INTRODUCTION
Colorectal cancer (CRC), also called colon cancer or large bowel cancer, is the most common type of gastrointestinal cancer and one of the major contributors to cancer‐related death worldwide. Metastasis is considered to be the leading cause of mortality in CRC patients. Recently, genetic and epigenetic alterations in CRC have been extensively studied. However, the underlying molecular mechanisms and primary biomarkers for metastasis are still not well determined, which is urgently required for the development of effective therapeutic interventions and methods of managing this disease.
MORC2, also known as ZCWCC1, ZCW3, or KIAA0852, is a member of the MORC family of proteins. Our previous work showed that, in gastric cancer cells, MORC2 suppressed carbonic anhydrase IX (CAIX), p21Waf1/Cip1, and ArgBP2 (Arg‐binding protein 2) gene expression through histone deacetylase 4 (HDAC4), HDAC1, and EZH2, respectively. It has been reported that MORC2 facilitated chromatin remodeling following the DNA damage response and promoted lipogenesis. We also showed that phosphorylation of MORC2 on serine 677 by PAK1 promoted gastric tumorigenesis. It is reported that MORC2 promoted breast cancer invasion and metastasis through a PRD domain‐mediated interaction with catenin delta 1. Recently, it has been shown that MORC2‐mutant M276I promotes metastasis of triple‐negative breast cancer by regulating CD44 splicing. Moreover, MORC2 promotes cancer stemness and tumorigenesis by facilitating DNA methylation‐dependent silencing of Hippo signaling in hepatocellular carcinoma. Additionally, MORC2 was found to be one of the mutation hotspot oncogenes in CRCs with microsatellite instability. However, the potential oncogenic roles and molecular mechanisms of MORC2 in CRC remain elusive.
N‐myc downstream regulated gene 1 (NDRG1) is a well‐characterized metastasis suppressor that has shown the potential to be developed as a target for antimetastatic therapy. NDRG1 mediates its activity through various signaling pathways and molecular motors. It has been reported that NDRG1 was downregulated in CRC tissues and it was a prognostic biomarker for human colorectal cancer. Moreover, NDRG1 inhibited epithelial‐mesenchymal transition, migration, and invasion of CRC cells through interaction and promotion of caveolin‐1 ubiquitylation.
In this study, we found that MORC2 bound to NDRG1 promoter and inhibited NDRG1 expression in CRC cells. We also show that MORC2 interacted with sirtuin 1 (SIRT1) and inhibited NDRG1 promoter activity independently and cumulatively with SIRT1. We reveal that NDRG1 was required in MORC2‐mediated promotion of CRC cell migration and invasion in vitro, as well as lung metastasis of CRC cells in vivo. Furthermore, we show the negative correlation between MORC2 and NDRG1 in CRC samples. We found that high expression of MORC2 was significantly associated with lymph node metastasis and poor pTNM stage. Decreased expression of NDRG1 was significantly related to lymph node metastasis in CRC samples. Our results might thus contribute to understanding the mechanisms of NDRG1 transcriptional regulation and suggest MORC2 as a potential therapeutic target for CRC.
MATERIALS AND METHODS
Cell culture
HT‐29, SW‐480, and SW‐620 cells were cultured in RPMI‐1640 medium, and HEK‐293 cells were cultured in DMEM, supplemented with 10% FBS, 100 μg/mL streptomycin, 100 U/mL penicillin, and 1% glutamine at 37°C in 5% CO2 and 95% air.
Plasmids, transient transfection, and luciferase assay
For the construction of NDRG1 promoter‐driven luciferase reporter plasmid, a series of fragments were amplified by PCR from human genomic DNA. These PCR products were digested with BglII/HindIII and inserted into the firefly luciferase reporter vector pGL3‐basic (Promega, Madison, WI, USA). The sense primers were 5′‐GGAAGATCTACGGTGCTAAGGTTGGAAAGGG‐3′ (−759 to +69 bp), 5′‐GGAAGATCTCCGAGCTGGTGAGACCTACA‐3′(−446 to +69 bp), and 5′‐GGAAGATCTACTGCAGAGCCGACCCACAA‐3′(−213 to +69 bp). The antisense primer was 5′‐CCCAAGCTTGGAGCCAGGCGAGGTTTGTTTA‐3′. We constructed 3× Flag‐CMV‐MORC2 in our laboratory. Transient transfection was carried out using Lipofectamine 3000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's instructions. Luciferase assay has been described previously in detail. Cells were transfected with indicated reporter and Renilla‐encoding plasmids. All measurements were repeated at least 3 times and luciferase values were normalized to internal Renilla control.
Lentiviral vector production and generation of stable cell lines
Flag‐vector lentivirus, Flag‐MORC2 lentivirus, nonsilencing (NC)‐shRNA lentivirus, MORC2‐shRNA lentivirus, SIRT1‐shRNA lentivirus, and NDRG1‐shRNA lentivirus were purchased from GeneChem (Shanghai, China). HT‐29, SW‐620, and SW‐480 cells were transfected with various plasmids using lentivirus according to the manufacturer's instructions. Stable clonal cell lines were selected with 2 μg/mL puromycin.
Immunoprecipitation and western blot analyses
Immunoprecipitation and western blot analyses have been described previously in detail. The samples were incubated with anti‐MORC2 (Bethyl Laboratories, Montgomery, TX, USA), anti‐NDRG1 (Cell Signaling Technology, Danvers, MA, USA) and anti‐SIRT1 (Cell Signaling Technology) antibodies.
RNA isolation, reverse transcription, and real‐time PCR
RNA was extracted with TRIzol (Invitrogen). cDNA was synthesized by reverse transcription using an RT reaction kit (Takara, Dalian, China), according to the manufacturer's instructions. Real‐time PCR was carried out according to the protocol used in our previous study. The primers for NDRG1 were: 5′‐TGGACCCAACAAAGACCACT‐3′ (sense) and 5′‐CCATCCAGAGAAGTGACGCT‐3′ (antisense); and for β‐actin were: 5′‐TCGTGCGTGACATTAAGGAG‐3′ (sense) and 5′‐ATGCCAGGGTACATGGTGGT‐3′ (antisense). Gene expression levels were calculated relative to the housekeeping gene β‐actin by using Stratagene Mx 3000P software (Agilent Technologies Inc., CA, USA).
Tissue samples and immunohistochemical staining
Nontumor colon tissues (5 cm away from the cancer edge) from 36 patients and human CRC tissues from 119 patients undergoing radical colon resection were obtained at the First Hospital of China Medical University (Shenyang, China). Fresh samples were snap frozen in liquid nitrogen immediately after resection and stored at −80°C. All samples were obtained with patients’ informed consent. The samples were histologically confirmed by staining with H&E. The histological grade of cancer was assessed according to criteria set by the WHO. The TNM classification was undertaken according to the standard criteria of the 7th TNM staging system. Immunohistochemistry has been described previously, and immunohistochemical results were judged by HSCORE (histological score).
Transwell migration and invasion assays and wound healing assays
Transwell migration and invasion assays have been described previously. The number of migrated cells was counted in 5 representative microscopic fields that were photographed. Three independent experiments were carried out. The invasion assay was undertaken using BD BioCoat Matrigel invasion chambers (8 mm pore size; BD Biosciences, San Jose, CA, USA). The same procedures described above were used, except that the filters were precoated with 100 mL Matrigel at a 1:4 dilution in 1640 medium to form a genuine reconstituted basement membrane. For wound healing assays, cells were seeded and grown to confluency. Then the cells were gently scratched with tips to create a mechanical wound. Images were taken at 0, 24, and 48 hours using a microscope.
Nude mice xenografts
Five‐ to 6‐week‐old female athymic nude BALB/c mice were purchased from Vital River Laboratory Animal Technology (Beijing, China). To induce tumor formation, 5 × 106 tumor cells were injected into the tail vein. Eight weeks after injection, liver and lung samples were collected and subjected to histological examination. Visible lung metastases were measured and counted using a microscope. All experimental procedures involving animals were carried out in accordance with the Guide for the Care and Use of Laboratory Animals (NIH publication no. 80‐23, revised 1996) and were undertaken according to the institutional ethical guidelines for animal experiments.
Chromatin immunoprecipitation
Cells were cross‐linked with 1% formaldehyde at 37°C for 10 minutes, resuspended in lysis buffer (50 mmol/L Tris‐HCl [pH = 8.1], 10 mmol/L EDTA, 1% SDS, and protease inhibitor cocktail). DNA was sheared by sonication to lengths between 200 and 1000 bp. Protein‐DNA complexes were precipitated by anti‐MORC2 antibody (Bethyl Laboratories) and control IgG, respectively, followed by the elution of the complex from the Ab. Real‐time PCR was carried out with primers specific for the NDRG1 promoter region: 5′‐CCGAGCTGGTGAGACCTACA‐3′(sense) and 5′‐TTGTGGGTCGGCTCTGCAGT‐3′(antisense).
Cell viability and cell cycle analysis
Cells were seeded in 96‐well plates (2000 cells/well) in triplicate, and cell viability was evaluated by CCK‐8 (Dojindo Laboratories, Kumamoto, Japan). For cell cycle analyses, cells were harvested and fixed in 75% ethanol overnight. After washing with PBS, cells were stained with propidium iodide and analyzed on a BD FACSVerse flow cytometer (BD Biosciences).
Statistical analysis
The statistical analysis was carried out using SPSS (17.0) software (SPSS, Chicago, IL, USA). Data of MORC2 and NDRG1 expression in CRC were analyzed by Spearman's rank correlation coefficient test. Mann‐Whitney U test was used to analyze the association between MORC2 or NDRG1 expression and clinical features. Survival curve was estimated by the Kaplan‐Meier method. The statistical significance of difference was analyzed by ANOVA. Statistical significance was defined as P < 0.05. All experiments were repeated 3 times, and data were expressed as the mean ± SD from a representative experiment.
RESULTS
MORC2 downregulates NDRG1 in colorectal cancer cells
To search MORC2 target genes, we undertook DNA microarray hybridization (Affymetrix, Santa Clara, CA, USA) using stable pcDNA3.1 or MORC2‐overexpressed gastric cancer SGC‐7901 cells and found various MORC2 target genes, most of which were downregulated. The genes that were downregulated or upregulated by MORC2 are listed in Tables S1 and S2. NDRG1 was one of the genes that was downregulated by MORC2 in gastric cancer SGC‐7901 cells. Importantly, emerging evidence suggests that NDRG1 can serve as a biomarker for colorectal cancer and has tumor suppressive properties in colorectal cancer. These findings prompted us to investigate whether MORC2 regulates NDRG1 in CRC cells. Quantitative (q)RT‐PCR and western blot analyses were carried out in stable empty vector or MORC2‐overexpressed HT‐29 and SW‐620 cells. The results showed that MORC2 overexpression downregulated the mRNA and protein level of NDRG1 (Figure A,B). In contrast, stable knockdown of MORC2 increased the expression of NDRG1 mRNA and protein (Figure C‐F).
MORC2 binds to NDRG1 promoter and inhibits NDRG1 promoter activity
In order to elucidate whether the decrease in NDRG1 mRNA is dependent on MORC2 as a regulator of transcription, we examined the regulation of the NDRG1 promoter. As a reporter, we used the −759 to +69 fragment of the NDRG1 promoter fused to the luciferase reporter gene. This region has been shown to be sufficient for the transcription induction of the NDRG1 gene in neuroblastoma cells. The luciferase assay results showed that MORC2 downregulated NDRG1 promoter activity in a dose‐dependent manner (Figure A,B). To determine which region is required for the repression function of MORC2 on NDRG1 transcription, a series of 5′ promoter deletion mutants of the NDRG1 promoter proximal to the transcriptional initiation site were transfected into HEK‐293 cells with or without MORC2. Significant decrease of NDRG1 promoter activity was observed in pGL3‐NDRG1(−759/+69) and pGL3‐NDRG1(−446/+69), but not in pGL3‐NDRG1(−213/+69) (Figure C), indicating that the −446 to −213 bp region played an important role in the suppression function of MORC2 on NDRG1 transcription. In order to verify the binding of MORC2 to the NDRG1 promoter, ChIP assays were carried out. The results showed that MORC2 bound to the −446 to −213 bp region of NDRG1 promoter, but not to the −759 to −446 bp or −213 to +69 bp region in Flag‐MORC2 transfected cells (Figure D). Our ChIP‐qPCR results verified that endogenous MORC2 also bound to the −446 to −213 bp region of NDRG1 promoter (Figure E).
MORC2 inhibits NDRG1 expression by decreasing histone acetylation level of NDRG1 promoter
Our previous work showed that MORC2 downregulated CAIX transcription by recruiting HDAC4. Sirtuin 1, a member of the class III HDACs, has been implicated in the carcinogenesis and progression of various cancers, so we wondered whether SIRT1 participates in MORC2‐mediated NDRG1 transcriptional regulation. Then we used sirtinol, a specific inhibitor of SIRT1, to treat stable empty vector or MORC2 overexpressed SW‐620 cells. Obviously, the inhibition of NDRG1 by MORC2 was inverted after treatment with sirtinol (Figure A,B). To further detect whether SIRT1 plays a role in downregulation of NDRG1, qRT‐PCR and western blot were carried out. As can been seen in Figure C,D, depletion of the endogenous SIRT1 by specific shRNA resulted in an increase of NDRG1 mRNA and protein level. Moreover, SIRT1 inhibited NDRG1 promoter activity in a dose‐dependent manner (Figure E). To test whether MORC2 could physically interact with SIRT1, we carried out immunoprecipitation followed by western blot analysis. We transfected HCT‐116 cells with Flag‐tagged MORC2 or Flag empty vector. The results indicated that Flag‐tagged MORC2 could interact with endogenous SIRT1 in vivo (Figure F). To investigate how the NDRG1 promoter activity was affected by MORC2 and SIRT1, we undertook luciferase assays. The results showed that MORC2 was able to inhibit NDRG1 promoter activity independently and cumulatively with SIRT1 (Figure G). To detect the influence of MORC2 on the acetylation level of histone H3 and H4 in the NDRG1 promoter, ChIP‐qPCR was carried out. The results showed that MORC2 overexpression led to a significant decrease of H4Ac and a slight decrease of H3Ac at the −446 to −213 bp region in NDRG1 promoter (Figure H). These data suggest that MORC2 and SIRT1 inhibit NDRG1 transcription cumulatively and MORC2 inhibits NDRG1 expression by decreasing the histone acetylation level of the NDRG1 promoter.
NDRG1 is essential in MORC2‐mediated promotion of CRC cell migration and invasion
To study the role of MORC2 in CRC cell migration and invasion, we used HT‐29 and SW‐480 cells that stably express MORC2 and analyzed their migratory capacities in vitro. Compared with the control, MORC2 overexpression enhanced the migration of cells, as shown by Transwell assays (Figure A) and wound healing assays (Figures B,S1). We carried out CCK‐8 cell growth assays and flow cytometric analyses to evaluate whether MORC2 affected cell proliferation and the cell cycle. As shown in Figure S2A, MORC2 overexpression or MORC2 and SIRT1 overexpression did not affect cell proliferation in SW‐480 cells (Figure S2A). The flow cytometric analysis showed that MORC2 significantly inhibited cell cycle progression in HCT‐116 cells (Figure S2B) and slightly inhibited cell cycle progression in SW‐480 cells (Figure S2C). As NDRG1 is a potent suppressor of metastasis in colon cancer, and MORC2 inhibit NDRG1 expression in CRC cells, we wondered whether NDRG1 is required for MORC2‐mediated promotion of CRC cell migration and invasion. We examined the effect of NDRG1 knockdown on shMORC2‐induced reduction of cell migration and invasion capacities in SW‐480 cells. As shown in Figure C,D, NDRG1 knockdown abrogated the shMORC2‐induced reduction of the migration and invasion ability of SW‐480 cells, indicating that NDRG1 is essential in MORC2‐mediated promotion of CRC cell migration and invasion.
NDRG1 is required for MORC2‐mediated promotion of CRC cell lung metastasis
To further examine the effect of MORC2 and NDRG1 on CRC cell metastasis in vivo, shNC, shMORC2, and shMORC2 + shNDRG1 SW‐480 cells were injected into nude mice through the tail vein. Eight weeks later, mice were killed and necropsied to find metastases in the lung. Indeed, SW‐480 cells stably silencing MORC2 developed less metastases in the lung of nude mice than the shNC group and shMORC2 + shNDRG1 group (Figure A,B). Lung metastasis was detected in all of the 8 mice in the shNC group, 1 of 9 mice in the shMORC2 group, and 8 of 9 mice in the shMORC2 + shNDRG1 group (Figure C). Hematoxylin–eosin and immunohistochemical staining confirmed that shMORC2 group resulted in much less marked metastatic spread to the lungs of the mice compared with the shNC group and shMORC2 + shNDRG1 group (Figure D). These findings indicate that NDRG1 is required for MORC2‐mediated promotion of CRC cell pulmonic metastasis.
Negative correlation of MORC2 and NDRG1 expression in CRC samples
In order to detect the expression of MORC2 and NDRG1 and the correlation between MORC2 and NDRG1 in CRC samples, 36 noncancerous tissues and 119 CRC tissues were immunostained for MORC2 and NDRG1. Representative images are shown in Figure A. Unlike noncancerous tissue, high expression level of MORC2 and low expression level of NDRG1 were observed in CRC tissues (Figure A). More importantly, the MORC2 expression level was negatively correlated with the NDRG1 expression level (Figure B, r = −0.782, P < 0.0001). To better understand the correlation between MORC2 or NDRG1 expression and progression of CRC, these samples were divided into 2 groups based on MORC2 or NDRG1 levels (histological score). The data showed that high expression of MORC2 was significantly associated with lymph node metastasis (P = 0.019) and poor pTNM stage (P = 0.02) (Table ), but not with tumor size (P = 0.916). The data also showed that decreased expression of NDRG1 was significantly related to lymph node metastasis (P = 0.003) (Table ). Moreover, Kaplan‐Meier curves using The Cancer Genome Atlas database showed that colon cancer patients with higher MORC2 expression had a significantly shorter overall survival (P = 0.033, Figure C). Taken together, these data indicate that MORC2 is overexpressed in CRC tissues, leading to the decreased expression of NDRG1 and influencing the lymph node metastasis of CRC. The expression of MORC2 correlates with poor prognosis in colon cancer patients.
Feature | n | MORC2 expression | P value | |
Weak | Strong | |||
Age (years) | ||||
<65 | 63 | 13 | 50 | 0.916 |
≥65 | 56 | 12 | 44 | |
Gender | ||||
Male | 66 | 10 | 56 | 0.081 |
Female | 53 | 15 | 38 | |
Tumor size (cm) | ||||
<5 | 56 | 12 | 44 | 0.916 |
≥5 | 63 | 13 | 50 | |
Histological grade | ||||
Poorly | 17 | 6 | 11 | 0.136 |
Moderately | 88 | 17 | 71 | |
Well | 14 | 2 | 12 | |
Depth of invasion (pT) | ||||
T1,T2 | 18 | 6 | 12 | 0.165 |
T3,T4 | 101 | 19 | 82 | |
Lymph node metastasis (pN) | ||||
No | 76 | 21 | 55 | 0.019* |
Yes | 43 | 4 | 39 | |
Distant metastasis (pM) | ||||
No | 108 | 24 | 84 | 0.310 |
Yes | 11 | 1 | 10 | |
Pathological stage (pStage) | ||||
Stages I, II | 71 | 20 | 51 | 0.020 * |
Stages III, IV | 48 | 5 | 43 |
Bold values indicate statistical significance,*P < 0.05
Feature | n | NDRG1 expression | P value | |
Weak | Strong | |||
Age (years) | ||||
<65 | 63 | 43 | 20 | 0.293 |
≥65 | 56 | 33 | 23 | |
Gender | ||||
Male | 66 | 42 | 24 | 0.954 |
Female | 53 | 34 | 19 | |
Tumor size (cm) | ||||
<5 | 56 | 38 | 18 | 0.395 |
≥5 | 63 | 38 | 25 | |
Histological grade | ||||
Poorly | 17 | 15 | 2 | 0.239 |
Moderately | 88 | 51 | 37 | |
Well | 14 | 10 | 4 | |
Depth of invasion (pT) | ||||
T1,T2 | 18 | 14 | 4 | 0.184 |
T3,T4 | 101 | 62 | 39 | |
Lymph node metastasis (pN) | ||||
No | 76 | 41 | 35 | 0.003 * |
Yes | 43 | 35 | 8 | |
Distant metastasis (pM) | ||||
No | 108 | 67 | 41 | 0.195 |
Yes | 11 | 9 | 2 | |
Pathological stage (pStage) | ||||
Stages I, II | 71 | 44 | 27 | 0.603 |
Stages III, IV | 48 | 32 | 16 |
Bold values indicate statistical significance.*P < 0.05.
DISCUSSION
In our previous study, we found that MORC2 could act as a transcriptional repressor and inhibit the transcription of a number of target genes, including CAIX, p21Waf/Cip1, and ArgBP2 in gastric cancer cells. We also reported the potential role of the PAK1‐MORC2 axis in gastric tumorigenesis. Other studies revealed that MORC2 promoted chromatin remodeling during the DNA‐damage response and had a cytosolic role in lipogenesis and adipogenesis. However, little is known about its functions in colorectal cancer. Here, we show that MORC2 is upregulated in CRC tissues (Figure A), which is consistent with the findings of another group. We provide the first evidence that MORC2 promotes CRC cell migration and invasion in vitro and metastasis in vivo (Figures ). Our data also show that MORC2 inhibits cell cycle progression in HCT‐116 cells (Figure S2B), indicating that increased migration and invasion by MORC2 are not due to increased proliferation.
NDRG1 is a member of the NDRG family. Previous studies have revealed that NDRG1 shows tissue‐specific expression patterns in different human cancers. NDRG1 is downregulated in colorectal cancer, prostate cancer, and glioma, whereas it is upregulated in breast cancer, liver cancer, and lung cancer. Our study shows the low expression of NDRG1 in colorectal cancer samples compared with noncancerous tissues (Figure A), which is consistent with a previous study. NDRG1 plays pleiotropic roles in tumor metastasis depending on cell context. It inhibits tumor progression and metastasis in colon, prostate, and breast cancers, indicating that NDRG1 is an effective metastasis suppressor in these cancers. However, NDRG1 promotes portal vein invasion and intrahepatic metastasis in hepatocellular carcinoma. In this study, we show that NDRG1 prevents the migration and invasion of CRC cells (Figure C,D). Importantly, NDRG1 is essential in MORC2‐mediated enhancement of CRC cell migration and invasion (Figure C,D).
NDRG1 is regulated by a variety of factors and stress related to cancer progression, including metal ions, cytokines, oncogenes, tumor suppressors, hormones, and a hypoxic microenvironment. As the name implies, N‐myc transcriptionally repress NDRG1 expression in human neuroblastomas cells. The von Hippel‐Lindau protein has been reported to inhibit NDRG1 expression in renal cancer cells. However, NDRG1 expression is able to be promoted by hypoxia‐inducible factor‐1α, p53, PTEN, and eIF3a. Here, our data show that MORC2 downregulated NDRG1 promoter activity in a dose‐dependent manner (Figure A,B). Through a series of deletions of the NDRG1 promoter luciferase constructs, we found that the −446 to −213 bp region was important for the promoter activities of NDRG1 (Figure C). Moreover, ChIP assay indicated the enrichment of MORC2 on the NDRG1 promoter (Figure D). The direct role of MORC2 in the negative regulation of the NDRG1 gene was further evidenced by both overexpression and knockdown of MORC2 in CRC cells (Figure ). These data suggest that NDRG1 is a novel target gene of MORC2 in CRC cells.
Reversible acetylation is one of the best‐characterized post‐transcriptional modifications of the histone amino termini. In general, histone acetylation is correlated with a positive transcriptional regulation, whereas histone deacetylation is associated with transcriptional repression. It has been reported that HDAC inhibitor trichostatin A promotes NDRG1 mRNA expression, indicating that histone deacetylation is involved in transcriptional silencing of NDRG1. Sirtuin 1 is a highly conserved NAD(+)‐dependent HDAC that is upregulated in colon cancers. Sirtuin 1 could promote cancer cell proliferation by enhancing expression of several epigenetic silenced tumor suppressor genes. Sirtuin 1 is known to promote NDRG1 expression in hypoxic trophoblasts. However, the influence of SIRT1 on NDRG1 expression in cancer cells remains unknown. Here, we found that sirtinol treatment inverted the inhibition of NDRG1 by MORC2 (Figure A,B), indicating that SIRT1 is involved in the MORC2‐mediated NDRG1 transcriptional inhibition. Moreover, we showed that SIRT1 downregulated NDRG1 mRNA, protein level, and promoter activity (Figure C‐E).
Sirtuin 1 interacts with many important transcription factors or cofactors to repress target gene transcription through deacetylation, which is involved in tumorigenesis and cancer progression. In this study, we showed that SIRT1 interacted with MORC2 and inhibited NDRG1 transcription independently and cumulatively with MORC2 (Figure F,G). Because SIRT1 is a member of the class III HDACs, we speculate that SIRT1 might contribute to MORC2‐mediated NDRG1 transcription inhibition through its histone deacetylation function. Further studies are needed to verify the speculation.
Based on previous knowledge, as well as findings from this study, we propose a hypothesized model (Figure D). In our model, MORC2 binds the −446 to −213 bp region of NDRG1 promoter, and interacts with SIRT1, leading to a decrease in the histone H3 and H4 acetylation level of the NDRG1 promoter and thus the transcriptional repression of the NDRG1 gene (Figure D).
In conclusion, we recognize NDRG1 as a novel target gene of MORC2 in CRC cells. MORC2 inhibits NDRG1 transcription by binding the −446 to −213 bp region of NDRG1 promoter and interacting with SIRT1. Moreover, MORC2 promotes the migration and invasion, as well as the pulmonic metastasis, of CRC cells through NDRG1. Notably, MORC2 expression correlates negatively with NDRG1 expression in CRC tissues and high expression of MORC2 is significantly associated with lymph node metastasis and poor pTNM stage. Our findings highlight a novel mechanism of MORC2 in the progression of CRC and provide MORC2 as a potential therapeutic target for CRC.
ACKNOWLEDGMENTS
This work was supported by the National Natural Science Foundation of China (31271389, 31571457 and 31771553) and the Scientific and Technological Research Project of Liaoning Provincial Education Department (LK201630). Human studies were approved by the Ethics Committee of the Medical Faculty of the China Medical University. All procedures in animal experiments were approved by the Animal Ethics Committee of the China Medical University.
CONFLICT OF INTEREST
Authors declare no conflict of interests for this article.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2019. This work is published under http://creativecommons.org/licenses/by-nc-nd/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details


1 Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, China
2 Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, China