestyle factors such as age, gender, alcohol use, smoking, high body mass index (BMI), and low physical activity have also been reported to be associated with an increased risk of developing CRC and higher mortality rates among patients [5]. At present, many common single-nucleotide polymorphisms (SNPs) associated with CRC risk have been successfully known through genome-wide association studies (GWAS), however, there are still many SNPs susceptible to CRC that have not been identified [6, 7]. Therefore, further investigation into oncogenic SNPs associated with CRC risk is imperative.
MMP8 (matrix metallopeptidase 8), a member of the MMP family, is an endopeptidase primarily produced by neutrophils. It plays a crucial role in degrading extracellular matrix proteins, growth factors, and cytokines [8]. According to reports, MMP8 is involved in the progression, metastasis, and invasion of cancer through its pro-cancer and anti-tumor functions [9]. For instance, MMP8 has been shown to increase cell-cell adhesion and reduce migration of tongue carcinoma cells by cleaving the anti-adhesive protein FXYD5 [10]. High serum MMP8 levels are associated with reduced survival and systemic inflammation of CRC patients [11]. The enhanced-serum MMP8 level in CRC patients was significantly related to advanced-stage CRC, distant metastasis, lack of MMR, and poor survival [12]. Relevant studies have concluded that the polymorphisms of MMP8 are associated with the risk of a variety of cancers, including breast cancer [13], thyroid cancer [14], and laryngeal squamous cell carcinoma [15]. Previously, the association between MMP8 rs11225395 and CRC susceptibility has been reported [16]. The impact of MMP8 genotypes on CRC risk in Taiwan has been explored [17]. The relationship between other loci in MMP8 and the risk of CRC has not been reported yet.
In previous studies, the association of MMP8 rs3740938 with the risk of breast cancer has been explored [18]. MMP8 rs1940475 is associated with the risk of breast cancer [18] and gastric ulcer [19]. MMP8 rs3765620 is related to ischemic stroke susceptibility [20]. However, the role of these polymorphisms in CRC susceptibility has not been reported. In this case-control study, we selected three variants (rs3740938, rs1940475, and rs3765620) in the exon region of the MMP8 gene to explore their role in CRC occurrence in the Chinese Han population.

Methods

Subjects

To ensure the accuracy and credibility of the research results, before we plan to conduct this study, we used G*power 3.1.9.7 software (https://​stats.​idre.​ucla.​edu/​other/​gpower/​) to estimate the sample size. The specific parameters we set are as follows: effect size d = 0.2; α error probability = 0.05; power (1-β err prob) = 95%. This calculation yielded a sample consisting of at least 651 cases and 651 controls. Here, we enrolled a total of 1378 subjects (688 CRC cases and 690 healthy controls) from Hainan Cancer Hospital from 2020 to 2023. The inclusion criteria for CRC cases are: newly diagnosed and histologically confirmed by rectoscopy, endorectal ultrasonography, magnetic resonance imaging (MRI), computed tomography, and histopathological results based on the American Joint Committee on Cancer (AJCC) classification. Patients with a history of cancer or severe chronic diseases were excluded. Prior to any treatment, blood samples were collected from the patient. For the control group, we selected healthy individuals without malignant tumors or digestive diseases from the same hospital as cases, ensuring they were genetically unrelated to CRC patients. All subjects belonged to the Chinese Han ethnicity, and no minors or illiterates were involved in our study. Questionnaires surveys and medical records were used to obtain epidemiological characteristics and pathological data. This research plan was implemented in accordance with the Helsinki Declaration and approved by the Ethics Committee of Hainan Cancer Hospital (No. ZDKJ202008). All subjects provided written informed consent before registration in this study.

Genotyping

Three SNPs (rs3740938, rs1940475, and rs3765620) in MMP8 were chosen for analysis. The selection criteria for these SNPs included (1) the dbSNP database with minor allele frequency (MAFs) ≥ 5%; (2) with MassARRAY primer design, a call rate > 99%; (3) and previous association studies [1820]. Bioinformatics tools such as dbSNP (https://​www.​ncbi.​nlm.​nih.​gov/​snp/​), HaploReg v4.1 (https://​pubs.​broadinstitute.​org/​mammals/​haploreg/​haploreg.​php), RegulomeDB (https://​regulome.​stanford.​edu/​regulome-search/​), and QTLbase (http://​www.​mulinlab.​org/​qtlbase/​index.​html) were employed to identify the potentially functional SNPs.
Peripheral whole blood samples (5 mL) were obtained from each participant and stored in tubes containing Ethylene Diamine Tetraacetic Acid (EDTA) anticoagulant. Within 24 h, genomic DNA was isolated using the GoldMag DNA Purification Kit (GoldMag Co. Ltd., Xi’an, China). The DNA samples were quantified using NanoDrop 2000 (Thermo Scientific, Waltham, MA, USA) and stored at − 20 °C. DNA samples with low concentrations or poor quality were excluded from subsequent studies. The MassARRAY platform is based on MALDI-TOF (matrix-assisted laser desorption/ionization—time of flight) mass spectrometry [21, 22]. The analytical accuracy of MALDI-TOF MS is quite high, 0.1–0.01% of the determined mass. Genotyping was performed using the Agena MassARRAY system (Agena, San Diego, CA, USA) with incorporated software (https://​www.​agenabio.​com/​). In addition, this study also set up double wells for each sample to ensure the accuracy of the results. For quality control, about 10% of the total samples were chosen randomly and re-genotyped, and the concordance rate reached 100%.

Statistical data

The demographic data between two groups were tested by student t-test or χ2 test for continuous or categorical variables, respectively. The Hardy Weinberg balance (HWE) of the control group was assessed by a goodness-of-fit χ2 test. The relationship of MMP8 polymorphisms with CRC risk was determined by calculating odd ratios (ORs) and 95% confidence intervals (CIs) using a multi-genetic model adjusted for age, sex, BMI, smoking, and alcohol consumption. SNPstats (https://​www.​snpstats.​net/​start.​htm) was utilized for this analysis. The subgroup analyses were completed within specific subpopulations stratified by age, sex, BMI, smoking, and drinking status. In addition, the impact of genotypes on different pathological types was also evaluated. The false positive reporting probability (FPRP) threshold was set at 0.2 with a prior probability of 0.1, which is used to evaluate the significant association of significant findings [23]. The optimal SNP-SNP interaction model was determined through multifactor dimensionality reduction (MDR) analysis. The data analysis was conducted using SPSS version 18.0 (SPSS Inc., Chicago, Illinois, USA) and MDR version 3.0.2 software. A statistical significance was defined as p value < 0.05, and a Bonferroni-corrected p < 0.05/3 was considered significance.

Results

Subjects characteristics

The case group (59.78 ± 11.29 years) included 402 males and 286 females, and the control group (59.62 ± 9.55 years) consisted of 404 males and 286 females (Table 1). No significant differences between the two groups were found in terms of age (p = 0.774), gender (p = 0.964), smoking (p = 0.624), and drinking (p = 0.828). There was a significant difference in BMI between the two groups (p < 0.001). Among the patient cohort, there were 320 (46.5%) individuals diagnosed with colon cancer and 368 (53.5%) with rectal cancer. Within this group, 183 (26.6%) patients experienced lymph node metastasis, while 263 (38.2%) patients were classified as stage III-IV based on their cancer staging.
Table 1
Characteristics of patients with CRC and health controls
Variable
Cases (688)
Control (690)
p
Age
Mean ± SD, years
59.78 ± 11.29
59.62 ± 9.55
0.774
 
> 60years
353 (51.3%)
380 (55.1%)
 
≤ 60 years
335 (48.7%)
310 (44.9%)
Gender
   
0.964
 
Male
402 (58.4%)
404 (58.6%)
 
Female
286 (41.6%)
286 (41.4%)
Smoking
   
0.624
 
Yes
312 (45.3%)
322 (46.7%)
 
No
376 (54.7%)
368 (53.3%)
Drinking
   
0.828
 
Yes
330 (48.0%)
335 (48.6%)
 
No
358 (52.0%)
355 (51.4%)
BMI
   
< 0.001
 
> 24 kg/m2
150 (33.0%)
216 (51.2%)
 
≤ 24 kg/m2
304 (67.0%)
206 (48.8%)
Lymph nodes metastasis
    
 
Yes
183 (26.6%)
  
No
53 (7.7%)
Unavailable
452 (65.7%)
Pathological type
    
 
Colon cancer
320 (46.5%)
Rectal cancer
368 (53.5%)
Stage
    
 
I-II
109 (15.8%)
  
III-IV
263 (38.2%)
Unavailable
316 (45.9%)
CRC, colorectal cancer; BMI, body mass index
p values were calculated by χ2 test or the Student’s t test
p < 0.05 indicates statistical significance

Relationship of selected variants with CRC risk

Three SNPs (rs3740938, rs1940475, and rs3765620) in MMP8 were genotyped, and the MAFs of these three SNPs in the two groups were all > 0.05 (Table 2). All HWE p–-values for these variants were > 0.05. The results of genotyping displayed that the genotyping success rate of each SNP was > 99.5%. RegulomeDB analysis displayed that rs1940475 was associated with eQTL/caQTL, transcription factor (TF) binding/chromatin accessibility peak. HaploReg v4.1 database displayed that these SNPs might be associated with the regulation of promoter/ enhancer histone marks, DNAse, and /or motif changes. Moreover, the genotypes of rs3740938 (p = 0.027), rs1940475 (p = 2.720e-13), and rs3765620 (p = 1.620e-12) were associated with the expression of MMP8 in blood. In the allele model, rs3740938 was associated with the higher CRC risk (p = 0.016, OR = 1.24, 95% CI: 1.04–1.48).
Table 2
Details of MMP8 SNPs and the allele model for the association with CRC risk
SNP ID
Chr: Position
Alleles
(Ref/Alt)
MAF
HWE
Call rate
OR (95% CI)
p
Exon
dbSNP func annot
Haploreg4.1
RegulomeDB
P-Value for eQTL in Blood
Cases
Controls
O(HET)
E(HET)
p
rs3740938
11:102716331
 A/G
0.257
0.218
0.326
0.341
0.264
99.8%
1.24 (1.04–1.48)
0.016*
6
Synonymous
L (Leu) > L (Leu)
Promoter histone marks; Enhancer histone marks; Motifs changed
other
0.027
rs1940475
11:102722517
T/C
0.393
0.367
0.454
0.465
0.514
100.0%
1.12 (0.96–1.30)
0.163
2
Missense
K (Lys) > E (Glu)
Enhancer histone marks; DNAse; Motifs changed
eQTL/caQTL + TF binding / chromatin accessibility peak
2.720e-13
rs3765620
11:102724761
G/A
0.386
0.359
0.448
0.460
0.508
99.8%
1.13 (0.96–1.31)
0.134
1
Missense
T (Thr) > I (Ile)
Enhancer histone marks; DNAse; Motifs changed
other
1.620e-12
SNP, Single nucleotide polymorphism; CRC, colorectal cancer; MAF, Minor allele frequency; HWE, Hardy-Weinberg equilibrium; O(HET), Observed heterozygosity frequency; E(HET), Expected heterozygosity frequency
p values were calculated from Person’s chi-square test (two-sided)
*p < 0.05 indicates statistical significance. Bold p means that the data is statistically significant after Bonferroni correction (p < 0.05/3)
Genetic model analysis between selected variants and CRC risk was shown in Table 3. MMP8 rs3740938 contributed to an increased predisposition to CRC under the codominant (p = 0.044, OR = 1.31, 95% CI: 1.04–1.64), dominant (p = 0.013, OR = 1.31, 95% CI: 1.06–1.63) and log-additive (p = 0.017, OR = 1.24, 95% CI: 1.04–1.47) models. The risk-increasing significance of rs3740938 for CRC occurrence still existed after Bonferroni multiple correction (p < 0.05/3).
Table 3
Effect of MMP8 variants on the susceptibility to CRC
SNP ID
Model
Genotype
Control
Case
OR (95% CI)
p
rs3740938
Codominant
GG
427 (61.9%)
379 (55.2%)
1
0.044*
GA
225 (32.6%)
261 (38%)
1.31 (1.04–1.64)
AA
38 (5.5%)
46 (6.7%)
1.37 (0.87–2.15)
Dominant
GG
427 (61.9%)
379 (55.2%)
1
0.013*
GA-AA
263 (38.1%)
307 (44.8%)
1.31 (1.06–1.63)
Recessive
GG-GA
652 (94.5%)
640 (93.3%)
1
0.350
AA
38 (5.5%)
46 (6.7%)
1.24 (0.79–1.93)
Log-additive
---
---
---
1.24 (1.04–1.47)
0.017*
rs1940475
Codominant
CC
280 (40.6%)
261 (37.9%)
1
0.360
CT
313 (45.4%)
313 (45.5%)
1.08 (0.85–1.35)
TT
97 (14.1%)
114 (16.6%)
1.26 (0.92–1.74)
Dominant
CC
280 (40.6%)
261 (37.9%)
1
0.310
CT-TT
410 (59.4%)
427 (62.1%)
1.12 (0.90–1.39)
Recessive
CC-CT
593 (85.9%)
574 (83.4%)
1
0.200
TT
97 (14.1%)
114 (16.6%)
1.21 (0.90–1.63)
Log-additive
---
---
---
1.11 (0.96–1.30)
0.170
rs3765620
Codominant
A/A
288 (41.7%)
263 (38.3%)
1
0.330
G/A
309 (44.8%)
316 (46.1%)
1.12 (0.89–1.41)
G/G
93 (13.5%)
107 (15.6%)
1.26 (0.91–1.75)
Dominant
A/A
288 (41.7%)
263 (38.3%)
1
0.190
G/A-G/G
402 (58.3%)
423 (61.7%)
1.15 (0.93–1.43)
Recessive
A/A-G/A
597 (86.5%)
579 (84.4%)
1
0.270
G/G
93 (13.5%)
107 (15.6%)
1.19 (0.88–1.60)
Log-additive
---
---
---
1.12 (0.96–1.31)
0.140
CRC, colorectal cancer; SNP, single nucleotide polymorphism; OR, odds ratio; 95% CI, 95% confidence interval; BMI, body mass index
p values were calculated by logistic regression analysis with adjustments for age, gender, BMI, smoking and drinking
*p < 0.05 respects the data is statistically significant. Bold p means that the data is statistically significant after Bonferroni correction (p < 0.05/3)

Stratification analysis

Stratification analyses by age, gender, BMI, tobacco use, and alcohol were displayed in Fig. 1; Table 4 and Suppl_Table 1. In subjects aged > 60 years, rs3740938 (codominant: p = 0.045, OR = 1.49; and dominant: p = 0.018, OR = 1.44) might contribute to the increasing CRC risk. After stratification by gender, an association between rs3740938 and an increased CRC susceptibility was observed among females under the dominant (p = 0.014, OR = 1.53) and log-additive (p = 0.027, OR = 1.37) models, and this SNP could be identified as a potential risk marker with a marginal p value in codominant model (p = 0.048, OR = 1.55). In subjects with BMI > 24 kg/m2, rs3740938 conferred to the higher susceptibility to CRC (codominant: p = 0.033, OR = 1.67; dominant: p = 0.011, OR = 1.74; and log-additive: p = 0.010, OR = 1.58). In smokers, rs3740938 might be a -risk-increasing factor for CRC under the codominant (p = 0.044, OR = 1.44), dominant (p = 0.015, OR = 1.49) and log-additive (p = 0.014, OR = 1.38) models. Among drinkers, rs3740938 was associated with the increased CRC susceptibility (codominant: p = 0.042, OR = 1.48; dominant: p = 0.012, OR = 1.49; and log-additive: p = 0.018, OR = 1.34). After Bonferroni multiple correction, the relationships of rs3740938 with CRC susceptibility in females, subjects with BMI > 24 kg/m2, smokers and drinkers were also remarkable.
Table 4
Stratification for the effect of MMP8 rs3740938 variant on CRC susceptibility
Model
Genotype
Control
Case
OR (95% CI)
p -value
Control
Case
OR (95% CI)
p -value
Age stratification
 
Age > 60 years
Age ≤ 60 years
Codominant
G/G
239 (62.9%)
191 (54.1%)
1
0.045*
188 (60.6%)
188 (56.5%)
1
0.210
 
G/A
117 (30.8%)
140 (39.7%)
1.49 (1.09–2.05)
108 (34.8%)
121 (36.3%)
1.14 (0.82–1.59)
 
A/A
24 (6.3%)
22 (6.2%)
1.17 (0.63–2.16)
14 (4.5%)
24 (7.2%)
1.83 (0.91–3.67)
Dominant
G/G
239 (62.9%)
191 (54.1%)
1
0.018*
188 (60.6%)
188 (56.5%)
1
0.230
 
G/A-A/A
141 (37.1%)
162 (45.9%)
1.44 (1.06–1.94)
122 (39.4%)
145 (43.5%)
1.22 (0.88–1.67)
Recessive
G/G-G/A
356 (93.7%)
331 (93.8%)
1
0.980
296 (95.5%)
309 (92.8%)
1
0.110
 
 A/A
24 (6.3%)
22 (6.2%)
1.01 (0.55–1.85)
14 (4.5%)
24 (7.2%)
1.73 (0.87–3.44)
Log-additive
---
---
---
1.26 (0.99–1.61)
0.056
---
---
1.24 (0.95–1.61)
0.110
Gender stratification
 
Males
Females
Codominant
G/G
235 (58.2%)
217 (54.0%)
1
0.450
192 (67.1%)
162 (57.0%)
1
0.048*
G/A
145 (35.9%)
156 (38.8%)
1.16 (0.86–1.55)
80 (28.0%)
105 (37.0%)
1.55 (1.08–2.22)
A/A
24 (5.9%)
29 (7.2%)
1.33 (0.75–2.36)
14 (4.9%)
17 (6.0%)
1.42 (0.68–2.99)
Dominant
G/G
235 (58.2%)
217 (54.0%)
1
0.240
192 (67.1%)
162 (57.0%)
1
0.014*
G/A-A/A
169 (41.8%)
185 (46.0%)
1.18 (0.89–1.56)
94 (32.9%)
122 (43.0%)
1.53 (1.09–2.16)
Recessive
G/G-G/A
380 (94.1%)
373 (92.8%)
1
0.430
272 (95.1%)
267 (94.0%)
1
0.590
 A/A
24 (5.9%)
29 (7.2%)
1.25 (0.71–2.20)
14 (4.9%)
17 (6.0%)
1.22 (0.59–2.54)
Log-additive
---
---
---
1.16 (0.92–1.45)
0.210
---
---
1.37 (1.03–1.81)
0.027*
BMI stratification
 
BMI > 24 kg/m2
BMI ≤ 24 kg/m2
Codominant
G/G
141 (65.3%)
79 (52.7%)
1
0.033*
131 (63.6%)
179 (59.1%)
1
0.300
G/A
65 (30.1%)
59 (39.3%)
1.67 (1.06–2.62)
62 (30.1%)
110 (36.3%)
1.29 (0.88–1.90)
A/A
10 (4.6%)
12 (8%)
2.25 (0.92–5.48)
13 (6.3%)
14 (4.6%)
0.78 (0.35–1.73)
Dominant
G/G
141 (65.3%)
79 (52.7%)
1
0.011*
131 (63.6%)
179 (59.1%)
1
0.320
G/A-A/A
75 (34.7%)
71 (47.3%)
1.74 (1.13–2.69)
75 (36.4%)
124 (40.9%)
1.20 (0.83–1.74)
Recessive
G/G-G/A
206 (95.4%)
138 (92.0%)
1
0.170
193 (93.7%)
289 (95.4%)
1
0.400
 A/A
10 (4.6%)
12 (80.0%)
1.85 (0.77–4.42)
13 (6.3%)
14 (4.6%)
0.72 (0.33–1.56)
Log-additive
---
---
---
1.58 (1.11–2.24)
0.010*
---
---
1.08 (0.80–1.46)
0.620
Smoking stratification
 
Smokers
Non-smokers
Codominant
G/G
206 (64.0%)
169 (54.3%)
1
0.044*
221 (60.0%)
210 (56.0%)
1
0.470
G/A
100 (31.1%)
119 (38.3%)
1.44 (1.03–2.02)
125 (34.0%)
142 (37.9%)
1.21 (0.89–1.65)
A/A
16 (5.0%)
23 (7.4%)
1.77 (0.90–3.47)
22 (6.0%)
23 (6.1%)
1.10 (0.60–2.04)
Dominant
G/G
206 (64.0%)
169 (54.3%)
1
0.015*
221 (60.0%)
210 (56.0%)
1
0.240
G/A-A/A
116 (36.0%)
142 (45.7%)
1.49 (1.08–2.05)
147 (40.0%)
165 (44.0%)
1.19 (0.89–1.60)
Recessive
G/G-G/A
306 (95.0%)
288 (92.6%)
1
0.200
346 (94.0%)
352 (93.9%)
1
0.930
 A/A
16 (5.0%)
23 (7.4%)
1.54 (0.79–2.99)
22 (6.0%)
23 (6.1%)
1.03 (0.56–1.88)
Log-additive
---
---
---
1.38 (1.07–1.80)
0.014*
---
---
1.13 (0.89–1.43)
0.320
Drinking stratification
 
Drinkers
Non-drinkers
Codominant
G/G
211 (63.0%)
176 (53.5%)
1
0.042*
216 (60.9%)
203 (56.9%)
1
0.600
G/A
101 (30.1%)
124 (37.7%)
1.48 (1.06–2.06)
124 (34.9%)
137 (38.4%)
1.16 (0.85–1.59)
A/A
23 (6.9%)
29 (8.8%)
1.53 (0.85–2.75)
15 (4.2%)
17 (4.8%)
1.20 (0.58–2.47)
Dominant
G/G
211 (63.0%)
176 (53.5%)
1
0.012*
216 (60.9%)
203 (56.9%)
1
0.310
G/A-A/A
124 (37.0%)
153 (46.5%)
1.49 (1.09–2.03)
139 (39.1%)
154 (43.1%)
1.17 (0.86–1.58)
Recessive
G/G-G/A
312 (93.1%)
300 (91.2%)
1
0.330
340 (95.8%)
340 (95.2%)
1
0.740
 A/A
23 (6.9%)
29 (8.8%)
1.32 (0.75–2.35)
15 (4.2%)
17 (4.8%)
1.13 (0.55–2.30)
Log-additive
---
---
---
1.34 (1.05–1.70)
0.018*
---
---
1.13 (0.88–1.46)
0.330
CRC, colorectal cancer; SNP, single nucleotide polymorphism; OR, odds ratio; 95% CI, 95% confidence interval; BMI, body mass index
p values were calculated by logistic regression analysis with adjustments for age, gender, BMI, smoking or drinking
*p < 0.05 respects the data is statistically significant. Bold p means that the data is statistically significant after Bonferroni correction (p < 0.05/3)
We also explored the correlation of selected SNPs with the pathological types of CRC (Table 5). Stratifying by pathological type, rs3740938 was related to an increased risk of rectal cancer (dominant: p = 0.022, OR = 1.35; and log-additive: p = 0.018, OR = 1.34). Due to lack of information, the correlation of MMP8 variants with stage and lymph node metastasis in CRC patients has not been explored.
Table 5
Association between MMP8 polymorphisms and the risk of colon cancer and rectal cancer
SNP ID
Model
Genotype
Control
Colon cancer
Rectal cancer
N
OR (95% CI)
p -value
N
OR (95% CI)
p -value
rs3740938
Codominant
G/G
427 (61.9%)
179 (55.9%)
1
0.200
200 (54.6%)
1
0.064
G/A
225 (32.6%)
122 (38.1%)
1.29 (0.97–1.71)
139 (38.0%)
1.32 (1.01–1.73)
A/A
38 (5.5%)
19 (5.9%)
1.19 (0.67–2.13)
27 (7.4%)
1.53 (0.90–2.58)
Dominant
G/G
427 (61.9%)
179 (55.9%)
1
0.076
200 (54.6%)
1
0.022*
G/A-A/A
263 (38.1%)
141 (44.1%)
1.28 (0.98–1.67)
166 (45.4%)
1.35 (1.04–1.75)
Recessive
G/G-G/A
652 (94.5%)
301 (94.1%)
1
0.790
339 (92.6%)
1
0.230
 A/A
38 (5.5%)
19 (5.9%)
1.08 (0.61–1.91)
27 (7.4%)
1.37 (0.82–2.29)
Log-additive
---
---
---
1.19 (0.96–1.48)
0.120
---
1.28 (1.04–1.57)
0.021*
rs1940475
Codominant
C/C
280 (40.6%)
127 (39.7%)
1
0.790
134 (36.4%)
1
0.240
 C/T
313 (45.4%)
143 (44.7%)
1.01 (0.76–1.35)
170 (46.2%)
1.14 (0.86–1.50)
T/T
97 (14.1%)
50 (15.6%)
1.15 (0.77–1.71)
64 (17.4%)
1.38 (0.95–2.01)
Dominant
C/C
280 (40.6%)
127 (39.7%)
1
0.750
134 (36.4%)
1
0.180
 C/T-T/T
410 (59.4%)
193 (60.3%)
1.05 (0.80–1.37)
234 (63.6%)
1.19 (0.92–1.55)
Recessive
C/C-C/T
593 (85.9%)
270 (84.4%)
1
0.500
304 (82.6%)
1
0.160
T/T
97 (14.1%)
50 (15.6%)
1.14 (0.78–1.65)
64 (17.4%)
1.29 (0.91–1.82)
Log-additive
---
---
---
1.06 (0.87–1.28)
0.570
---
1.17 (0.97–1.40)
0.095
rs3765620
Codominant
A/A
288 (41.7%)
128 (40.0%)
1
0.750
135 (36.9%)
1
0.240
G/A
309 (44.8%)
144 (45.0%)
1.06 (0.79–1.41)
172 (47.0%)
1.19 (0.90–1.57)
G/G
93 (13.5%)
48 (15.0%)
1.17 (0.78–1.76)
59 (16.1%)
1.36 (0.92-2.00)
Dominant
A/A
288 (41.7%)
128 (40.0%)
1
0.560
135 (36.9%)
1
0.120
G/A-G/G
402 (58.3%)
192 (60.0%)
1.08 (0.83–1.42)
231 (63.1%)
1.23 (0.94–1.59)
Recessive
A/A-G/A
597 (86.5%)
272 (85.0%)
1
0.510
307 (83.9%)
1
0.240
G/G
93 (13.5%)
48 (15.0%)
1.14 (0.78–1.66)
59 (16.1%)
1.24 (0.87–1.77)
Log-additive
---
---
---
1.08 (0.89–1.30)
0.460
---
1.17 (0.97–1.41)
0.092
SNP, single nucleotide polymorphism; OR, odds ratio; 95% CI, 95% confidence interval
p values were calculated by logistic regression analysis with adjustments for age, gender, BMI, smoking or drinking
*p < 0.05 respects the data is statistically significant

FPRP analysis

Table 6 exhibited the results of the FPRP analysis, with a prior probability level of 0.1 and FPRP of < 0.2, for the positive results. The significant association between rs3740938 and CRC susceptibility remained noteworthy in the overall analysis. And this correlation persisted in females, subjects aged > 60 years, subjects with BMI > 24 kg/m2, smokers, and drinkers. Furthermore, the significant association of rs3740938 with susceptibility to rectal cancer remained prominent.
Table 6
False-positive report probability for the associations of variants in MMP with CRC risk
SNP ID
Model
OR (95% CI)
Prior probability
0.25
0.1
0.01
0.001
0.0001
Overall
       
 
Allele
1.24 (1.04–1.48)
0.049
0.134
0.630
0.945
0.994
 
Codominant
1.31 (1.04–1.64)
0.053
0.143
0.647
0.949
0.995
 
Dominant
1.31 (1.06–1.63)
0.044
0.122
0.605
0.939
0.994
 
Log-additive
1.24 (1.04–1.47)
0.038
0.106
0.567
0.930
0.992
Females
       
 
Codominant
1.55 (1.08–2.22)
0.052
0.141
0.644
0.948
0.995
 
Dominant
1.53 (1.09–2.16)
0.048
0.131
0.623
0.943
0.994
 
Log-additive
1.37 (1.03–1.81)
0.075
0.195
0.727
0.964
0.996
Age > 60 years
       
 
Codominant
1.49 (1.09–2.05)
0.043
0.118
0.595
0.937
0.993
 
Dominant
1.44 (1.06–1.94)
0.048
0.131
0.624
0.944
0.994
BMI > 24 kg/m2
       
 
Codominant
1.67 (1.06–2.62)
0.089
0.227
0.764
0.970
0.997
 
Dominant
1.74 (1.13–2.69)
0.049
0.135
0.631
0.945
0.994
 
Log-additive
1.58 (1.11–2.24)
0.033
0.092
0.527
0.918
0.991
Smokers
       
 
Codominant
1.44 (1.03–2.02)
0.097
0.243
0.780
0.973
0.997
 
Dominant
1.49 (1.08–2.05)
0.043
0.118
0.595
0.937
0.993
 
Log-additive
1.38 (1.07–1.80)
0.050
0.136
0.635
0.946
0.994
Drinkers
       
 
Codominant
1.48 (1.06–2.06)
0.059
0.158
0.674
0.954
0.995
 
Dominant
1.49 (1.09–2.03)
0.034
0.096
0.540
0.922
0.992
 
Log-additive
1.34 (1.05–1.70)
0.046
0.125
0.612
0.941
0.994
Rectal cancer
       
 
Codominant
1.32 (1.01–1.73)
0.117
0.285
0.814
0.978
0.998
 
Dominant
1.35 (1.04–1.75)
0.066
0.174
0.699
0.959
0.996
 
Log-additive
1.28 (1.04–1.57)
0.051
0.138
0.638
0.947
0.994
SNP, single nucleotide polymorphism; OR, odds ratio; 95% CI, 95% confidence interval
The level of false-positive report probability threshold was set at 0.2, and Bold represent that noteworthy findings are presented

MDR analysis

The interaction between these SNPs was evaluated using MDR analysis, and the results were shown in Table 7 and Suppl_Figure 1. Single–locus rs3740938 was the optimal model for evaluating CRC susceptibility (p = 0.012, testing accuracy = 0.5336, cross–validation consistency, 10/10) with the information gain of 0.34%.
Table 7
SNP–SNP interaction models of MMP8 SNPs analyzed by the MDR method
Model
Training Bal. Acc.
Testing Bal. Acc.
CVC
p
rs3740938
0.5336
0.5336
10/10
0.012*
rs3740938,rs1940475
0.5340
0.5263
9/10
0.012*
rs3740938,rs1940475,rs3765620
0.5378
0.5270
10/10
0.005*
MDR, multifactor dimensionality reduction; Bal. Acc., balanced accuracy; CVC, cross–validation consistency
p values were calculated using χ2 tests
*p < 0.05: indicates statistical significance

Discussion

In this study, we first examined the association of MMP8 rs3740938 with an increased CRC predisposition in the Chinese Han population. This relationship was particularly significant in subjects aged > 60 years, females, people with BMI > 24 kg/m2, smokers and drinkers. Moreover, MMP8 rs3740938 was related to the pathological type of rectal cancer. These findings contributed valuable data that could potentially be utilized in constructing a genetic panel for predicting CRC risk.
MMP8 is known to be expressed in various cancer types and may be associated with cancer cell invasion, proliferation, metastasis, and the poor prognosis of cancer patients [9]. MMP8 in serum was identified to be related to CRC patients with bad prognosis [11]. Protein array analysis showed decreased levels of circulating angiogenesis factor MMP8 during treatment with bevacizumab in metastatic CRC [24]. In CRC tissues, YKL-40 is associated with the expression of MMP8 and may be involved in the immunological properties of the tumor microenvironment [25]. MMP8. MMP8 rs11225395 was connected to the higher CRC risk in a Chinese Han population [16]. However, the contribution of three variants (rs3740938, rs1940475, and rs3765620) in the exon region of MMP8 gene to CRC occurrence has not been reported. Here, we examined these three SNPs in the MMP8 gene. Our data displayed a significant association between MMP8 rs3740938 and an increased CRC predisposition in the Chinese Han population. Bioinformatics analysis using HaploReg v4.1 demonstrated that rs3740938 was related to promoter histone marks, enhancer histone marks and motifs changed. Furthermore, according to the QTLbase database, the genotypes of rs3740938 (p = 0.027) were found to be negatively associated with the expression of MMP8 in blood. Compared with rs3740938-GG and -GA genotypes, AA genotype may be associated with the lower expression of MMP8 mRNA. These findings suggested that the role of rs3740938 in CRC may be through affecting gene expression of MMP8, thereby affecting CRC occurrence. However, further experimental confirmation is needed to validate this hypothesis.
The incidence and mortality rates related to CRC have shown a steady increase [26]. The incidence and mortality of CRC are often higher in men than in women [27]. Sex hormones are considered to be the factors leading to gender differences in the incidence and mortality of CRC [28]. Here, the relationship of MMP8 polymorphisms with CRC occurrence under the stratified analysis by age, sex and BMI was explored. MMP8 rs3740938 might contribute to an increased susceptibility to CRC in participants aged > 60 years, females, and people with BMI > 24 kg/m2, indicating that the effects of rs3740938 on CRC occurrence are specific to age, gender, and BMI. As is well known, smoking increases the risk of various cancers, such as lung cancer, head and neck cancer, stomach cancer, etc [29]. It is a significant risk factor for CRC, with a dose-dependent relationship where the risk increases with the intensity and duration of smoking [30]. For a long time, alcohol consumption has been shown to be associated with the development of CRC and is considered as a crucial targeted factor related to the adverse consequences of CRC [31]. We also explored the correlation of selected SNPs with cigarette and alcohol in relation to CRC occurrence, and our results indicated that rs3740938 might act as an increasing-risk factor for CRC in smokers and drinkers. Additionally, rs3740938 was also observed to be related to an increased risk of rectal cancer. Therefore, according to our research results, this locus can be identified as a key research object for further investigation into CRC risk.
There are also potential limitations to our research. First, the sample size of our study was limited, and all participants were Chinese Han people. Future studies will involve a larger and more diverse sample population, along with validation experiments to confirm our results. Second, the lack of comprehensive information on various factors such as environmental exposure, lifestyle choices, and clinicopathological data (including cancer progression, metastasis, and invasion) hinders a thorough understanding of their role in the association between the selected SNPs and CRC risk. Therefore, further studies is required to investigate these aspects and collected complete clinicopathological data to evaluate the relationship accurately. Third, the potential mechanisms and functions of these SNPs in relation to CRC risk, including association of high serum MMP8 levels withMMP8 SNPs have not been fully elucidated. In subsequent researches, we will design detailed experiments to explore the expression data of MMP8 and the potential mechanisms and functions of these SNPs in CRC. Fourth, our study only assesses the correlation between three SNPs in the exon region of the MMP8 gene and CRC risk, and a large number of exonic or intronic variants remain to be studied. In subsequent studies, we will further explore the association of other loci in MMP8 with CRC susceptibility. Despite the above limitations, this is the first study that has reported MMP8 rs3740938 was associated with the increased CRC predisposition in a Chinese Han population, and this variant could serve as potential biomarkers of CRC susceptibility. These findings increased our knowledge regarding the effect of MMP8 on the process of CRC occurrence, provided some data for future explorations of the relationship between MMP8 and CRC risk in different populations, and also helped to establish new warning and treatment methods for CRC in futures studies. In the next step, we will further explore the functions of these SNPs based on the results of this study, in order to provide new theoretical basis and targets for the diagnosis and treatment of CRC.

Conclusion

Our results show that MMP8 rs3740938 might be a risk-increasing factor for CRC, revealing for the first time the role of rs3740938 in MMP8 in CRC risk among the Chinese Han population. Our findings might provide new biological insights into the role of MMP8 gene in the formation and progression of CRC.

Acknowledgements

The authors thank all participants and volunteers in this study.

Declarations

This research plan was implemented in accordance with the Helsinki Declaration and approved by the Ethics Committee of Hainan Cancer Hospital (No. ZDKJ202008). All subjects have obtained written informed consent before registration.
Not applicable.

Competing interests

The authors declare that they have no conflict of interest..
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge
Literatur
1.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. Cancer J Clin. 2021;71(3):209–49.CrossRef Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. Cancer J Clin. 2021;71(3):209–49.CrossRef
2.
Zurück zum Zitat Maomao C, He L, Dianqin S, Siyi H, Xinxin Y, Fan Y, Shaoli Z, Changfa X, Lin L, Ji P, et al. Current cancer burden in China: epidemiology, etiology, and prevention. Cancer Biology & Medicine. 2022;19(8):1121–38.CrossRef Maomao C, He L, Dianqin S, Siyi H, Xinxin Y, Fan Y, Shaoli Z, Changfa X, Lin L, Ji P, et al. Current cancer burden in China: epidemiology, etiology, and prevention. Cancer Biology & Medicine. 2022;19(8):1121–38.CrossRef
3.
Zurück zum Zitat Qu R, Ma Y, Zhang Z, Fu W. Increasing burden of Colorectal cancer in China. Lancet Gastroenterol Hepatol. 2022;7(8):700.CrossRefPubMed Qu R, Ma Y, Zhang Z, Fu W. Increasing burden of Colorectal cancer in China. Lancet Gastroenterol Hepatol. 2022;7(8):700.CrossRefPubMed
4.
Zurück zum Zitat Biller LH, Schrag D. Diagnosis and treatment of metastatic Colorectal Cancer: a review. JAMA. 2021;325(7):669–85.CrossRefPubMed Biller LH, Schrag D. Diagnosis and treatment of metastatic Colorectal Cancer: a review. JAMA. 2021;325(7):669–85.CrossRefPubMed
5.
Zurück zum Zitat Global regional. National burden of Colorectal cancer and its risk factors, 1990–2019: a systematic analysis for the global burden of Disease Study 2019. Lancet Gastroenterol Hepatol. 2022;7(7):627–47.CrossRef Global regional. National burden of Colorectal cancer and its risk factors, 1990–2019: a systematic analysis for the global burden of Disease Study 2019. Lancet Gastroenterol Hepatol. 2022;7(7):627–47.CrossRef
6.
Zurück zum Zitat Thomas M, Sakoda LC, Hoffmeister M, Rosenthal EA, Lee JK, van Duijnhoven FJB, Platz EA, Wu AH, Dampier CH, de la Chapelle A, et al. Genome-wide modeling of polygenic risk score in Colorectal Cancer Risk. Am J Hum Genet. 2020;107(3):432–44.CrossRefPubMedCentralPubMed Thomas M, Sakoda LC, Hoffmeister M, Rosenthal EA, Lee JK, van Duijnhoven FJB, Platz EA, Wu AH, Dampier CH, de la Chapelle A, et al. Genome-wide modeling of polygenic risk score in Colorectal Cancer Risk. Am J Hum Genet. 2020;107(3):432–44.CrossRefPubMedCentralPubMed
7.
Zurück zum Zitat Meng Y, Du M, Gu D, Li C, Li S, Zhang Q, Ben S, Zhu Q, Xin J, Zhang Z, et al. Genome-Wide Association Analyses Identify CATSPERE as a mediator of Colorectal Cancer susceptibility and progression. Cancer Res. 2022;82(6):986–97.CrossRefPubMed Meng Y, Du M, Gu D, Li C, Li S, Zhang Q, Ben S, Zhu Q, Xin J, Zhang Z, et al. Genome-Wide Association Analyses Identify CATSPERE as a mediator of Colorectal Cancer susceptibility and progression. Cancer Res. 2022;82(6):986–97.CrossRefPubMed
8.
Zurück zum Zitat Bassiouni W, Ali MAM, Schulz R. Multifunctional intracellular matrix metalloproteinases: implications in Disease. FEBS J. 2021;288(24):7162–82.CrossRefPubMed Bassiouni W, Ali MAM, Schulz R. Multifunctional intracellular matrix metalloproteinases: implications in Disease. FEBS J. 2021;288(24):7162–82.CrossRefPubMed
10.
Zurück zum Zitat Juurikka K, Dufour A, Pehkonen K, Mainoli B, Campioni Rodrigues P, Solis N, Klein T, Nyberg P, Overall CM, Salo T, et al. MMP8 increases tongue carcinoma cell-cell adhesion and diminishes migration via cleavage of anti-adhesive FXYD5. Oncogenesis. 2021;10(5):44.CrossRefPubMedCentralPubMed Juurikka K, Dufour A, Pehkonen K, Mainoli B, Campioni Rodrigues P, Solis N, Klein T, Nyberg P, Overall CM, Salo T, et al. MMP8 increases tongue carcinoma cell-cell adhesion and diminishes migration via cleavage of anti-adhesive FXYD5. Oncogenesis. 2021;10(5):44.CrossRefPubMedCentralPubMed
11.
Zurück zum Zitat Reijonen P, Peltonen R, Tervahartiala T, Sorsa T, Isoniemi H. Serum Matrix Metalloproteinase-8 and Myeloperoxidase Predict Survival after Resection of Colorectal Liver metastases. Oncology. 2021;99(12):766–79.CrossRefPubMed Reijonen P, Peltonen R, Tervahartiala T, Sorsa T, Isoniemi H. Serum Matrix Metalloproteinase-8 and Myeloperoxidase Predict Survival after Resection of Colorectal Liver metastases. Oncology. 2021;99(12):766–79.CrossRefPubMed
12.
Zurück zum Zitat Pezeshkian Z, Nobili S, Peyravian N, Shojaee B, Nazari H, Soleimani H, Asadzadeh-Aghdaei H, Ashrafian Bonab M, Nazemalhosseini-Mojarad E, Mini E. Insights into the role of Matrix metalloproteinases in Precancerous conditions and in Colorectal Cancer. Cancers. 2021;13(24):6226.CrossRefPubMedCentralPubMed Pezeshkian Z, Nobili S, Peyravian N, Shojaee B, Nazari H, Soleimani H, Asadzadeh-Aghdaei H, Ashrafian Bonab M, Nazemalhosseini-Mojarad E, Mini E. Insights into the role of Matrix metalloproteinases in Precancerous conditions and in Colorectal Cancer. Cancers. 2021;13(24):6226.CrossRefPubMedCentralPubMed
13.
Zurück zum Zitat Bartnykaitė A, Savukaitytė A, Bekampytė J, Ugenskienė R, Laukaitienė D, Korobeinikova E, Gudaitienė J, Juozaitytė E. The role of Matrix Metalloproteinase single-nucleotide polymorphisms in the Clinicopathological properties of Breast Cancer. Biomedicines. 2022;10(8):1891.CrossRefPubMedCentralPubMed Bartnykaitė A, Savukaitytė A, Bekampytė J, Ugenskienė R, Laukaitienė D, Korobeinikova E, Gudaitienė J, Juozaitytė E. The role of Matrix Metalloproteinase single-nucleotide polymorphisms in the Clinicopathological properties of Breast Cancer. Biomedicines. 2022;10(8):1891.CrossRefPubMedCentralPubMed
14.
Zurück zum Zitat Murugan AK, Qasem E, Al-Hindi H, Alzahrani AS. Analysis of ALK, IDH1, IDH2 and MMP8 somatic mutations in differentiated thyroid cancers. Mol Clin Oncol. 2021;15(4):210.CrossRefPubMedCentralPubMed Murugan AK, Qasem E, Al-Hindi H, Alzahrani AS. Analysis of ALK, IDH1, IDH2 and MMP8 somatic mutations in differentiated thyroid cancers. Mol Clin Oncol. 2021;15(4):210.CrossRefPubMedCentralPubMed
15.
Zurück zum Zitat Insodaite R, Liutkevicius V, Uloza V, Solovejute R, Smalinskiene A. Association between MMP8 gene polymorphisms and laryngeal squamous cell carcinoma. Anticancer Res. 2020;40(4):2003–9.CrossRefPubMed Insodaite R, Liutkevicius V, Uloza V, Solovejute R, Smalinskiene A. Association between MMP8 gene polymorphisms and laryngeal squamous cell carcinoma. Anticancer Res. 2020;40(4):2003–9.CrossRefPubMed
16.
Zurück zum Zitat Tai J, Sun D, Wang X, Kang Z. Matrix metalloproteinase-8 rs11225395 polymorphism correlates with Colorectal cancer risk and survival in a Chinese Han population: a case-control study. Aging. 2020;12(19):19618–27.CrossRefPubMedCentralPubMed Tai J, Sun D, Wang X, Kang Z. Matrix metalloproteinase-8 rs11225395 polymorphism correlates with Colorectal cancer risk and survival in a Chinese Han population: a case-control study. Aging. 2020;12(19):19618–27.CrossRefPubMedCentralPubMed
17.
Zurück zum Zitat Deng Y, Ke TW, Wang YC, Chin YT, Yueh TC, Hung YC, Mong MC, Yang YC, Chang WS, Shen TC, et al. Impact of Matrix Metalloproteinase-8 genotypes on Colorectal Cancer Risk in Taiwan. Anticancer Res. 2023;43(9):3979–85.CrossRefPubMed Deng Y, Ke TW, Wang YC, Chin YT, Yueh TC, Hung YC, Mong MC, Yang YC, Chang WS, Shen TC, et al. Impact of Matrix Metalloproteinase-8 genotypes on Colorectal Cancer Risk in Taiwan. Anticancer Res. 2023;43(9):3979–85.CrossRefPubMed
18.
Zurück zum Zitat Wang K, Zhou Y, Li G, Wen X, Kou Y, Yu J, He H, Zhao Q, Xue F, Wang J, et al. MMP8 and MMP9 gene polymorphisms were associated with Breast cancer risk in a Chinese Han population. Sci Rep. 2018;8(1):13422.CrossRefPubMedCentralPubMed Wang K, Zhou Y, Li G, Wen X, Kou Y, Yu J, He H, Zhao Q, Xue F, Wang J, et al. MMP8 and MMP9 gene polymorphisms were associated with Breast cancer risk in a Chinese Han population. Sci Rep. 2018;8(1):13422.CrossRefPubMedCentralPubMed
19.
Zurück zum Zitat Dvornyk V, Ponomarenko I, Minyaylo O, Reshetnikov E, Churnosov M. Association of the functionally significant polymorphisms of the MMP9 gene with H. Pylori-positive gastric Ulcer in the caucasian population of Central Russia. PLoS ONE. 2021;16(9):e0257060.CrossRefPubMedCentralPubMed Dvornyk V, Ponomarenko I, Minyaylo O, Reshetnikov E, Churnosov M. Association of the functionally significant polymorphisms of the MMP9 gene with H. Pylori-positive gastric Ulcer in the caucasian population of Central Russia. PLoS ONE. 2021;16(9):e0257060.CrossRefPubMedCentralPubMed
20.
Zurück zum Zitat Zhao Y, Zhang Q, Zhang X, Zhang Y, Lu Y, Ma X, Li W, Niu X, Zhang G, Chang M, et al. The roles of MMP8/MMP10 polymorphisms in ischemic Stroke susceptibility. Brain and Behavior. 2022;12(12):e2797.CrossRefPubMedCentralPubMed Zhao Y, Zhang Q, Zhang X, Zhang Y, Lu Y, Ma X, Li W, Niu X, Zhang G, Chang M, et al. The roles of MMP8/MMP10 polymorphisms in ischemic Stroke susceptibility. Brain and Behavior. 2022;12(12):e2797.CrossRefPubMedCentralPubMed
21.
Zurück zum Zitat Oeth P, del Mistro G, Marnellos G, Shi T, van den Boom D. Qualitative and quantitative genotyping using single base primer extension coupled with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MassARRAY). Methods in Molecular Biology (Clifton NJ). 2009;578:307–43.CrossRef Oeth P, del Mistro G, Marnellos G, Shi T, van den Boom D. Qualitative and quantitative genotyping using single base primer extension coupled with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MassARRAY). Methods in Molecular Biology (Clifton NJ). 2009;578:307–43.CrossRef
22.
Zurück zum Zitat Ellis JA, Ong B. The MassARRAY(®) system for targeted SNP genotyping. Methods in Molecular Biology (Clifton NJ). 2017;1492:77–94.CrossRef Ellis JA, Ong B. The MassARRAY(®) system for targeted SNP genotyping. Methods in Molecular Biology (Clifton NJ). 2017;1492:77–94.CrossRef
23.
Zurück zum Zitat Wacholder S, Chanock S, Garcia-Closas M, El Ghormli L, Rothman N. Assessing the probability that a positive report is false: an approach for molecular epidemiology studies. J Natl Cancer Inst. 2004;96(6):434–42.CrossRefPubMedCentralPubMed Wacholder S, Chanock S, Garcia-Closas M, El Ghormli L, Rothman N. Assessing the probability that a positive report is false: an approach for molecular epidemiology studies. J Natl Cancer Inst. 2004;96(6):434–42.CrossRefPubMedCentralPubMed
24.
Zurück zum Zitat Hagman H, Bendahl PO, Lidfeldt J, Belting M, Johnsson A. Protein array profiling of circulating angiogenesis-related factors during bevacizumab containing treatment in metastatic Colorectal cancer. PLoS ONE. 2018;13(12):e0209838.CrossRefPubMedCentralPubMed Hagman H, Bendahl PO, Lidfeldt J, Belting M, Johnsson A. Protein array profiling of circulating angiogenesis-related factors during bevacizumab containing treatment in metastatic Colorectal cancer. PLoS ONE. 2018;13(12):e0209838.CrossRefPubMedCentralPubMed
25.
Zurück zum Zitat Ochman B, Mielcarska S, Kula A, Dawidowicz M, Robotycka J, Piecuch J, Szrot M, Dzięgielewska-Gęsiak S, Muc-Wierzgoń M, Waniczek D, et al. Do elevated YKL-40 levels drive the immunosuppressive Tumor Microenvironment in Colorectal Cancer? Assessment of the Association of the expression of YKL-40, MMP-8, IL17A, and PD-L1 with coexisting type 2 Diabetes, obesity, and active Smoking. Curr Issues Mol Biol. 2023;45(4):2781–97.CrossRefPubMedCentralPubMed Ochman B, Mielcarska S, Kula A, Dawidowicz M, Robotycka J, Piecuch J, Szrot M, Dzięgielewska-Gęsiak S, Muc-Wierzgoń M, Waniczek D, et al. Do elevated YKL-40 levels drive the immunosuppressive Tumor Microenvironment in Colorectal Cancer? Assessment of the Association of the expression of YKL-40, MMP-8, IL17A, and PD-L1 with coexisting type 2 Diabetes, obesity, and active Smoking. Curr Issues Mol Biol. 2023;45(4):2781–97.CrossRefPubMedCentralPubMed
26.
Zurück zum Zitat Forsberg A, Westerberg M, Metcalfe C, Steele R, Blom J, Engstrand L, Fritzell K, Hellström M, Levin L, Löwbeer C, et al. Once-only colonoscopy or two rounds of faecal immunochemical testing 2 years apart for Colorectal cancer screening (SCREESCO): preliminary report of a randomised controlled trial. Lancet Gastroenterol Hepatol. 2022;7(6):513–21.CrossRefPubMed Forsberg A, Westerberg M, Metcalfe C, Steele R, Blom J, Engstrand L, Fritzell K, Hellström M, Levin L, Löwbeer C, et al. Once-only colonoscopy or two rounds of faecal immunochemical testing 2 years apart for Colorectal cancer screening (SCREESCO): preliminary report of a randomised controlled trial. Lancet Gastroenterol Hepatol. 2022;7(6):513–21.CrossRefPubMed
27.
Zurück zum Zitat Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, Cercek A, Smith RA, Jemal A. Colorectal cancer statistics, 2020. Cancer J Clin. 2020;70(3):145–64.CrossRef Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, Cercek A, Smith RA, Jemal A. Colorectal cancer statistics, 2020. Cancer J Clin. 2020;70(3):145–64.CrossRef
28.
Zurück zum Zitat Bull CJ, Bell JA, Murphy N, Sanderson E, Davey Smith G, Timpson NJ, Banbury BL, Albanes D, Berndt SI, Bézieau S, et al. Adiposity, metabolites, and Colorectal cancer risk: mendelian randomization study. BMC Med. 2020;18(1):396.CrossRefPubMedCentralPubMed Bull CJ, Bell JA, Murphy N, Sanderson E, Davey Smith G, Timpson NJ, Banbury BL, Albanes D, Berndt SI, Bézieau S, et al. Adiposity, metabolites, and Colorectal cancer risk: mendelian randomization study. BMC Med. 2020;18(1):396.CrossRefPubMedCentralPubMed
29.
Zurück zum Zitat Larsson SC, Carter P, Kar S, Vithayathil M, Mason AM, Michaëlsson K, Burgess S. Smoking, alcohol consumption, and cancer: a mendelian randomisation study in UK Biobank and international genetic consortia participants. PLoS Med. 2020;17(7):e1003178.CrossRefPubMedCentralPubMed Larsson SC, Carter P, Kar S, Vithayathil M, Mason AM, Michaëlsson K, Burgess S. Smoking, alcohol consumption, and cancer: a mendelian randomisation study in UK Biobank and international genetic consortia participants. PLoS Med. 2020;17(7):e1003178.CrossRefPubMedCentralPubMed
30.
Zurück zum Zitat Botteri E, Borroni E, Sloan EK, Bagnardi V, Bosetti C, Peveri G, Santucci C, Specchia C, van den Brandt P, Gallus S, et al. Smoking and Colorectal Cancer risk, overall and by Molecular subtypes: a Meta-analysis. Am J Gastroenterol. 2020;115(12):1940–9.CrossRefPubMed Botteri E, Borroni E, Sloan EK, Bagnardi V, Bosetti C, Peveri G, Santucci C, Specchia C, van den Brandt P, Gallus S, et al. Smoking and Colorectal Cancer risk, overall and by Molecular subtypes: a Meta-analysis. Am J Gastroenterol. 2020;115(12):1940–9.CrossRefPubMed
31.
Zurück zum Zitat Sapkota R, Zakaria J, Glenn E, Richard H, Rimawi A, Tobi M, McVicker B. Alcohol Use and the risk of colorectal liver Metastasis: a systematic mapping review. Biology. 2023;12(2):257.CrossRefPubMedCentralPubMed Sapkota R, Zakaria J, Glenn E, Richard H, Rimawi A, Tobi M, McVicker B. Alcohol Use and the risk of colorectal liver Metastasis: a systematic mapping review. Biology. 2023;12(2):257.CrossRefPubMedCentralPubMed
Metadaten
Titel
Association study for the role of MMP8 gene polymorphisms in Colorectal cancer susceptibility
verfasst von
Shuyong Yu
Jiajia Cheng
Ping Li
Le Tian
Zhuang Chen
Zhaowei Chen
Yongyu Li
Jian Song
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2023
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-11662-z

Weitere Artikel der Ausgabe 1/2023

BMC Cancer 1/2023 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Update Onkologie

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