Open Access

Downregulation of O‑linked N‑acetylglucosamine transferase by RNA interference decreases MMP9 expression in human esophageal cancer cells

  • Authors:
    • Zhe Qiao
    • Chengxue Dang
    • Bin Zhou
    • Shaomin Li
    • Wei Zhang
    • Jiantao Jiang
    • Jin Zhang
    • Yuefeng Ma
    • Ranran Kong
    • Zhenchuan Ma
  • View Affiliations

  • Published online on: April 7, 2016     https://doi.org/10.3892/ol.2016.4428
  • Pages: 3317-3323
  • Copyright: © Qiao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

O-linked N-acetylglucosamine transferase (OGT) catalyzes O-linked glycosylation (O-GlcNAcylation). O-GlcNAcylation is a post‑translational carbohydrate modification of diverse nuclear and cytosolic proteins by the addition of O‑linked β‑N‑acetylglucosamine. It was recently demonstrated that OGT and the level of O‑GlcNAcylation are upregulated in esophageal cancer; however, the physiological consequences of this upregulation remain unknown. The current study reports that OGT knockdown by short hairpin RNA (shRNA) did not affect cell viability; however, cell migration in esophageal cancer Eca‑109 cells was significantly reduced. OGT‑specific shRNA vectors efficiently decreased the protein and mRNA levels of OGT and the RL2 level (a marker of O‑GlcNAcylation levels) in Eca‑109 esophageal cancer cells. In addition, colony formation and cell proliferation assays demonstrated that OGT‑specific shRNA decreased the proliferation of Eca‑109 cells; however, there was no significant statistical difference between OGT‑specific shRNA and control shRNA. Notably, transwell assays demonstrated that the migratory ability of Eca‑109 cells was significantly suppressed following knockdown of the OGT gene. Correspondingly, western blot analyses demonstrated that OGT knockdown significantly downregulated the expression of matrix metalloproteinase 9 (MMP9) in Eca‑109 cells. These results suggest that OGT may promote the migration, invasion and metastasis of esophageal cancer cells by enhancing the stability or expression of MMP9.

Introduction

Esophageal cancer is one of the most common types of cancer worldwide with >480,000 new cases diagnosed annually (1,2). Globally, the disease accounts for ~400,000 cancer-associated mortalities annually (3) with a 5-year survival rate of <20% (4). According to the National Comprehensive Cancer Network guidelines (5), surgery is the optimal treatment choice, however, chemotherapy and radiotherapy may also be administered (6). Squamous cell carcinoma is one of the most common types of cancer and this reacts poorly to common chemotherapy compared with other types of cancer, such as adenocarcinoma (7). Novel studies are required to provide evidence for effective therapy on this disease. Glycolysis and the uptake of glucose are enhanced in cancer cells in order to meet the increased energy requirements of the rapidly proliferating cells, including esophageal caner cells, which are known of elevated Wurberg Effects. A fraction of the glucose in cancer cells is metabolized by the hexosamine biosynthetic pathway (HBP) (810). The HBP regulates enzymatic O-linked glycosylation (O-GlcNAcylation), a post-translational carbohydrate modification of diverse nuclear and cytosolic proteins by the addition of O-linked β-N-acetylglucosamine (O-GlcNAc). O-GlcNAcylation of a protein alters the protein's stability, intracellular localization and function. O-GlcNAcylation serves important roles in an array of normal biological processes, and its dysregulation is involved in a variety of human diseases, including diabetes mellitus (11,12) and various neurological disorders (13). Several GlcNAcylated tumor-associated proteins have recently been identified, including c-Myc (14) and p53 (15), each one of the most important oncogenes and tumor suppressor genes, respectively. These findings suggest that O-GlcNAcylation serves a significant role in oncogenesis and tumor progression (1618). In our previous study, OGT and a marker of O-GlcNAcylation levels, mouse monoclonal anti-O-linked N-acetylglucosamine antibody (RL2), were observed to be upregulated in esophageal cancer (19). However, the physiological consequences of this upregulation remain to be determined.

In the current study, RNA interference (RNAi) was used to knock down OGT in esophageal cancer Eca-109 cells, and the cell proliferation and migration capabilities were subsequently assessed. The results demonstrated that cell viability was unaffected by RNAi knockdown of OGT in Eca-109 esophageal cancer cells; however, the knockdown significantly reduced cell migration and markedly decreased matrix metalloproteinase 9 (MMP9) levels in knockdown cells.

Materials and methods

Reagents

Polyclonal rabbit anti-human OGT antibody was purchased from ProteinTech Group, Inc. (Chicago, IL, USA; catalog no., 11576-2-AP) and mouse monoclonal anti-human MMP9 antibody was purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA; catalog no., sc-12759). Mouse monoclonal RL2 antibody (catalog no., MA1-072) and the Invitrogen™ BLOCK-iT™ Pol II miR RNAi Expression Vector Kit (catalog no., K4935-00) were purchased from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Transwell plates were purchased from Corning Incorporated (Corning, NY, USA).

Cell culture and RNAi

The human esophageal cancer Eca-109 cell line was obtained from the Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China) and cultured in RPMI-1640 medium (Thermo Fisher Scientific, Inc.) containing 10% fetal bovine serum (FBS; HyClone; GE Healthcare Life Sciences, Logan, UT, USA), 100 U/ml penicillin and 100 µg/ml streptomycin (Gibco; Thermo Fisher Scientific, Inc.). Cells were incubated at 37°C in an atmosphere of 5% CO2. Three shRNAs were designed with the assistance of the online RNAi software BLOCK-iT™ RNAi Designer (http://rnaidesigner.lifetechnologies.com/rnaiexpress/). The names and sequences of the three shRNA segments are listed in Table I. Eca-109 cells were transfected with an RNAi-OGT-EmGFP Vector according to the BLOCK-iT™ Pol II miR RNAi Expression Vector Kit manufacturer's instructions. Negative control cells were composed of Eca-109 cells transfected with negative control shRNA, as described in Table I.

Table I.

shRNA segment names and sequences.

Table I.

shRNA segment names and sequences.

Sequence

shRNA segmentTop strandBottom strand
RNAi-OGTA 5′-TGCTGGATGTGCCAACTCAGC 5′-CCTGGATGTGCCAACAGCTAA
TAACCGTTTTGGCCACTGACTG CCGTCAGTCAGTGGCCAAAACG
ACGGTTAGCTGTTGGCACATC-3′ GTTAGCTGAGTTGGCACATCC-3′
RNAi-OGTB 5′-TGCTGTAGAGTAGGCATCAGC 5′-CCTGTAGAGTAGGCAAGCAA
AAAGGGTTTTGGCCACTGACTG AGGGTCAGTCAGTGGCCAAAAC
ACCCTTTGCTTGCCTACTCTA-3′ CCTTTGCTGATGCCTACTCTAC-3′
RNAi-OGTC 5′-TGCTGAATACTGCTCAGCAAC 5′-CCTGAATACTGCTCAAACTTC
TTCAGGTTTTGGCCACTGACTG AGGTCAGTCAGTGGCCAAAACC
ACCTGAAGTTTGAGCAGTATT-3′ TGAAGTTGCTGAGCAGTATTC-3′
Negative control 5′-GAAATGTACTGCGCGTGGAG
ACGTTTTGGCCACTGACTGACG
TCTCCACGCAGTACATTT-3′

[i] shRNA, short hairpin RNA; RNAi, RNA interference; OGT, O-linked N-acetylglucosamine transferase.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from the Eca-109 cells using TRIzol reagent according to the manufacturers' instructions (Takara Bio, Otsu, Japan), and cDNA was synthesized using GoScript™ Reverse Transcription System, according to the manufacturer's instructions (Promega Corporation, Madison, WI, USA; catalog no., A5001). In total, 950 ng of total RNA from each sample was used for the synthesis of cDNA. The target gene mRNA levels relative to the GAPDH control were determined by qPCR in an Applied Biosystems™ 7900 HT Fast Real-Time PCR System (Thermo Fisher Scientific, Inc.) using the GoTaq® qPCR Master Mix (Promega Corporation; catalog no., A6001). The data were analyzed using the 2−ΔΔCq method (20). The genes of interest were OGT1, OGT2, OGT3 and MMP9. The primers were designed using BLOCK-iT™ RNAi Designer software (Invitrogen; Thermo Fisher, Inc.) and synthesized by BGI Shenzhen (Shenzhen, China). The following primer sequences were used: MMP9, forward 5′-CCTGGAGACCTGAGAACCAATCT-3′ and reverse 5′-CCACCCGAGTGTAACCATAGC-3′. PCR was performed under the following conditions: Initial denaturation at 95°C for 30 sec, followed by 40 cycles of 95°C for 5 sec, and elongation at 60°C for 30 sec. Each of the cDNA samples from each group were assessed for gene expression in duplicate. All the tests were repeated for 3 times.

Western blot

Total protein was extracted on ice from Eca-109 cells with cell lysis buffer (Cell Signaling Technology, Inc., Danvers, MA, USA). Equal amounts of protein were separated by 10% SDS-PAGE and then transferred to a polyvinylidene difluoride membrane (catalog no., P2813; Sigma-Aldrich, St. Louis, MO, USA). Membranes were blocked with 2% fat-free milk in phosphate-buffered saline (PBS) at room temperature for 1 h and then probed with primary antibodies (dilution, 1:200) overnight at 4°C in PBS containing 0.1% Tween 20 (PBST) and 1% fat-free milk. Following the overnight incubation, membranes were washed four times in PBST and then incubated with anti-rabbit horseradish peroxidase-conjugated secondary antibody (dilution, 1:200; catalog no., 1662408; Bio-Rad Laboratories, Inc., Hercules, CA, USA). Signals were developed using enhanced chemiluminescence reagents (Amersham Pharmacia Biotech, Piscataway, NJ, USA). Densitometric analysis was performed to quantify the results using Image Pro Plus 6.0 software (Media Cybernetics, Inc., Rockville, MD, USA).

Cell proliferation assay

Cells were seeded into 96-well plates at a density of 5×104/ml in 100 µl of medium, and grown for 24 h. Following transfection for 24–72 h, the CellTiter 96® AQueous One Solution Cell Proliferation Assay System (Promega Corporation) was added (20 µl/well) and incubated for 90 min. Finally, the optical densities (OD) were measured at 492 nm with a scanning microplate reader (EnSpire® Multimode Plate Reader; Perkin Elmer, Waltham, MA, USA).

Colony formation assay

A total of 200 cells were plated in a 10-cm Petri dish. Two weeks following transfection, the cells were washed with PBS, fixed with cold methanol for 15 min at −20°C, and stained with 1% crystal violet (Thermo Fisher Scientific, Inc.) in 25% methanol for 15 min. The dishes were thoroughly washed with water, and the blue colonies were counted.

Transwell chamber assay

Transwell chamber migration assays were performed using Nunc 24-well 8.0-µm pore Transwell plates (Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. Eca-109 cells were plated at a density of 5×104cells/ml in each well with RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.) free of FBS, and 500 µl culture medium containing 10% FBS was added to the bottom of the 24-well plate. Following incubation for 24 h, non-invading cells were removed from the upper surface of the membrane using a cotton-tipped swab. The invading cells were subsequently fixed in methanol for 10 min and stained with 0.1% crystal violet hydrate (Sigma-Aldrich) for 30 min. The stained cells were counted as cells per field at 10× magnification in 3 fields (CX31 microscope; Olympus Corporation, Tokyo, Japan).

Statistical analysis

All data are presented as the mean ± standard error of the mean of at least three individual experiments. Student's t-test was used to analyze the differences between groups. Statistical significance was determined using SPSS software version 13.0 (SPSS Inc., Chicago, IL, USA). P<0.05 was considered to indicate statistically significant differences.

Results

Silencing of OGT by RNAi in Eca-109 cells

Three pairs of double-stranded OGT oligonucleotides (RNAi-OGTA, RNAi-OGTB and RNAi-OGTC) were designed and cloned into the transfection vector RNAi-OGT-EmGFP. To assess the transfection efficiency, Eca-109 cells were transfected with RNAi-OGT-EmGFP and EmGFP fluorescence was observed after 7-, 14- and 21-day incubations. After 7 days of incubation, >90% of the cells fluoresced green, and the cells remained strongly fluorescent until at least 21 days (Fig. 1A), indicating efficient transfection of the construct.

To assess the RNAi efficiency of the RNAi-OGT constructs, the OGT mRNA levels were measured using RT-qPCR following three days of transfection. The results indicated that RNAi-OGTA, RNAi-OGTB and RNAi-OGTC decreased OGT mRNA levels compared with negative control shRNA transfected cells. Furthermore, the strongest OGT knockdown was induced by RNAi-OGTB (Fig. 1B). Consistent with OGT mRNA level alterations, western blot analysis demonstrated that OGT protein levels were also markedly downregulated by RNAi-OGTA, RNAi-OGTB and RNAi-OGTC. Once again, the strongest OGT knockdown was induced by RNAi-OGTB (Fig. 1C and D). Therefore, the subsequent functional analyses of OGT knockdown in Eca-109 cells were performed using the RNAi-OGTB construct.

OGT knockdown by RNAi decreases O-GlcNAcylation in Eca-109 cells

In our previous study, OGT and the O-GlcNAcylation marker RL2 were observed to be highly expressed in esophageal cancer (19). The present study aimed to determine whether RNAi silencing of OGT leads to decreased O-GlcNAcylation in esophageal cancer cells. RL2 protein levels were assessed by western blot analysis of Eca-109 cells transfected with RNAi-OGTB for 72 h, revealing that, compared with cells transfected with the control shRNA, RNAi-OGTB-transfected cells exhibited significantly decreased RL2 protein levels (P=0.002; Fig. 2).

RNAi knockdown of OGT does not affect the viability of Eca-109 cells

Colony formation assays demonstrated that RNAi-OGTB transfection of Eca-109 cells led to decreased colony formation. However, the observed decrease in colony formation did not reach statistical significance when compared with the negative control shRNA transfected cells (P=0.232; Fig. 3A–C). Similarly, cell proliferation assays revealed that RNAi-OGTB transfection of Eca-109 cells did not significantly inhibit cell proliferation when compared with negative control shRNA transfected cells (P=0.728; Fig. 3D). These results indicate that the RNAi knockdown of OGT had no effect on Eca-109 cell viability.

RNAi knockdown of OGT inhibits Eca-109 cell migration

To investigate whether RNAi knockdown of OGT affects cell migration, Transwell assays were performed using Eca-109 cells transfected with RNAi-OGTB for 72 h. In comparison with the control (Fig. 4A), RNAi-OGTB transfection significantly decreased cell migration (P<0.001; Fig. 4B and C), suggesting that OGT promotes cell migration.

RNAi knockdown of OGT downregulates MMP9 expression in Eca-109 cells

To investigate the underlying mechanism by which OGT downregulation inhibits cell migration, MMP9 mRNA and protein levels were assessed by RT-qPCR and western blot analysis, respectively, in Eca-109 cells transfected with RNAi-OGTB for 72 h. Compared to the control, RNAi-OGTB transfection significantly downregulated MMP9 mRNA levels (P<0.001; Fig. 5A). Correspondingly, western blot analysis also demonstrated the marked downregulation of MMP9 by RNAi-OGTB transfection (P=0.003; Fig. 5B and C).

Discussion

In this study, OGT was successfully knocked down using RNAi in human Eca-109 esophageal cancer cells. OGT downregulation resulted in a decrease of the O-GlcNAcylation marker RL2. OGT knockdown did not significantly alter cell viability; however, it did significantly reduce Eca-109 cell migration. Moreover, MMP9 mRNA and protein expression was significantly decreased following RNAi knockdown of OGT in Eca-109 cells. These findings suggest that OGT may promote the migration, invasion and metastasis of esophageal cancer cells by enhancing the stability or expression of MMP9.

Types of protein modification include phosphorylation, ubiquitination, glycosylation, nitrosylation and O-GlcNAcylation. O-GlcNAcylation is a kind of post-translational modification that targets diverse nuclear and cytosolic proteins by the cycling of a single O-linked β-N-acetylglucosamine on the hydroxyl groups of the serine and threonine residues of target proteins (17,18,21). O-GlcNAcylation is dynamically regulated by the polypeptides OGT and β-N-acetylglucosaminidase (OGA): OGT catalyzes the addition of O-linked β-N-acetylglucosamine from uridine diphosphate N-acetylglucosamine onto the hydroxyl group of a serine or threonine residue on the target protein substrate (22), whilst OGA is a neutral hexosaminidase with a catalytic site similar to a family of 84 glycoside hydrolases that specifically catalyze the removal of β-linked GlcNAc on their substrates (2325). O-GlcNAcylation is involved in cellular signal transduction pathways and regulates cell growth, proliferation and migration (26). Increased OGT expression and O-GlcNAcylation levels have been observed to promote tumorigenesis in numerous types of tissue, including lung, colon and breast cancers (27,28). Our group previously demonstrated that the expression of OGT was positively related to the level of O-GlcNAcylation, and that OGT expression and O-GlcNAcylation levels were increased in esophageal cancer when compared to normal esophageal tissues (19). These results led us to hypothesize that elevated OGT expression may promote the tumorigenesis and progression of esophageal cancer. To investigate this hypothesis, an RNAi knockdown of OGT in the Eca-109 human esophageal cell line was created.

The results of the present study demonstrated that neither the RNAi knockdown of OGT expression nor the decrease of O-GlcNAcylation significantly inhibited cellular proliferation. This negative result may be due to the incomplete depletion of OGT in these cells; in previous studies, complete deletion of OGT led to cell death (29,30). High levels of O-GlcNAcylation in breast cancer have recently been observed to promote metastasis, particularly to lymph nodes (31). Furthermore, in our previous report, high levels of O-GlcNAcylation were identified to be associated with lymph node metastasis in esophageal carcinoma (19). These results suggest that OGT may promote cancer cell metastasis. In accordance with this suggestion, the Transwell assay performed in the current study demonstrated that RNAi knockdown of OGT significantly reduced cell migration. Since matrix metalloproteinases (MMPs), including MMP9, serve crucial roles in cell migration (32,33), the effect of RNAi knockdown of OGT in Eca-109 cells on MMP9 expression was investigated. This revealed that MMP9 was significantly decreased in the RNAi-OGTB-transfected Eca-109 cells when compared with negative control shRNA transfected cell group, suggesting that knocking down the OGT gene in esophageal cancer may affect cellular migratory ability by decreasing MMP9, a phenomenon which may be predicted to be relative to the level of O-GlcNAcylation. Supporting this hypothesis, high OGT expression and high levels of O-GlcNAcylation have been observed to promote the metastasis of breast cancer cells (34). Also consistent with the current results, MMP9 has been reported to be involved in esophageal carcinoma metastasis (35), and high levels of O-GlcNAcylation have been associated with lymph node metastasis in esophageal carcinoma (19). However, the molecular mechanism by which O-GlcNAcylation affects MMP9 is largely unknown. O-GlcNAcylation has been reported to regulate extracellular signal-regulated kinases (ERKs) through MAPK/ERK kinases and Raf (36), and ERK regulates MMP2 and MMP9 (37,38). These facts imply a potential mechanism, in which OGT knockdown results in the downregulation of O-GlcNAcylation, which in turn results in the downregulation of MMP9 through the ERK pathway.

In summary, the current study demonstrated that RNAi is able to successfully downregulate OGT, and that OGT downregulation suppresses O-GlcNAcylation in Eca-109 cells. OGT knockdown did not affect cell viability; however, it did significantly reduce cell migration. Supporting this result, OGT knockdown was accompanied by a decrease in MMP9 expression. These findings suggest that O-GlcNAcylation may promote the migration, invasion and metastasis of esophageal cancer cells by enhancing the stability or expression of MMP9.

Acknowledgements

The present study was funded by the Scientific and Technological Research of Shaanxi Province of China (grant no. 2007K09-01), and was approved by the Ethics Review Committee of the Second Affiliated Hospital of Xi'an Jiaotong University (Xi'an, China).

References

1 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Pakzad R, Mohammadian-Hafshejani A, Khosravi B, Soltani S, Pakzad I, Mohammadian M, Salehiniya H and Momenimovahed Z: The incidence and mortality of esophageal cancer and their relationship to development in Asia. Ann Transl Med. 4:292016.PubMed/NCBI

3 

Parkin DM, Bray FI and Devesa SS: Cancer burden in the year 2000. The global picture. Eur J Cancer. 37(Suppl 8): S4–S66. 2001. View Article : Google Scholar : PubMed/NCBI

4 

van Rensburg SJ: Esophageal cancer, micronutrient malnutrition, and silica fragments. Lancet. 2:1098–1099. 1982. View Article : Google Scholar : PubMed/NCBI

5 

National Comprehensive Cancer Network: NCCN Guidelines and Clinical Resources: Guidelines for Treatment of Cancer by Site. Version 7. 2015.

6 

Brescia AA, Broderick SR, Crabtree TD, Puri V, Musick JF, Bell JM, Kreisel D, Krupnick AS, Patterson GA and Meyers BF: Adjuvant therapy for positive nodes after induction therapy and resection of esophageal cancer. Ann Thorac Surg. 101:200–210. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Zhang L, Ma J, Han Y, Liu J, Zhou W, Hong L and Fan D: Targeted therapy in esophageal cancer. Expert Rev Gastroenterol Hepatol. Feb 19–2016.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

8 

Zeidan Q and Hart GW: The intersections between O-GlcNAcylation and phosphorylation: Implications for multiple signaling pathways. J Cell Sci. 123:13–22. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Marshall S, Bacote V and Traxinger RR: Discovery of a metabolic pathway mediating glucose-induced desensitization of the glucose transport system. Role of hexosamine biosynthesis in the induction of insulin resistance. J Biol Chem. 266:4706–4712. 1991.PubMed/NCBI

10 

Copeland RJ, Bullen JW and Hart GW: Cross-talk between GlcNAcylation and phosphorylation: Roles in insulin resistance and glucose toxicity. Am J Physiol Endocrinol Metab. 295:E17–E28. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Housley MP, Rodgers JT, Udeshi ND, Kelly TJ, Shabanowitz J, Hunt DF, Puigserver P and Hart GW: O-GlcNAc regulates FoxO activation in response to glucose. J Biol Chem. 283:16283–16292. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Vosseller K, Wells L, Lane MD and Hart GW: Elevated nucleocytoplasmic glycosylation by O-GlcNAc results in insulin resistance associated with defects in Akt activation in 3T3-L1 adipocytes. Proc Natl Acad Sci USA. 99:5313–5318. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Yang JY, Gu JL, Shi JH, Liu F and Shen Q: The inhibitory effect of OGT gene expression on the level of tau phosphorylation. Prog Biochem Biophys. 36:346–352. 2009. View Article : Google Scholar

14 

Vervoorts J, Lüscher-Firzlaff J and Lüscher B: The ins and outs of MYC regulation by posttranslational mechanisms. J Biol Chem. 281:34725–34729. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Yang WH, Kim JE, Nam HW, Ju JW, Kim HS, Kim YS and Cho JW: Modification of p53 with O-linked N-acetylglucosamine regulates p53 activity and stability. Nat Cell Biol. 8:1074–1083. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Barone BB, Yeh HC, Snyder CF, Peairs KS, Stein KB, Derr RL, Wolff AC and Brancati FL: Long-term all-cause mortality in cancer patients with preexisting diabetes mellitus: A systematic review and meta-analysis. Jama. 300:2754–2764. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Torres CR and Hart GW: Topography and polypeptide distribution of terminal N-acetylglucosamine residues on the surfaces of intact lymphocytes. Evidence for O-linked GlcNAc. J Biol Chem. 259:3308–3317. 1984.PubMed/NCBI

18 

Hart GW, Housley MP and Slawson C: Cycling of O-linked beta-N-acetylglucosamine on nucleocytoplasmic proteins. Nature. 446:1017–1022. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Qiao Z, Dang C, Zhou B, Li S, Zhang W, Jiang J, Zhang J, Kong R and Ma Y: O-linked N-acetylglucosamine transferase (OGT) is overexpressed and promotes O-linked protein glycosylation in esophageal squamous cell carcinoma. J Biomed Res. 26:268–273. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Love DC and Hanover JA: The hexosamine signaling pathway: Deciphering the ‘O-GlcNAc code’. Sci STKE. 2005:re132005.PubMed/NCBI

22 

Haltiwanger RS, Holt GD and Hart GW: Enzymatic addition of O-GlcNAc to nuclear and cytoplasmic proteins. Identification of a uridine diphospho-N-acetylglucosamine: Peptide beta-N-acetylglucosaminyltransferase. J Biol Chem. 265:2563–2568. 1990.PubMed/NCBI

23 

Dong DL and Hart GW: Purification and characterization of an O-GlcNAc selective N-acetyl-beta-D-glucosaminidase from rat spleen cytosol. J Biol Chem. 269:19321–19330. 1994.PubMed/NCBI

24 

Cetinbas N, Macauley MS, Stubbs KA, Drapala R and Vocadlo DJ: Identification of Asp174 and Asp175 as the key catalytic residues of human O-GlcNAcase by functional analysis of site-directed mutants. Biochemistry. 45:3835–3844. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Gao Y, Wells L, Comer FI, Parker GJ and Hart GW: Dynamic O-glycosylation of nuclear and cytosolic proteins: Cloning and characterization of a neutral, cytosolic beta-N-acetylglucosaminidase from human brain. J Biol Chem. 276:9838–9845. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Rogacka D, Piwkowska A, Jankowski M, Kocbuch K, Dominiczak MH, Stepiński JK and Angielski S: Expression of GFAT1 and OGT in podocytes: Transport of glucosamine and the implications for glucose uptake into these cells. J Cell Physiol. 225:577–584. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Mi W, Gu Y, Han C, Liu H, Fan Q, Zhang X, Cong Q and Yu W: O-GlcNAcylation is a novel regulator of lung and colon cancer malignancy. Biochim Biophys Acta. 1812:514–519. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Caldwell SA, Jackson SR, Shahriari KS, Lynch TP, Sethi G, Walker S, Vosseller K and Reginato MJ: Nutrient sensor O-GlcNAc transferase regulates breast cancer tumorigenesis through targeting of the oncogenic transcription factor FoxM1. Oncogene. 29:2831–2842. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Ngoh GA, Facundo HT, Zafir A and Jones SP: O-GlcNAc signaling in the cardiovascular system. Circ Res. 107:171–185. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Slawson C, Copeland RJ and Hart GW: O-GlcNAc signaling: A metabolic link between diabetes and cancer? Trends Biochem Sci. 35:547–555. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Gu Y, Mi W, Ge Y, Liu H, Fan Q, Han C, Yang J, Han F, Lu X and Yu W: GlcNAcylation plays an essential role in breast cancer metastasis. Cancer Res. 70:6344–6351. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Akter H, Park M, Kwon OS, Song EJ, Park WS and Kang MJ: Activation of matrix metalloproteinase-9 (MMP-9) by neurotensin promotes cell invasion and migration through ERK pathway in gastric cancer. Tumour Biol. 36:6053–6062. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Song J, Wu C, Korpos E, Zhang X, Agrawal SM, Wang Y, Faber C, Schäfers M, Körner H, Opdenakker G, et al: Focal MMP-2 and MMP-9 activity at the blood-brain barrier promotes chemokine-induced leukocyte migration. Cell Rep. 10:1040–1054. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Slawson C, Pidala J and Potter R: Increased N-acetyl-beta-glucosaminidase activity in primary breast carcinomas corresponds to a decrease in N-acetylglucosamine containing proteins. Biochim Biophys Acta. 1537:147–157. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Wang YZ, Wu JN, Sun RQ and Wu WX: Expression of MMP-9 and MMP-9 mRNA in esophageal carcinoma its correlation with antioncogene p53. Jiang Su Da Xue Xue Bao. 2:113–116. 2005.(In Chinese).

36 

Kneass ZT and Marchase RB: Protein O-GlcNAc modulates motility-associated signaling intermediates in neutrophils. J Biol Chem. 280:14579–14585. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Hong S, Park KK, Magae J, Ando K, Lee TS, Kwon TK, Kwak JY, Kim CH and Chang YC: Ascochlorin inhibits matrix metalloproteinase-9 expression by suppressing activator protein-1-mediated gene expression through the erk1/2 signaling pathway: Inhibitory effects of ascochlorin on the invasion of renal carcinoma cells. J Biol Chem. 280:25202–25209. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Westermarck J and Kähäri VM: Regulation of matrix metalloproteinase expression in tumor invasion. Faseb J. 13:781–792. 1999.PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 11 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Qiao Z, Dang C, Zhou B, Li S, Zhang W, Jiang J, Zhang J, Ma Y, Kong R, Ma Z, Ma Z, et al: Downregulation of O‑linked N‑acetylglucosamine transferase by RNA interference decreases MMP9 expression in human esophageal cancer cells. Oncol Lett 11: 3317-3323, 2016
APA
Qiao, Z., Dang, C., Zhou, B., Li, S., Zhang, W., Jiang, J. ... Ma, Z. (2016). Downregulation of O‑linked N‑acetylglucosamine transferase by RNA interference decreases MMP9 expression in human esophageal cancer cells. Oncology Letters, 11, 3317-3323. https://doi.org/10.3892/ol.2016.4428
MLA
Qiao, Z., Dang, C., Zhou, B., Li, S., Zhang, W., Jiang, J., Zhang, J., Ma, Y., Kong, R., Ma, Z."Downregulation of O‑linked N‑acetylglucosamine transferase by RNA interference decreases MMP9 expression in human esophageal cancer cells". Oncology Letters 11.5 (2016): 3317-3323.
Chicago
Qiao, Z., Dang, C., Zhou, B., Li, S., Zhang, W., Jiang, J., Zhang, J., Ma, Y., Kong, R., Ma, Z."Downregulation of O‑linked N‑acetylglucosamine transferase by RNA interference decreases MMP9 expression in human esophageal cancer cells". Oncology Letters 11, no. 5 (2016): 3317-3323. https://doi.org/10.3892/ol.2016.4428