NDRG1 was downregulated and worked as favorable biomarker in the development of gastric cancer
Original Article

NDRG1 was downregulated and worked as favorable biomarker in the development of gastric cancer

Xing-Jun Xiao, Hua-Chuan Zheng

Tumor Basic and Translational Laboratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China

Contributions: (I) Conception and design: XJ Xiao; (II) Administrative support: HC Zheng; (III) Provision of study materials or patients: HC Zheng; (IV) Collection and assembly of data: XJ Xiao; (V) Data analysis and interpretation: XJ Xiao; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Hua-Chuan Zheng, PhD. Tumor Basic and Translational Laboratory, The First Affiliated Hospital of Jinzhou Medical University, 5-2 Renmin Street, Jinzhou 121001, China. Email: zheng_huachuan@hotmail.com.

Background: This study clarified the relationship between N-myc downstream regulated gene 1 (NDRG1) expression and the clinicopathological features, DNA methylation, prognosis and relevant signal pathways in gastric cancer (GC).

Methods: NDRG1 expression was examined by Western blot, immunohistochemistry and qRT-PCR. The clinical, transcriptome and methylation data of GC was downloaded from The Cancer Genome Atlas (TCGA), and extracted by R software. The overall survival (OS) rate of NDRG1 was analyzed by Kaplan-Meier plotter. The NDRG1-related gene set enrichment analysis (GSEA) was performed by GSEA-3.0.

Results: NDRG1 expression was down-regulated at both mRNA and protein levels, and immunohistochemically correlated with tumor diameter, depth of invasion, lymph node metastasis and lymphatic invasion, tissue differentiation at a negative manner. The mRNA expression of NDRG1 was negatively related to its methylation. Kaplan-Meier plotter results indicated that NDRG1 was positively correlated with the prognosis of GC patients. NDRG1 was involved in cancer, Notch, PPAR, ERBB, adherens junction, and tight junction signal pathways.

Conclusions: In GC, NDRG1 expression was down-regulated, possibly due to DNA methylation. NDRG1 could play a role of tumor suppressor in the tumorigenesis by inhibiting multiple oncogenic signal pathways. The hypo-expression of NDRG1 was positively associated with malignant biological behavior and adverse prognosis in gastric cancer.

Keywords: N-myc downstream regulated gene 1 (NDRG1); gastric cancer; methylation; bioinformatics analysis


Submitted Aug 19, 2019. Accepted for publication Nov 26, 2019.

doi: 10.21037/tcr.2019.12.76


Introduction

Gastric cancer is one of most common malignant cancers and the third reason of cancer-related deaths in the past two decades worldwide (1). Gastric cancer (GC) ranks the second among cancer deaths, and the incidence of GC is still increasing in China (2). Recent studies reported that GC is affected by Helicobacter pylori infection, high salty-diets, smoking and so on (3-5).

N-myc downstream regulated gene 1 (NDRG1), namely CAP43, DRG-1, RIT42, is a firstly-discovered member of NDRG family. Located at 8q24.22, it spans 60 kb (6). NDRG1 protein was about 43 kD (6,7) and located in cytoplasm, cellular membrane and nucleus (8,9). Reportedly, NDRG1 could inhibit proliferation (10), autophagy (11) and promote apoptosis (12,13), differentiation (14). Other researchers found that NDRG1 could suppress migration and invasion (15), thus inhibit epithelial-mesenchymal transition (EMT) (10,15,16). These evidences indicated that NDRG1 suppressed the aggressive phenotypes as a tumor suppressor.

A body of evidences reported that NDRG1 was decreased in glioma (6), prostate cancer (17), colorectal cancer (12,13,15,18), and ovarian cancer (19). But, NDRG1 expression is still controversial in GC (20,21). Ureshino et al. (20) reported that the NDRG1 over-expression was closely correlated with unfavorable prognosis of GC. However, Chang et al. (21) proved that NDRG1 was correlated inversely with clinicopathological characteristics of GC. Therefore, our aim was to illustrate the relationship between NDRG1 expression and GC by combining clinical data with bioinformatics data. Furthermore, combining with previous work by other researchers, we conducted GSEA to analyze NDRG1-related signal pathways, so as to reveal NDRG1 function in gastric cancer.


Methods

Subjects

All tissues were provided by our hospital between 2012 to 2015. Thirty-four pairs of gastric cancer and adjacent samples were stored in −80 °C refrigerator, and 160 gastric cancer and 86 adjacent samples were prepared in pathological blocks. Patients received no radiotherapy, chemotherapy, or adjuvant treatment before surgery. We were allowed to use their tissues for clinical research, and the study was approved by regional ethics committee of The First Affiliated Hospital of Jinzhou Medical University Medical Ethics Committee.

Quantitative reverse transcription- polymerase chain reaction (qRT-PCR)

Total RNA of 18 pairs of samples were extracted by RNeasy mini kit (QIAGEN, Germany), and then cDNA was synthesized by reverse transcriptase (M-MLV, Takara, Japan) and random primers (Takara, Japan). GAPDH primers: forward 5'-CAATGACCCCTTCATTGACC-3', reverse 5'-TGGAAGATGGTGATGGGATT-3'. NDRG1 primers: forward 5'-GGGCTGAAAAGCATTATTGG-3', reverse 5'-CTCCACCATCTCAGGGTTGT-3'. The iTaqTM universal SYBR® green supermix (BIO-RAD, USA) was used to amplify gene by CFX96TM real-time system (BIO-RAD, Bio-Rad Laboratories, Inc., Singapore). The three-step PCR was selected to amplify gene, and the thermocycling conditions were listed as follows: 1 cycle of 95 °C for 5 min, followed by 60 cycles of 95 °C, 55 °C and 72 °C for 30 s respectively. The gene expression level was calculated as 2−ΔΔCt, where ΔCt=Ct (NDRG1) − Ct (GAPDH), and ΔΔCt=ΔCt (Cancer) − ΔCt (Normal), with GAPDH as an internal control.

Western blot

Proteins were extracted from 16 pairs of samples by RIPA lysis buffer, segregated by 12% SDS-PAGE, and then transferred to PVDF membranes (Millipore, USA, IPVH00010). Skim milk powder was dissolved in TBST, and membranes were blocked in 5% skim milk for 2 h, and incubated with rabbit anti-NDRG1 (#9485, CST) or rabbit anti-GAPDH (AB-P-R 001, Hangzhou Goodhere Biotechnology Co.,). The PVDF membranes were washed for 3 times, and then incubated with anti-rabbit antibody with horseradish peroxidase (HRP, #7074S, CST). Protein Bands were captured with C300 (Azure Biosystems, Azure biosystem Inc, USA) by Western BrightTM ECL western blotting detection kit (Advansta, USA). The gray values of protein bands were measured by Image J software (v1.8.0), with GAPDH as an internal control.

Tissue microarray (TMA) and Immunohistochemistry (IHC)

Tumor or adjacent normal tissues were identified by microscope, and tissue microarrayer (KIN-2, Azumaya Co., Ltd., Japan) was used to transfer corresponding tissue cores to a recipient block. Consecutive sections were incised and then transferred to glass slides coated with poly-lysine. The slides were deparaffinized and rehydrated, and then conducted antigen retrieval. The slides were blocked with 3% hydrogen peroxide, and then 5% bovine serum albumin was used to block non-specific binding sites. The slides were incubated with rabbit anti-NDRG1 (#9485, CST) for overnight at 4 °C. After rinsed with PBS for 3 times, the slides were incubated with polyclonal swine anti-rabbit antibody with HRP (P0399, DAKO) in room temperature for 2 h. DAB (BOSTER, USA) was used to visualize the specific binding sites. After stained with hematoxylin (Solarbio, China), the slides were dehydrated, cleared, mounted and visualize by a microscope (Nikon, Nikon Corporation, Japan).

Evaluation of IHC

The proportion of positive cells (0 = negative; 1 = 1–50%; 2 = 50–74%; 3 ≥75%) was used to evaluate the positive rate of NDRG1 expression, and the staining intensity (0=negative; 1 = weak; 2 = intermediate; and 3 = strong) was used to estimate NDRG1 expression level. Therefore, a final score was obtained by multiplying the score of NDRG1 positive rate and staining intensity, which determined NDRG1 expression as (− = 0; + = 1–3; ++ = 4–6; +++ = 7–9). The final score of 0–3 was considered as low NDRG1 expression and that of 3–9 was considered as high NDRG1 expression.

Bioinformatics analysis

Transcriptome, clinical and methylation data of GC patients was downloaded from TCGA database (https://portal.gdc.cancer.gov/). Thirty adjacent normal and 344 GC samples were obtained in transcriptome data. The normalized transcriptome data of GC patients was also used to conduct GSEA. The raw NDRG1 clinical data was obtained by integrating clinical with transcriptome data, and used to compare NDRG1 expression with clinicopathological parameters. The transcriptome and methylation data were used to perform NDRG1 methylation analysis. The overall survival rate of NDRG1 was downloaded from Kaplan-Meier plotter (http://www.kmplot.com/).

Methylation analysis

The methylation and transcriptome data were employed to analyze the relationship between NDRG1 methylation and mRNA expression, survival rate. DNA methylation was measured by Illumina Human Methylation 450, and methylated and unmethylated signals were used to calculate beta (β) values, which were quantitative scores of the DNA methylation levels. The correlation between NDRG1 methylation and mRNA expression was analyzed by Pearson’s correlation analysis. The median values of β and mRNA expression were used as cut-off values, and then Kaplan-Meier survival curves of NDRG1 methylation were obtained.

Gene set enrichment analysis (GSEA)

GSEA-3.0 was downloaded from http://software.broadinstitute.org/, and GSEA was conducted by gsea.3.0. The mRNA expression data of NDRG1 was divided into two groups (H vs. L) according to the median value of GC patients. NDRG1 mRNA expression level was used as a phenotype label and the normalized expression matrix of GC patients was used as expression dataset. C2.cp.kegg.v6.2.symbols.gmt was used as gene set database to analyze NDRG1-related KEGG signal pathways.

Statistical analysis

R (v.3.5.3) was downloaded from https://www.r-project.org/ and used to extract and analyze raw data of TCGA-GC. Pearson’s correlation test, log-rank test and students’ t test were used to analyze the relationship between NDRG1 expression and methylation, NDRG1 methylation and survival and compare the means respectively. Chi-square test was used to evaluate NDRG1 expression level in gastric cancer and para-cancer tissues and the relationship between NDRG1 expression and clinicopathological parameters. All the data was handled by SPSS 12.0 software and graphpad prism 6. P<0.05 was regarded as statistically significant.


Results

NDRG1 expression in gastric cancer

TCGA database and qRT-PCR indicated that the mRNA expression was decreased in GC (P<0.001, Figure 1A,B,C). It was the same according to western blot results (P<0.001, Figure 1D,E,F). NDRG1 protein was positively detected in adjacent normal tissue (Figure 2A), intestinal-type (Figure 2B) and diffuse-type gastric cancer (Figure 2C). Immunohistochemically, the high expression rate of NDRG1 was lower in GC than that in adjacent normal tissues (25.6% vs. 69.8%, P=0.001, Table 1).

Figure 1 NDRG1 was downregulated at mRNA and protein levels in gastric cancer. NDRG1 mRNA expression in TCGA database (A) and in GC (Tumor) and adjacent normal tissues (B) respectively. The average NDRG1 mRNA in GC and adjacent normal tissues (C). The NDRG1 protein level in GC and adjacent normal tissues (D-F). GAPDH was used as internal control and Image J was used to evaluate the gray value of protein bands. Data were presented as mean ± standard deviation. ***P<0.001; Normal (N): adjacent normal tissues; Tumor (T): gastric cancer tissues.
Figure 2 Typical pictures of IHC. NDRG1 expression was observed in normal tissue (A), intestinal gastric cancer (B), diffuse gastric cancer (C). All the pictures were captured by 200× microscope.

Table 1

NDRG1 expression in gastric cancer

Group n NDRG1 expression HER (%) P value
Low (−, +) High (++, +++)
Cancer 160 119 41 25.6 0.001*
Normal 86 26 60 69.8

HER, high expression rate; Normal, adjacent normal tissues. *P<0.05.

The relationship between NDRG1 protein expression and clinicopathological characteristics of gastric cancer

The IHC results showed that NDRG1 expression was negatively associated with tumor diameter (P=0.002), depth of invasion (P=0.002), lymph node metastasis (P=0.012), lymphatic invasion (P=0.035) and differentiation (P=0.013), and not correlated with age, sex or distant metastasis (Table 2). And NDRG1 expression was lower in diffuse-type than intestinal-type carcinomas (P=0.034).

Table 2

The relationship between NDRG1 protein expression and pathological parameters in gastric cancer

Clinicopathological features n NDRG1 expression, n (%) P value
Low (−, +) High (++, +++)
Age (years) 0.276
   ≤60 73 51 (69.9) 22 (30.1)
   >60 87 68 (78.2) 19 (21.8)
Gender 1.000
   Male 116 86 (74.1) 30 (25.9)
   Female 44 33 (75.0) 11 (25.0)
Tumor diameter (cm) 0.002*
   ≤5 116 81 (69.8) 35 (30.2)
   >5 41 36 (87.8) 5 (12.2)
Depth of invasion 0.002*
   Tis–2 37 20 (54.1) 17 (45.9)
   T3–4 123 99 (80.5) 24 (19.5)
Lymph node metastasis 0.012*
   Yes 108 87 (80.6) 21 (19.4)
   No 52 32 (61.5) 20 (38.5)
Distant metastasis 0.115
   Yes 8 8 (100.0) 0 (0.0)
   No 152 111 (73.0) 41 (27.0)
Lymphatic invasion 0.035*
   Yes 73 60 (82.2) 13 (17.8)
   No 87 59 (67.8) 28 (32.2)
Lauren’s classification 0.034*
   Intestinal-type 42 25 (59.5) 17 (40.5)
   Diffuse-type 79 62 (78.5) 17 (21.5)
   Mixed-type 39 32 (82.1) 7 (17.9)
Differentiation 0.013*
   Poorly 114 91 (79.8) 23 (20.2)
   Well & moderately 40 24 (60.0) 16 (40.0)

Tis, cancer in situ; T1, lamina propria and submucosa; T2, muscularis propria and subserosa; T3, exposure to serosa; T4, invasion into serosa. *P<0.05.

The correlation between NDRG1 mRNA expression and clinicopathological features of gastric cancer

The transcriptome and clinical data were extracted as mentioned above, and then raw NDRG1 clinical data was obtained. Regardless of survival days, NDRG1 expression was lower in T3–4 than that in T1–2 group (P<0.05, Figure 3A). In the old (age >60 years), male, G3, stage I–II, N0–1 and M0 groups, NDRG1 expression was also lower in T3–4 than that in T1–2 group (P<0.05, Figure 3B,C,D,E,F,G). In the old group (age >60 years), we observed that NDRG1 expression was weaker in stage III–IV than that in stage I–II group (P<0.05, Figure 3H).

Figure 3 The correlation between NDRG1 mRNA expression and clinicopathological features of gastric cancer based on TCGA. NDRG1 expression was negatively correlated with T staging in the survival time, old, male, grade 3, stage I–II, N0–1 and M0 groups respectively (A-G). And NDRG1 was also negatively associated with stage in the old group (H). Data were presented as mean ± standard deviation. *P<0.05; ***P<0.001; Note: 388 was the median survival time of the total gastric cancer patients.

The methylation analysis of NDRG1 in gastric cancer

NDRG1 mRNA expression was negatively associated with DNA methylation (P=8.282e−08, Figure 4A). However, there was no relationship between NDRG1 methylation or mRNA expression and survival rate (P=0.806 and P=0.663, Figure 4B,C). Twenty-six methylation sites of NDRG1 were extracted, but only seven significant methylation sites were negatively correlated with NDRG1 expression (Table 3). Pearson’s correlation coefficient (PCC ≥−0.30) was considered as significant.

Figure 4 The analysis of NDRG1 promoter methylation in gastric cancer. The relationship between NDRG1 methylation and its mRNA expression level (A). The Kaplan-Meier survival curves for NDRG1 methylation (B) and the integrative analysis of NDRG1 (C). NDRG1 hypo & high expression: NDRG1 hypo-methylation and high mRNA expression; NDRG1 hyper & low expression: NDRG1 hyper- methylation and low mRNA expression.

Table 3

NDRG1 methylation sites were negatively correlated with mRNA expression level in gastric cancer

Number Methylation site Start End PCC P value
1 cg00799984 133294862 133294863      −0.31 2.945e−08
2 cg09102409 133296491 133296492      −0.30 8.663e−08
3 cg16001384 133294558 133294559      −0.416 2.648e−14
4 cg17129188 133295354 133295355      −0.44 5.698e−16
5 cg20100745 133295485 133295486      −0.426 5.868e−15
6 cg23417096 133237773 133237774      −0.332 2.614e−09
7 cg25232510 133293980 133293981      −0.372 1.577e−11

PCC, Pearson correlation coefficient.

The relationship between NDRG1 expression and prognosis in gastric cancer

According to Kaplan-Meier plotter, in most cases, the mRNA down-regulation of NDRG1 was positively associated with low overall survival rate in GC (Table 4), especially with higher invasion (T), lymph node metastasis (N), higher pathological stage (Stage), diffuse-type manner and HER2 positivity. Taken together, the low NDRG1 expression may be related to poor prognosis of GC patients.

Table 4

The relationship between NDRG1 expression and overall survival rate of gastric cancer patients

Number Overall survival rate (OS) Total HR (95%CI) P value
1 Diffuse type & no perforation 69 0.44 (0.24–0.82)        0.0077
2 Diffuse type & poor differentiation 74 0.53 (0.29–0.97)        0.035
3 Female & no perforation 62 0.44 (0.2–0.95)        0.033
4 Intestinal type & HER positive 147 0.58 (0.37–0.89)        0.011
5 M0 & no perforation 140 0.6 (0.38–0.96)        0.032
6 N0 & no perforation 36 0.18 (0.05–0.57)        0.0013
7 N1 & other adjuvant 34 0.16 (0.03–0.85)        0.014
8 N2 & HER positive 39 0.37 (0.15–0.91)        0.025
9 N2 & intestinal type 53 0.44 (0.22–0.88)        0.016
10 N2 & M0 104 0.57 (0.34–0.95)        0.029
11 N3 & female 28 0.4 (0.18–0.98)        0.04
12 N3 & HER negative 57 0.52 (0.27–0.99)        0.042
13 N3 & surgery alone 52 0.49 (0.24–1)        0.047
14 No perforation & HER positive 79 0.42 (0.21–0.81)        0.0079
15 Stage2 & difuse type 53 0.3 (0.11–0.81)        0.012
16 Stage2 & intestinal type 134 0.65 (0.43–0.99)        0.041
17 Stage2 & other adjuvant 25 0.12 (0.01–1.33)        0.038
18 Stage3 & female 87 0.54 (0.31–0.95)        0.029
19 Stage3 & HER positive 137 0.56 (0.37–0.85)        0.0056
20 Stage 3 & N2 96 0.55 (0.32–0.94)        0.028
21 Stage 4 & N1–3 124 0.64 (0.41–1)        0.048
22 Stage 4 & N3 76 0.56 (0.32–1)        0.047
23 Stage 4 & T3 66 0.51 (0.26–0.98)        0.039
24 Stage 4 & T4 36 0.34 (0.13–0.99)        0.022
25 T3 & diffuse type 113 0.56 (0.35–0.91)        0.016
26 T3 & HER positve 65 0.41 (0.22–0.75)        0.0032
27 T3 & M0 174 0.66 (0.45–0.97)        0.032
28 T3 & N1–3 186 0.67 (0.47–0.97)        0.033
29 T3 & N2 59 0.42 (0.22–0.82)        0.0086
30 T3 & N3 42 0.38 (0.18–0.83)        0.012
31 T3 & no perforation 98 0.59 (0.35–0.98)        0.04
32 T4 & intestinal type 20 0.2 (0.04–0.93)        0.024
33 Well differentiation & HER positive 27 0.25 (0.09–0.71)        0.0048

HR, hazard ratio; CI, confidence interval; T, tumor; N, lymph node metastasis; M, distant metastasis; Stage, pathological stage.

NDRG1-related signal pathways in gastric cancer

As mentioned above, we conducted a GSEA to analyze NDRG1-related signal pathways in GC. According to nominal P value, significant enriched pathways were related to biological process of GC. The results showed that TCGA-GC patients in high NDRG1 expression group were enriched in many signal pathways, including cancer, Notch, PPAR, ERBB, adherens junction and tight junction signal pathways (P<0.05, Figure 5). NDRG1 was also enriched in other cancer signal pathways, such as bladder, thyroid, prostate, renal cell, pancreatic cancer and acute myeloid leukemia (P<0.05, Figure S1).

Figure 5 Enrichment plots from gene set enrichment analysis (GSEA). GSEA results showed that NDRG1 mRNA expression was positively correlated with the cancer (A), Notch (B), PPAR (C), ERBB (D), adherens junction (E) and tight junction (F) signal pathways respectively. ES, enrichment score; NES, normalized ES; FDR, false discovery rate.

Discussion

Based on pathological and bioinformatics results, NDRG1 expression was decreased in GC, which was consistent with many other cancers (6,13-14,17-19). Chang et al. (22) reported that NDRG1 was down-regulated significantly in gastric cancer. The results of IHC and TCGA were basically consistent with our previous article (23). In addition, NDRG1 expression was decreased not only in prostate cancer (17), pancreatic cancer (24) and but also acute myeloid leukemia (25), renal cell carcinoma (26). Compared with these tumors, NDRG1 may have similar tumorigenesis mechanism in GC. However, the NDRG1 expression was up-regulated in bladder cancer (27) and thyroid cancer (28), contrary to our GSEA results. This discrepancy might be owing to tissue specificity or other mechanisms might be involved in the tumorigenesis. It was indicated that NDRG1 hypo-expression might participated in the tumorgenesis. As we all known, DNA methylation might inhibit gene expression. NDRG1 methylation has been found in prostate cancer (17), pancreatic cancer (24), breast cancer (29), but was barely reported in GC. In our study, NDRG1 down-regulation was associated with gene methylation. NDRG1 methylation could silence gene expression and activate oncogenes, and then disorder cell proliferation and apoptosis (17).

Reportedly, NDRG1 was involved in inhibiting multiple oncogenic signal pathways. Therefore, we conducted a GSEA to investigate the function of NDRG1 in GC. In high expression phenotype group, NDRG1 was enriched in cancer signal pathway. Notch signal pathway is a conserved signaling system, the activation of which may induce the expression of downstream target genes, including NF-κB, Cyclin D1, p21, GATA3, c-Myc, Deltex1 (30). Notch signal pathway could induce cell proliferation, metastasis, drug resistance, and inhibit apoptosis (31). In GC, the activation of Notch signal pathway could promote GC progression (32). However, it was reported that NDRG1 could inhibit NF-κB signal pathway (33-35). Cai et al. (36) reported that NDRG1 could induce PPARγ expression. PPARγ could inhibit the proliferation, migration and Wnt/β-catenin signal pathway and promoted apoptosis of GC cell lines (37,38). Jin et al. (39) reported that NDRG1 could suppress nuclear translocation of β-catenin via WNT/β-catenin signal pathway. The activation of ERBB signal pathway could continue activating downstream oncogenic signal pathways, such as Ras-Raf-MAPK signal pathway and PI3K-AKT signal pathway, which were involved in suppressing apoptosis, promoting proliferation, angiogenesis, migration, invasion, and metastasis (40). NDRG1 could suppress ERBB signal pathway by inhibiting the expression and activation of key factors, such as EGFR (ERBB), HER2 (ERBB2) and HER3, and the formation of the heterodimers (41). Over-expression or amplification of EGFR and HER2 were associated with a poor prognosis in GC (42). NDRG1 could suppress Ras-Raf-MAPK, PI3K-AKT signal pathways (43-45). Reportedly, NDRG1 was closely associated with adherens junction, and participated in forming E-cadherin/catenin complex (46). Gon et al. (47) proved that NDRG1 down-regulation disrupted tight junctions. The disruption of adherens junction and tight junction was one of critical processes of EMT. Increasing evidences (10,15,16) proved that NDRG1 inhibited EMT. Taken together, NDRG1 could play a role of tumor suppressor via the six signal pathways in the tumorigenesis of GC. NDRG1 down-regulation was negatively correlated with some clinicopathological characteristics and positively associated with poor prognosis in GC, which was validated by pathological and Kaplan-Meier plotter results.

Several limitations should be considered in this paper. Firstly, the tissues of gastric cancer patients were too few. For example, the patients with distant metastasis only has eight samples. Secondly, without corresponding experimental data, we analyzed the signal pathways by combining limited existing data with the published works in the PubMed, which was the biggest drawback in this work.


Conclusions

NDRG1 expression was down-regulated in gastric cancer, which was negatively associated with gene methylation. NDRG1 could play a role of tumor suppressor in the tumorgenesis by inhibiting multiple oncogenic signal pathways. NDRG1 down-regulation was positively associated with aggressive behaviors and poor prognosis, which could be used as a new biomarker for the assessment and treatment of gastric cancer.

Figure S1 NDRG1 was enriched in other cancer-related signal pathways, including bladder, thyroid, prostate, renal cell, pancreatic cancer and acute myeloid leukemia signal pathways (S1A-F) respectively.

Acknowledgments

Funding: This study was financed by National Natural Scientific Foundation of China (81672700), Natural Scientific Foundation of Liaoning Province (20180530105) and Shenyang Science and Technology Grand (18-013-0-59).


Footnote

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/tcr.2019.12.76). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The human tissue collection was approved by the Ethics Committee of Jinzhou Medical University (Jinzhou, Liaoning province). All patients consented to participate in this research.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA Cancer J Clin 2017;67:7-30. [Crossref] [PubMed]
  2. Zheng R, Zeng H, Zhang S, et al. Estimates of cancer incidence and mortality in China, 2013. Chin J Cancer 2017;36:66. [Crossref] [PubMed]
  3. Wu Y, Shen L, Liang X, et al. Helicobacter pylori-induced YAP1 nuclear translocation promotes gastric carcinogenesis by enhancing IL-1β expression. Cancer Med 2019;8:3965-80. [PubMed]
  4. Kim SA, Choi BY, Song KS, et al. Prediagnostic Smoking and Alcohol Drinking and Gastric Cancer Survival: A Korean Prospective Cohort Study. Korean J Gastroenterol 2019;73:141-51. [Crossref] [PubMed]
  5. Hu J, La Vecchia C, Morrison H, et al. Salt, processed meat and the risk of cancer. Eur J Cancer Prev 2011;20:132-9. [Crossref] [PubMed]
  6. Sun B, Chu D, Li W, et al. Decreased expression of NDRG1 in glioma is related to tumor progression and survival of patients. J Neurooncol 2009;94:213-9. [Crossref] [PubMed]
  7. Kovacevic Z, Fu D, Richardson DR. The iron-regulated metastasis suppressor, Ndrg-1: identification of novel molecular targets. Biochim Biophys Acta 2008;1783:1981-92. [Crossref] [PubMed]
  8. Sun J, Zhang D, Bae DH, et al. Metastasis suppressor, NDRG1, mediates its activity through signaling pathways and molecular motors. Carcinogenesis 2013;34:1943-54. [Crossref] [PubMed]
  9. Wangpu X, Lu J, Xi R, et al. Targeting the Metastasis Suppressor, N-Myc Downstream Regulated Gene-1, with Novel Di-2-Pyridylketone Thiosemicarbazones: Suppression of Tumor Cell Migration and Cell-Collagen Adhesion by Inhibiting Focal Adhesion Kinase/Paxillin Signaling. Mol Pharmacol 2016;89:521-40. [Crossref] [PubMed]
  10. Chen Z, Sun J, Li T, et al. Iron chelator-induced up-regulation of Ndrg1 inhibits proliferation and EMT process by targeting Wnt/β-catenin pathway in colon cancer cells. Biochem Biophys Res Commun 2018;506:114-21. [Crossref] [PubMed]
  11. Sahni S, Bae DH, Lane DJ, et al. The metastasis suppressor, N-myc downstream-regulated gene 1 (NDRG1), inhibits stress-induced autophagy in cancer cells. J Biol Chem 2014;289:9692-709. [Crossref] [PubMed]
  12. Zhang X, Feng B, Zhu F, et al. N-myc downstream-regulated gene 1 promotes apoptosis in colorectal cancer via up-regulating death receptor 4. Oncotarget 2017;8:82593-608. [PubMed]
  13. Yang X, Zhu F, Yu C, et al. N-myc downstream-regulated gene 1 promotes oxaliplatin-triggered apoptosis in colorectal cancer cells via enhancing the ubiquitination of Bcl-2. Oncotarget 2017;8:47709-24. [PubMed]
  14. Fotovati A, Abu-Ali S, Kage M, et al. N-myc downstream-regulated gene 1 (NDRG1) a differentiation marker of human breast cancer. Pathol Oncol Res 2011;17:525-33. [Crossref] [PubMed]
  15. Mi L, Zhu F, Yang X, et al. The metastatic suppressor NDRG1 inhibits EMT, migration and invasion through interaction and promotion of caveolin-1 ubiquitylation in human colorectal cancer cells. Oncogene 2017;36:4323-35. [Crossref] [PubMed]
  16. Xi R, Pun IH, Menezes SV, et al. Novel Thiosemicarbazones Inhibit Lysine-Rich Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1 (CEACAM1) Coisolated (LYRIC) and the LYRIC-Induced Epithelial-Mesenchymal Transition via Upregulation of N-Myc Downstream-Regulated Gene 1 (NDRG1). Mol Pharmacol 2017;91:499-517. [Crossref] [PubMed]
  17. Li Y, Pan P, Qiao P, et al. Downregulation of N-myc downstream regulated gene 1 caused by the methylation of CpG islands of NDRG1 promoter promotes proliferation and invasion of prostate cancer cells. Int J Oncol 2015;47:1001-8. [Crossref] [PubMed]
  18. Liu J, Shao Y, He Y, et al. MORC2 promotes development of an aggressive colorectal cancer phenotype through inhibition of NDRG1. Cancer Sci 2019;110:135-46. [Crossref] [PubMed]
  19. Wang B, Li J, Ye Z, et al. N-myc downstream regulated gene 1 acts as a tumor suppressor in ovarian cancer. Oncol Rep 2014;31:2279-85. [Crossref] [PubMed]
  20. Ureshino H, Murakami Y, Watari K, et al. N-myc downstream regulated gene 1 (NDRG1) promotes metastasis of human scirrhous gastric cancer cells through epithelial mesenchymal transition. PLoS One 2012;7:e41312. [Crossref] [PubMed]
  21. Chang X, Xu X, Xue X, et al. NDRG1 Controls Gastric Cancer Migration and Invasion through Regulating MMP-9. Pathol Oncol Res 2016;22:789-96. [Crossref] [PubMed]
  22. Chang X, Xu X, Ma J, et al. NDRG1 expression is related to the progression and prognosis of gastric cancer patients through modulating proliferation, invasion and cell cycle of gastric cancer cells. Mol Biol Rep 2014;41:6215-23. [Crossref] [PubMed]
  23. Xiao XJ, Wu JC, Zheng HC. Retrospective study of the roles of NDRG1 expression in gastric cancer: a meta- and bioinformatics analysis. Int J Clin Exp Med 2018;11:11889-900.
  24. Angst E, Dawson DW, Nguyen A, et al. Epigenetic regulation affects N-myc downstream-regulated gene 1 expression indirectly in pancreatic cancer cells. Pancreas 2010;39:675-9. [Crossref] [PubMed]
  25. Tschan MP, Shan D, Laedrach J, et al. NDRG1/2 expression is inhibited in primary acute myeloid leukemia. Leuk Res 2010;34:393-8. [Crossref] [PubMed]
  26. Hosoya N, Sakumoto M, Nakamura Y, et al. Proteomics identified nuclear N-myc downstream-regulated gene 1 as a prognostic tissue biomarker candidate in renal cell carcinoma. Biochim Biophys Acta 2013;1834:2630-9. [Crossref] [PubMed]
  27. Li A, Zhu X, Wang C, et al. Upregulation of NDRG1 predicts poor outcome and facilitates disease progression by influencing the EMT process in bladder cancer. Sci Rep 2019;9:5166. [Crossref] [PubMed]
  28. Gerhard R, Nonogaki S, Fregnani JH, et al. NDRG1 protein overexpression in malignant thyroid neoplasms. Clinics (Sao Paulo) 2010;65:757-62. [PubMed]
  29. Han LL, Hou L, Zhou MJ, et al. Aberrant NDRG1 methylation associated with its decreased expression and clinicopathological significance in breast cancer. J Biomed Sci 2013;20:52. [Crossref] [PubMed]
  30. Gu Y, Masiero M, Banham AH. Notch signaling: its roles and therapeutic potential in hematological malignancies. Oncotarget 2016;7:29804-23. [Crossref] [PubMed]
  31. Zang MD, Hu L, Fan ZY, et al. Luteolin suppresses gastric cancer progression by reversing epithelial-mesenchymal transition via suppression of the Notch signaling pathway. J Transl Med 2017;15:52. [Crossref] [PubMed]
  32. Menezes SV, Fouani L, Huang MLH, et al. The Metastasis Suppressor, NDRG1, Attenuates Oncogenic TGF-β and NF-ĸB Signaling to Enhance Membrane E-Cadherin Expression in Pancreatic Cancer Cells. Carcinogenesis 2019;40:805-18. [Crossref] [PubMed]
  33. Chen L, Xing C, Ma G, et al. N-myc downstream-regulated gene 1 facilitates influenza A virus replication by suppressing canonical NF-κB signaling. Virus Res 2018;252:22-8. [Crossref] [PubMed]
  34. Fouani L, Kovacevic Z, Richardson DR. Targeting Oncogenic Nuclear Factor Kappa B Signaling with Redox-Active Agents for Cancer Treatment. Antioxid Redox Signal 2019;30:1096-23. [Crossref] [PubMed]
  35. Jiang L, Lin T, Xu C, et al. miR-124 interacts with the Notch1 signalling pathway and has therapeutic potential against gastric cancer. J Cell Mol Med 2016;20:313-22. [Crossref] [PubMed]
  36. Cai K, El-Merahbi R, Loeffler M, et al. Ndrg1 promotes adipocyte differentiation and sustains their function. Sci Rep 2017;7:7191. [Crossref] [PubMed]
  37. Guo F, Ren X, Dong Y, et al. Constitutive expression of PPARγ inhibits proliferation and migration of gastric cancer cells and down-regulates Wnt/β-Catenin signaling pathway downstream target genes TERT and ENAH. Gene 2016;584:31-7. [Crossref] [PubMed]
  38. Liu L, Si N, Ma Y, et al. Hydroxysafflor-Yellow A Induces Human Gastric Carcinoma BGC-823 Cell Apoptosis by Activating Peroxisome Proliferator- Activated Receptor Gamma (PPARγ). Med Sci Monit 2018;24:803-11. [Crossref] [PubMed]
  39. Jin R, Liu W, Menezes S, et al. The metastasis suppressor NDRG1 modulates the phosphorylation and nuclear translocation of β-catenin through mechanisms involving FRAT1 and PAK4. J Cell Sci 2014;127:3116-30. [Crossref] [PubMed]
  40. Resende C, Ristimäki A, Machado JC. Genetic and epigenetic alteration in gastric carcinogenesis. Helicobacter 2010;15:34-9. [Crossref] [PubMed]
  41. Kovacevic Z, Menezes SV, Sahni S, et al. The Metastasis Suppressor, N-MYC Downstream-regulated Gene-1 (NDRG1), Down-regulates the ErbB Family of Receptors to Inhibit Downstream Oncogenic Signaling Pathways. J Biol Chem 2016;291:1029-52. [Crossref] [PubMed]
  42. Thiel A, Ristimaki A. Targeted therapy in gastric cancer. APMIS 2015;123:365-72. [Crossref] [PubMed]
  43. Yang Y, Liu Y, Guo R, et al. The novel dithiocarbamate, DpdtC suppresses HER2-overexpressed cancer cells by up-regulating NDRG1 via inactivation of HER2-ERK 1/2 signaling. Sci Rep 2018;8:3398. [Crossref] [PubMed]
  44. Fu Y, Wei J, Dai X, et al. Increased NDRG1 expression attenuate trophoblast invasion through ERK/MMP-9 pathway in preeclampsia. Placenta 2017;51:76-81. [Crossref] [PubMed]
  45. Kovacevic Z, Chikhani S, Lui GY, et al. The iron-regulated metastasis suppressor NDRG1 targets NEDD4L, PTEN, and SMAD4 and inhibits the PI3K and Ras signaling pathways. Antioxid Redox Signal 2013;18:874-87. [Crossref] [PubMed]
  46. Lachat P, Shaw P, Gebhard S, et al. Expression of NDRG1, a differentiation- related gene, in human tissues. Histochem Cell Biol 2002;118:399-408. [Crossref] [PubMed]
  47. Gon Y, Maruoka S, Kishi H, et al. NDRG1 is important to maintain the integrity of airway epithelial barrier through claudin-9 expression. Cell Biol Int 2017;41:716-25. [Crossref] [PubMed]
Cite this article as: Xiao XJ, Zheng HC. NDRG1 was downregulated and worked as favorable biomarker in the development of gastric cancer. Transl Cancer Res 2020;9(1):210-221. doi: 10.21037/tcr.2019.12.76

Download Citation