Skip to main content
Erschienen in: Diagnostic Pathology 1/2019

Open Access 01.12.2019 | Research

High expression of MRE11 correlates with poor prognosis in gastric carcinoma

verfasst von: Junqing Li, Taiqiang Su, Liang Yang, Changhua Zhang, Yulong He

Erschienen in: Diagnostic Pathology | Ausgabe 1/2019

Abstract

Background

MRE11, a protein known to play a vital role in DNA double-strand break repair, is associated with the prognosis of a variety of tumours, but there are few studies regarding the role of MRE11 in gastric carcinoma (GC). The present study aimed to explore the clinicopathological significance and prognostic value of MRE11 expression in GC.

Methods

Data from the TCGA, GEO and Oncomine databases were analysed to assess MRE11 mRNA levels in GC. The prognostic role of the level of MRE11 mRNA was examined via the Kaplan-Meier plotter. MRE11 protein expression in tumour tissues from 155 GC patients was analysed by immunohistochemistry. Relationships between MRE11 expression and clinicopathological characteristics, overall survival (OS) and recurrence-free survival (RFS) were evaluated by Cox proportional hazards regression models and Kaplan-Meier survival curves.

Results

The results of bioinformatics analysis showed that MRE11 mRNA levels in GC tissues were higher than those in normal tissues (P < 0.01). Tissue microarray analysis showed that MRE11 protein expression was increased in GC tissues (P < 0.001), and MRE11 overexpression in GC tissues was significantly related to lymph node metastasis (P < 0.05), distant metastasis (P < 0.05) and tumour-node-metastasis stage (P < 0.05). Kaplan-Meier analyses showed that patients with GC who exhibited MRE11 overexpression had worse OS and RFS. According to Cox proportional hazards analyses, MRE11 overexpression was an independent prognostic factor for OS and RFS in these GC patients.

Conclusions

MRE11 overexpression is significantly associated with poor prognosis, and MRE11 may serve as a prognostic biomarker in GC patients.
Hinweise
Junqing Li and Taiqiang Su contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CIs
Confidence intervals
DSBs
DNA double-strand breaks
HR
Homologous recombination
HRs
Hazard ratios
IHC
immunohistochemical
MRN
MRE11/RAD50/NBS1
NHEJ
Non-homologous end joining
OS
Overall survival
RFS
Recurrence-free survival

Introduction

Gastric carcinoma (GC) is the fifth most common malignant tumour worldwide and frequently leads to death [1, 2]. Surgery is considered to be the only curative treatment, and there is no satisfactory treatment, including molecular-targeted therapy and immunotherapy, for patients with recurrent or unresectable GC [35]. Overall, there is a need to explore novel mechanisms of GC development to improve the prognosis of GC patients.
DNA double-strand breaks (DSBs) are a major threat to genomic integrity and cause chromosome breaks, deletions, and translocations in cancer cells [6, 7]. DSBs are repaired by the homologous recombination (HR) or non-homologous end joining (NHEJ) pathway [7, 8]. The MRE11/RAD50/NBS1 (MRN) complex plays a central role in most aspects of the cellular response to DSBs, including HR, NHEJ, telomere activity and DNA damage checkpoint activation [911]. As a core protein of the MRN repair complex, MRE11 may also be associated with the prognosis and development of human cancers. In addition, MRE11 protein expression was proved to be a predictive factor associated with survival following bladder cancer radiotherapy [12]. Furthermore, a randomized clinical trial showed that MRE11 deficiency is associated with improved long-term disease-free survival and overall survival (OS) in a subset of stage III colon cancer patients [13], and Yuan SS et al. showed that high MRE11 expression is associated with increased malignant behaviour in breast cancer [14]. Nonetheless, it remains unclear whether MRE11 regulates the progression and development of GC.
Therefore, in this study, we assessed MRE11 expression in GC specimens and explored its association with clinicopathologic parameters and long-term OS and recurrence-free survival (RFS) in GC patients.

Methods

Patients

Surgically treated GC patients (n = 155) with confirmed pathology at the First Affiliated Hospital of Sun Yat-sen University (FAHSYSU) between 2004 and 2007 were randomly chosen. Follow-up was terminated by December 2017. For this study, we excluded patients who received chemotherapy or other treatment before sampling or were lost during follow-up. We reviewed the patients’ clinicopathological characteristics, including gender, age, tumour size, tumour location, Bormann classification, and differentiation. We performed tumour staging for patients according to the 8th Edition of the American Joint Cancer Committee TNM classifications. Another 61 GC patients who had received surgical treatment in 2013 were randomly chosen for tissue microarray analysis.
Patient consent and ethical approval from the Institutional Review Board of FAHSYSU were obtained for this study.

Tissue microarrays

We collected adjacent normal and GC tissues of 61 patients from the FAHSYSU Department of Pathology, and these tissues were assembled into tissue microarrays (Servicebio, Wuhan, China) for immunohistochemical (IHC) staining.

Immunohistochemical staining

We obtained 155 paraffin-embedded GC specimens from the FAHSYSU Department of Pathology, and IHC staining of these specimens and the tissue microarrays were conducted as previously described [15] using an anti-MRE11 antibody (1:200; Sigma-Aldrich, Darmstadt, Germany).
Evaluation of the IHC results was performed by two independent investigators who were blind to the specimens, and scoring was determined using a semi-quantitative method [16]. Samples in which more than 10% of the tumour cells were stained were considered positive. The staining intensity was defined as follows: 0 (negative), 1 (weak), 2 (moderate), and 3 (strong). Negative and weak staining was considered to indicate low MRE11 expression, and moderate and strong staining was considered to indicate high MRE11 expression.

Bioinformatics analysis

We downloaded RNA-Seq data for GC from The Cancer Genome Atlas (TCGA), GEO (GSE139911) and Oncomine databases. We analysed the prognostic role of MRE11 mRNA levels using the Kaplan-Meier plotter.

Statistical analysis

Statistical analyses were performed using SPSS 17.0 (IBM, NY, USA). The chi-square test was employed for numerical data. Survival curves were generated using the Kaplan-Meier estimator. Hazard ratios (HRs) and 95% confidence intervals (CIs) were computed from univariate and multivariate Cox proportional hazards regression models to examine associations between prognosis and clinicopathological characteristics. P values less than 0.05 were considered statistically significant.

Results

Overexpression of MRE11 mRNA in GC tissues

Data from TCGA showed many genes to be up- or downregulated in GC samples relative to their expression in normal samples, with the MRE11 gene being significantly overexpressed in the former (Fig. 1a). Further analysis of unpaired GC and normal tissues from TCGA also indicated markedly upregulated expression of MRE11 mRNA in GC tissues (P < 0.001; Fig. 1b). The same results were found for paired GC and normal tissues from the GEO cohort (P < 0.01; Fig. 1c), and Oncomine cohort data were consistent (Table 1). Taken together, these findings indicate that MRE11 mRNA expression was upregulated in GC tissues.
Table 1
Oncomine analysis of MRE11 expression in GC (total of 5 GC cohorts)
Cohort
Sample (n)
t Test
Fold change
P
D’Errico et al. [17]
Gastric Mixed Adenocarcinoma (4) vs Normal (31)
6.160
2.953
3.34E-6
Cho et al. [18]
Diffuse Gastric Adenocarcinoma (31) vs Normal (19)
4.419
1.308
2.84E-5
Gastric Mixed Adenocarcinoma (10) vs Normal (19)
3.969
1.383
7.56E-4
Gastric Adenocarcinoma (4) vs Normal (19)
2.401
1.386
0.042
Gastric Intestinal-Type Adenocarcinoma (20) vs Normal (19)
2.340
1.200
0.013
Wang et al. [19]
Gastric Cancer (3) vs Normal (12)
2.958
1.668
0.003
Cui et al. [20]
Gastric Cancer (80) vs Normal (80)
1.882
1.283
0.031
Deng et al. [21]
Gastric Intestinal Type Adenocarcinoma (44) vs Normal (17)
2.575
1.022
0.007
Diffuse Gastric Adenocarcinoma (13) vs Normal (17)
2.163
1.011
0.018

Associations of MRE11 expression with clinical parameters in GC

To evaluate the relationship between MRE11 expression and clinicopathological characteristics in GC, we performed IHC staining to detect MRE11 expression in 155 paraffin-embedded GC specimens. As shown in Fig. 2, the MRE11 protein was mainly distributed in the nucleus. MRE11 expression was further measured using tissue microarrays containing adjacent normal and GC tissues from 61 patients, and MRE11 protein expression was found to be significantly higher in GC tissues than in adjacent normal tissues (Fig. 2c). As shown in Fig. 2d, there were 28 cases (18.1%) with an IHC score of 0, 45 (29.0%) with an IHC score of 1, 49 (31.6%) with an IHC score of 2, and 33 (21.3%) with an IHC score of 3. IHC scores of 0 and 1 were defined as low MRE11 expression (47.1%, 73/155); IHC scores of 2 and 3 were defined as high MRE11 expression (52.9%, 82/155).
The relationships between MRE11 expression and clinicopathological parameters are summarized in Table 2. MRE11 overexpression in GC tissues was significantly related to lymph node metastasis (P < 0.05), distant metastasis (P < 0.05) and tumour-node-metastasis (TNM) stage (P < 0.05).
Table 2
Associations of MRE11 expression with clinical parameters in GC
Characteristic
No.
MRE11 Expression
χ2
P
  
Low(N = 73)
High(N = 82)
Value
Value
Age
 
59.84 ± 10.87
56.92 ± 13.24
  
<60y
78
35
43
0.312
0.576
≥60y
77
38
39
Gender
 
Man
99
48
51
0.212
0.645
Female
56
25
31
Tumour location
     
Proximal
33
14
19
2.575
0.462
Middle
29
17
12
Distal
58
28
30
More than 2
35
14
21
Tumour size
     
< 5 cm
68
34
34
0.410
0.522
≥5 cm
87
39
48
Histologic type
     
Adenocarcinoma (NOS)
132
63
69
1.868
0.600
Signet ring
9
4
5
Mucinous
12
6
6
Undifferentiated
2
0
2
Bornmann classification
     
I
8
2
6
2.107
0.550
II
34
18
16
III
97
45
52
IV
16
8
8
Differentiation
 
Well
3
2
1
1.517
0.468
Moderate
35
19
16
Poor
117
52
65
Depth of invasion
 
T1
15
10
5
4.146
0.246
T2
13
8
5
T3
84
36
48
T4
43
19
24
Lymph node metastasis
 
N0
45
26
19
8.269
0.041
N1
54
25
29
N2
23
5
18
N3
33
17
16
Distant metastasis
 
M0
126
65
61
5.450
0.020
M1
29
8
21
Tumour-Node-Metastasis stage
 
I + II
49
30
19
5.740
0.017
III + IV
106
43
63
CEA level (μg/L)
     
< 5
135
64
71
0.041
0.840
≥5
20
9
11

High MRE11 expression predicted worse survival in GC

To define the prognostic role of MRE11 expression in GC, we first analysed data using the Kaplan-Meier plotter. Patients with high levels of MRE11 mRNA had worse OS (Fig. 3a) and RFS (Fig. 3b) than those with low MRE11 levels. Next, the prognostic value of MRE11 protein expression in our cohort was assessed by Kaplan-Meier analysis. The follow-up period of the 155 GC patients ranged from 2 to 156 months, with a mean survival time of 62.56 ± 4.64 months. The mean survival times of patients with low and high MRE11 expression were 81.83 ± 7.06 and 45.37 ± 5.47 months, respectively. The combined 5-year OS rate was 36.6%; the 5-year OS rate was 54.1% in the low MRE11 expression group and 20.9% in the high MRE11 expression group. Our data indicated that high MRE11 expression was associated with worse OS (P < 0.001, Fig. 3c) and RFS (P < 0.001, Fig. 3d). Furthermore, we defined the prognostic value of MRE11 expression in early (TNM stages I and II) and advanced (TNM stages III and IV) GC, and the results showed that high MRE11 expression was associated with worse OS (P < 0.05, Fig. 3e and g) and RFS (P < 0.05, Fig. 3f and h) in both early and advanced disease.
We also performed univariate and multivariate analyses to assess the ability of MRE11 expression to predict the prognosis of patients with GC. As shown in Table 3, univariate analysis revealed that certain clinical variables were significantly associated with OS, with multivariate analysis demonstrating that MRE11 expression (P < 0.01) was an independent predictor of OS in GC patients. In addition, MRE11 expression was an independent predictor of RFS (P < 0.01) in GC patients (Table 4). Taken together, our results indicate that MRE11 expression was an independent prognostic factor for the GC patients investigated and that MRE11 might serve as a molecular marker for GC prognosis.
Table 3
Cox proportional-hazard regression analysis for Overall Survival
Characteristic
Univariate analysis
Multivariate analysis
P-Value
HR
95.0% CI for Exp(B)
P-Value
HR
95.0% CI for Exp(B)
Lower
Upper
Lower
Upper
Gender
0.016
1.582
1.089
2.300
0.029
1.546
1.045
2.286
Age
0.325
0.833
0.580
1.198
    
Tumour location
0.497
1.064
0.889
1.274
    
Tumour size
0.001
1.898
1.303
2.763
    
Histologic type
0.335
1.174
0.847
1.628
    
Bornmann classification
0.008
1.460
1.103
1.932
    
Differentiation
0.034
1.550
1.033
2.326
    
Depth of invasion
0.000
1.757
1.382
2.235
    
Lymph node metastasis
0.000
1.521
1.289
1.794
    
Distant metastasis
0.000
3.509
2.267
5.429
    
Tumour-Node-Metastasis stage
0.000
2.134
1.698
2.681
0.016
1.735
1.107
2.719
CEA
0.019
1.832
1.104
3.040
    
MRE11 expression
0.000
2.069
1.422
3.012
0.001
1.900
1.297
2.782
Table 4
Cox proportional-hazard regression analysis for Recurrence Free Survival
Characteristic
Univariate analysis
Multivariate analysis
P-Value
HR
95.0% CI for Exp(B)
P-Value
HR
95.0% CI for Exp(B)
Lower
Upper
Lower
Upper
Gender
0.045
1.497
1.009
2.221
    
Age
0.404
0.850
0.581
1.245
    
Tumour location
0.901
0.988
0.820
1.191
    
Tumour size
0.006
1.735
1.173
2.565
    
Histologic type
0.464
1.144
0.798
1.641
    
Bornmann classification
0.025
1.402
1.043
1.884
    
Differentiation
0.036
1.589
1.030
2.452
    
Depth of invasion
0.001
1.526
1.199
1.940
    
Lymph node metastasis
0.001
1.331
1.116
1.586
    
Distant metastasis
0.000
3.108
1.965
4.915
    
Tumour-Node-Metastasis stage
0.000
1.928
1.527
2.433
0.005
1.969
1.232
3.147
CEA
0.042
1.744
1.021
2.979
    
MRE11 expression
0.000
2.220
1.489
3.309
0.001
2.041
1.358
3.068

Discussion

The results of this study show that MRE11 overexpression is significantly associated with poor prognosis and that MRE11 is a potential prognostic biomarker in patients with GC. First, by analysing TCGA, GEO and Oncomine data, we demonstrated that the MRE11 mRNA levels were significantly higher in GC tissues than in normal tissues. Second, using the Kaplan-Meier plotter, we found that patients with high MRE11 expression had poor OS and RFS. Third, we proved that MRE11 protein expression was significantly higher in GC tissues than in adjacent normal tissues, as based on tissue microarrays derived from 61 patients. Furthermore, IHC staining of tissues from 155 GC patients verified that MRE11 overexpression was associated with poor OS and RFS, a finding that is consistent with that of a previous study [22]. Finally, multivariate Cox regression analysis demonstrated MRE11 expression to be an independent prognostic factor for OS and RFS in GC patients.
The MRE11 gene encodes a nuclear protein involved in HR, telomere length maintenance, and DSB repair, and the MRN complex is required for NHEJ [68, 11]. Previous studies have reported that deficiency of the MRN complex may sensitize cancer cells to treatment with PARP inhibitors and might serve as a predictive biomarker for the efficacy of PARP inhibitor therapy [2325]. MRE11 protein expression is also associated with the prognosis and development of human cancers, such as bladder cancer, colon cancer, and breast cancer [1214]. Altan B et al. [22] found that high expression of MRE11 is associated with poor OS in GC, and we further proved that MRE11 overexpression is associated with poor RFS and is an independent prognostic factor for OS and RFS in GC. Some studies have suggested that high expression of MRE11 in tumour cells enhances DSB repair, leading to increased local recurrence and reduced survival rates [14, 26].
The findings of this study also demonstrate that MRE11 overexpression is significantly related to lymph node metastasis, distant metastasis and TNM stage. However, we did not explore the molecular mechanism of MRE11 in GC, which is a limitation of this study. Previous studies have shown that MRE11 overexpression in breast cancer cells leads to cell proliferation by stimulating STAT3 signalling and enhances migration and invasion capabilities through activation of MMP-2 and MMP-9 [14]. In addition, activation of the STAT3-MMP axis has been reported to promote cancer cell invasiveness and metastasis [2729]. Based on these results, we can conclude that the STAT3-MMP axis may be involved in the molecular mechanism of MRE11 in GC. In future work, we will verify the role of this molecular pathway in GC. As tumour cells overexpressing MRE11 exhibit a poor response to radiotherapy and chemotherapy and MRE11 deficiency leads to inhibition of DSB repair and enhances radio- and chemosensitivity in tumour cells [12, 14, 3032], MRE11 is a potential target for increasing radio- and chemosensitivity.
In conclusion, MRE11 overexpression is significantly associated with poor prognosis, and MRE11 may act as a prognostic biomarker in patients with GC. MRE11 may also serve as a target molecule for chemoradiotherapy.

Acknowledgments

The authors thank Ertao Zhai MD, Junbin Liao MD and Lingna Deng MD for their help in this study.
Lingna Deng MD, Department of Pathology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China. Email: denglna@163.​com.
Ethical approval was obtained by the First Affiliated Hospital, Sun Yat-sen University. Approval Letter for Research Protocol: No. 226 [2017].

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108.CrossRef Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108.CrossRef
2.
Zurück zum Zitat Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86.CrossRef Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86.CrossRef
3.
Zurück zum Zitat Cidon EU, Ellis SG, Inam Y, Adeleke S, Zarif S, Geldart T. Molecular targeted agents for gastric cancer: a step forward towards personalized therapy. Cancers (Basel). 2013;5(1):64–91.CrossRef Cidon EU, Ellis SG, Inam Y, Adeleke S, Zarif S, Geldart T. Molecular targeted agents for gastric cancer: a step forward towards personalized therapy. Cancers (Basel). 2013;5(1):64–91.CrossRef
4.
Zurück zum Zitat Song Z, Wu Y, Yang J, Yang D, Fang X. Progress in the treatment of advanced gastric cancer. Tumour Biol. 2017;39(7):1010428317714626.PubMed Song Z, Wu Y, Yang J, Yang D, Fang X. Progress in the treatment of advanced gastric cancer. Tumour Biol. 2017;39(7):1010428317714626.PubMed
5.
Zurück zum Zitat Vrána D, Matzenauer M, Neoral Č, Aujeský R, Vrba R, Melichar B, et al. From tumor immunology to immunotherapy in gastric and esophageal Cancer. Int J Mol Sci. 2018;20:1.CrossRef Vrána D, Matzenauer M, Neoral Č, Aujeský R, Vrba R, Melichar B, et al. From tumor immunology to immunotherapy in gastric and esophageal Cancer. Int J Mol Sci. 2018;20:1.CrossRef
6.
Zurück zum Zitat Powell SN, Bindra RS. Targeting the DNA damage response for cancer therapy. DNA Repair (Amst). 2009;8(9):1153–65.CrossRef Powell SN, Bindra RS. Targeting the DNA damage response for cancer therapy. DNA Repair (Amst). 2009;8(9):1153–65.CrossRef
7.
Zurück zum Zitat Rodgers K, McVey M. Error-prone repair of DNA double-Strand breaks. J Cell Physiol. 2016;231(1):15–24.CrossRef Rodgers K, McVey M. Error-prone repair of DNA double-Strand breaks. J Cell Physiol. 2016;231(1):15–24.CrossRef
8.
Zurück zum Zitat Helleday T, Lo J, van Gent DC, Engelward BP. DNA double-strand break repair: from mechanistic understanding to cancer treatment. DNA Repair (Amst). 2007;6(7):923–35.CrossRef Helleday T, Lo J, van Gent DC, Engelward BP. DNA double-strand break repair: from mechanistic understanding to cancer treatment. DNA Repair (Amst). 2007;6(7):923–35.CrossRef
9.
Zurück zum Zitat Assenmacher N, Hopfner KP. MRE11/RAD50/NBS1: complex activities. Chromosoma. 2004;113(4):157–66.CrossRef Assenmacher N, Hopfner KP. MRE11/RAD50/NBS1: complex activities. Chromosoma. 2004;113(4):157–66.CrossRef
10.
Zurück zum Zitat Williams RS, Williams JS, Tainer JA. Mre11-Rad50-Nbs1 is a keystone complex connecting DNA repair machinery, double-strand break signaling, and the chromatin template. Biochem Cell Biol. 2007;85(4):509–20.CrossRef Williams RS, Williams JS, Tainer JA. Mre11-Rad50-Nbs1 is a keystone complex connecting DNA repair machinery, double-strand break signaling, and the chromatin template. Biochem Cell Biol. 2007;85(4):509–20.CrossRef
11.
Zurück zum Zitat Kavitha CV, Choudhary B, Raghavan SC, Muniyappa K. Differential regulation of MRN (Mre11-Rad50-Nbs1) complex subunits and telomerase activity in cancer cells. Biochem Biophys Res Commun. 2010;399(4):575–80.CrossRef Kavitha CV, Choudhary B, Raghavan SC, Muniyappa K. Differential regulation of MRN (Mre11-Rad50-Nbs1) complex subunits and telomerase activity in cancer cells. Biochem Biophys Res Commun. 2010;399(4):575–80.CrossRef
12.
Zurück zum Zitat Choudhury A, Nelson LD, Teo MT, Chilka S, Bhattarai S, Johnston CF, et al. MRE11 expression is predictive of cause-specific survival following radical radiotherapy for muscle-invasive bladder cancer. Cancer Res. 2010;70(18):7017–26.CrossRef Choudhury A, Nelson LD, Teo MT, Chilka S, Bhattarai S, Johnston CF, et al. MRE11 expression is predictive of cause-specific survival following radical radiotherapy for muscle-invasive bladder cancer. Cancer Res. 2010;70(18):7017–26.CrossRef
13.
Zurück zum Zitat Pavelitz T, Renfro L, Foster NR, Caracol A, Welsch P, Lao VV, et al. MRE11-deficiency associated with improved long-term disease free survival and overall survival in a subset of stage III colon cancer patients in randomized CALGB 89803 trial. PLoS One. 2014;9(10):e108483.CrossRef Pavelitz T, Renfro L, Foster NR, Caracol A, Welsch P, Lao VV, et al. MRE11-deficiency associated with improved long-term disease free survival and overall survival in a subset of stage III colon cancer patients in randomized CALGB 89803 trial. PLoS One. 2014;9(10):e108483.CrossRef
14.
Zurück zum Zitat Yuan SS, Hou MF, Hsieh YC, Huang CY, Lee YC, Chen YJ, et al. Role of MRE11 in cell proliferation, tumor invasion, and DNA repair in breast cancer. J Natl Cancer Inst. 2012;104(19):1485–502.CrossRef Yuan SS, Hou MF, Hsieh YC, Huang CY, Lee YC, Chen YJ, et al. Role of MRE11 in cell proliferation, tumor invasion, and DNA repair in breast cancer. J Natl Cancer Inst. 2012;104(19):1485–502.CrossRef
15.
Zurück zum Zitat Yang L, Chen Z, Xiong W, Ren H, Zhai E, Xu K, et al. High expression of SLC17A9 correlates with poor prognosis in colorectal cancer. Hum Pathol. 2018;84:62–70.CrossRef Yang L, Chen Z, Xiong W, Ren H, Zhai E, Xu K, et al. High expression of SLC17A9 correlates with poor prognosis in colorectal cancer. Hum Pathol. 2018;84:62–70.CrossRef
16.
Zurück zum Zitat Zhai E, Liang W, Lin Y, Huang L, He X, Cai S, et al. HSP70/HSP90-organizing protein contributes to gastric Cancer progression in an autocrine fashion and predicts poor survival in gastric Cancer. Cell Physiol Biochem. 2018;47(2):879–92.CrossRef Zhai E, Liang W, Lin Y, Huang L, He X, Cai S, et al. HSP70/HSP90-organizing protein contributes to gastric Cancer progression in an autocrine fashion and predicts poor survival in gastric Cancer. Cell Physiol Biochem. 2018;47(2):879–92.CrossRef
17.
Zurück zum Zitat D'Errico M, de Rinaldis E, Blasi MF, Viti V, Falchetti M, Calcagnile A, et al. Genome-wide expression profile of sporadic gastric cancers with microsatellite instability. Eur J Cancer. 2009;45(3):461–9.CrossRef D'Errico M, de Rinaldis E, Blasi MF, Viti V, Falchetti M, Calcagnile A, et al. Genome-wide expression profile of sporadic gastric cancers with microsatellite instability. Eur J Cancer. 2009;45(3):461–9.CrossRef
18.
Zurück zum Zitat Cho JY, Lim JY, Cheong JH, Park YY, Yoon SL, Kim SM, et al. Gene expression signature-based prognostic risk score in gastric cancer. Clin Cancer Res. 2011;17(7):1850–7.CrossRef Cho JY, Lim JY, Cheong JH, Park YY, Yoon SL, Kim SM, et al. Gene expression signature-based prognostic risk score in gastric cancer. Clin Cancer Res. 2011;17(7):1850–7.CrossRef
19.
Zurück zum Zitat Wang Q, Wen YG, Li DP, Xia J, Zhou CZ, Yan DW, et al. Upregulated INHBA expression is associated with poor survival in gastric cancer. Med Oncol. 2012;29(1):77–83.CrossRef Wang Q, Wen YG, Li DP, Xia J, Zhou CZ, Yan DW, et al. Upregulated INHBA expression is associated with poor survival in gastric cancer. Med Oncol. 2012;29(1):77–83.CrossRef
20.
Zurück zum Zitat Cui J, Chen Y, Chou WC, Sun L, Chen L, Suo J, et al. An integrated transcriptomic and computational analysis for biomarker identification in gastric cancer. Nucleic Acids Res. 2011;39(4):1197–207.CrossRef Cui J, Chen Y, Chou WC, Sun L, Chen L, Suo J, et al. An integrated transcriptomic and computational analysis for biomarker identification in gastric cancer. Nucleic Acids Res. 2011;39(4):1197–207.CrossRef
21.
Zurück zum Zitat Deng N, Goh LK, Wang H, Das K, Tao J, Tan IB, et al. A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut. 2012;61(5):673–84.CrossRef Deng N, Goh LK, Wang H, Das K, Tao J, Tan IB, et al. A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut. 2012;61(5):673–84.CrossRef
22.
Zurück zum Zitat Altan B, Yokobori T, Ide M, Bai T, Yanoma T, Kimura A, et al. High expression of MRE11-RAD50-NBS1 is associated with poor prognosis and Chemoresistance in gastric Cancer. Anticancer Res. 2016;36(10):5237–47.CrossRef Altan B, Yokobori T, Ide M, Bai T, Yanoma T, Kimura A, et al. High expression of MRE11-RAD50-NBS1 is associated with poor prognosis and Chemoresistance in gastric Cancer. Anticancer Res. 2016;36(10):5237–47.CrossRef
23.
Zurück zum Zitat Oplustilova L, Wolanin K, Mistrik M, Korinkova G, Simkova D, Bouchal J, et al. Evaluation of candidate biomarkers to predict cancer cell sensitivity or resistance to PARP-1 inhibitor treatment. Cell Cycle. 2012;11(20):3837–50.CrossRef Oplustilova L, Wolanin K, Mistrik M, Korinkova G, Simkova D, Bouchal J, et al. Evaluation of candidate biomarkers to predict cancer cell sensitivity or resistance to PARP-1 inhibitor treatment. Cell Cycle. 2012;11(20):3837–50.CrossRef
24.
Zurück zum Zitat Koppensteiner R, Samartzis EP, Noske A, von Teichman A, Dedes I, Gwerder M, et al. Effect of MRE11 loss on PARP-inhibitor sensitivity in endometrial cancer in vitro. PLoS One. 2014;9(6):e100041.CrossRef Koppensteiner R, Samartzis EP, Noske A, von Teichman A, Dedes I, Gwerder M, et al. Effect of MRE11 loss on PARP-inhibitor sensitivity in endometrial cancer in vitro. PLoS One. 2014;9(6):e100041.CrossRef
25.
Zurück zum Zitat Brandt S, Samartzis EP, Zimmermann AK, Fink D, Moch H, Noske A, et al. Lack of MRE11-RAD50-NBS1 (MRN) complex detection occurs frequently in low-grade epithelial ovarian cancer. BMC Cancer. 2017;17(1):44.CrossRef Brandt S, Samartzis EP, Zimmermann AK, Fink D, Moch H, Noske A, et al. Lack of MRE11-RAD50-NBS1 (MRN) complex detection occurs frequently in low-grade epithelial ovarian cancer. BMC Cancer. 2017;17(1):44.CrossRef
26.
Zurück zum Zitat LeScodan R, Cizeron-Clairac G, Fourme E, Meseure D, Vacher S, Spyratos F, et al. DNA repair gene expression and risk of locoregional relapse in breast cancer patients. Int J Radiat Oncol Biol Phys. 2010;78(2):328–36.CrossRef LeScodan R, Cizeron-Clairac G, Fourme E, Meseure D, Vacher S, Spyratos F, et al. DNA repair gene expression and risk of locoregional relapse in breast cancer patients. Int J Radiat Oncol Biol Phys. 2010;78(2):328–36.CrossRef
27.
Zurück zum Zitat Huang C, Cao J, Huang KJ, Zhang F, Jiang T, Zhu L, et al. Inhibition of STAT3 activity with AG490 decreases the invasion of human pancreatic cancer cells in vitro. Cancer Sci. 2006;97(12):1417–23.CrossRef Huang C, Cao J, Huang KJ, Zhang F, Jiang T, Zhu L, et al. Inhibition of STAT3 activity with AG490 decreases the invasion of human pancreatic cancer cells in vitro. Cancer Sci. 2006;97(12):1417–23.CrossRef
28.
Zurück zum Zitat Xie TX, Wei D, Liu M, Gao AC, Ali-Osman F, Sawaya R, et al. Stat3 activation regulates the expression of matrix metalloproteinase-2 and tumor invasion and metastasis. Oncogene. 2004;23(20):3550–60.CrossRef Xie TX, Wei D, Liu M, Gao AC, Ali-Osman F, Sawaya R, et al. Stat3 activation regulates the expression of matrix metalloproteinase-2 and tumor invasion and metastasis. Oncogene. 2004;23(20):3550–60.CrossRef
29.
Zurück zum Zitat Wu X, Yan Q, Zhang Z, Du G, Wan X. Acrp30 inhibits leptin-induced metastasis by downregulating the JAK/STAT3 pathway via AMPK activation in aggressive SPEC-2 endometrial cancer cells. Oncol Rep. 2012;27(5):1488–96.PubMed Wu X, Yan Q, Zhang Z, Du G, Wan X. Acrp30 inhibits leptin-induced metastasis by downregulating the JAK/STAT3 pathway via AMPK activation in aggressive SPEC-2 endometrial cancer cells. Oncol Rep. 2012;27(5):1488–96.PubMed
30.
Zurück zum Zitat Kuroda S, Fujiwara T, Shirakawa Y, Yamasaki Y, Yano S, Uno F, et al. Telomerase-dependent oncolytic adenovirus sensitizes human cancer cells to ionizing radiation via inhibition of DNA repair machinery. Cancer Res. 2010;70(22):9339–48.CrossRef Kuroda S, Fujiwara T, Shirakawa Y, Yamasaki Y, Yano S, Uno F, et al. Telomerase-dependent oncolytic adenovirus sensitizes human cancer cells to ionizing radiation via inhibition of DNA repair machinery. Cancer Res. 2010;70(22):9339–48.CrossRef
31.
Zurück zum Zitat Deng R, Tang J, Ma JG, Chen SP, Xia LP, Zhou WJ, et al. PKB/Akt promotes DSB repair in cancer cells through upregulating Mre11 expression following ionizing radiation. Oncogene. 2011;30(8):944–55.CrossRef Deng R, Tang J, Ma JG, Chen SP, Xia LP, Zhou WJ, et al. PKB/Akt promotes DSB repair in cancer cells through upregulating Mre11 expression following ionizing radiation. Oncogene. 2011;30(8):944–55.CrossRef
32.
Zurück zum Zitat Zhang J, Xin X, Chen Q, Xie Z, Gui M, Chen Y, et al. Oligomannurarate sulfate sensitizes cancer cells to doxorubicin by inhibiting atypical activation of NF-kappaB via targeting of Mre11. Int J Cancer. 2012;130(2):467–77.CrossRef Zhang J, Xin X, Chen Q, Xie Z, Gui M, Chen Y, et al. Oligomannurarate sulfate sensitizes cancer cells to doxorubicin by inhibiting atypical activation of NF-kappaB via targeting of Mre11. Int J Cancer. 2012;130(2):467–77.CrossRef
Metadaten
Titel
High expression of MRE11 correlates with poor prognosis in gastric carcinoma
verfasst von
Junqing Li
Taiqiang Su
Liang Yang
Changhua Zhang
Yulong He
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Diagnostic Pathology / Ausgabe 1/2019
Elektronische ISSN: 1746-1596
DOI
https://doi.org/10.1186/s13000-019-0844-y

Weitere Artikel der Ausgabe 1/2019

Diagnostic Pathology 1/2019 Zur Ausgabe

Neu im Fachgebiet Pathologie