Skip to main content
Top
Published in: Tumor Biology 9/2016

01-09-2016 | Original Article

ZNF143 enhances metastasis of gastric cancer by promoting the process of EMT through PI3K/AKT signaling pathway

Authors: Song Wei, Linjun Wang, Lei Zhang, Bowen Li, Zheng Li, Qun Zhang, Jiwei Wang, Liang Chen, Guangli Sun, Qing Li, Hao Xu, Diancai Zhang, Zekuan Xu

Published in: Tumor Biology | Issue 9/2016

Login to get access

Abstract

The zinc finger protein 143 (ZNF143) is a transcription factor, which regulates many cell cycle-associated genes. ZNF143 expressed strongly in multiple solid tumors. However, the influence of ZNF143 on gastric cancer (GC) remains largely unknown. In this study, we investigated the ZNF143 mRNA level in GC tissues and cells by quantitative real-time PCR (qRT-PCR). The protein expression of ZNF143 in GC cells, and the signaling pathway proteins were detected by Western blotting. Transwell assay and wound healing assay were performed to explore the effects of ZNF143 for the migration ability of GC cells in vitro. We also performed the tail vein injection in nude mice with GC cells to explore the impact of ZNF143 on GC metastasis in vivo. ZNF143 was overexpressed in specimens of GC compared with adjacent normal tissues and increased more significantly in GC tissues of patients who had lymph node metastasis. Ectopic overexpression of ZNF143 enhanced GC migration, whereas ZNF143 knockdown suppressed this effect in vitro. In vivo, ZNF143 knockdown reduced distant metastasis of GC cells in nude mice. In addition, overexpression of ZNF143 reduced the expression of epithelial cell marker (E-cadherin) and induced the expression of mesenchymal cell marker (N-cadherin,Vimentin), Snail and Slug. We also found that ZNF143 enhanced GC cell migration by promoting the process of EMT through PI3K/AKT signaling pathway. In general, our findings show that ZNF143 expressed strongly in GC and enhanced migration of GC cells in vitro and in vivo. It is conceivable that ZNF143 could be a therapeutic genetic target for GC treatment.
Literature
1.
go back to reference Han C, Zhou Y, An Q, Li F, Li D, Zhang X, et al. Microrna-1 (mir-1) inhibits gastric cancer cell proliferation and migration by targeting met. Tumour Biol. 2015;36:6715–23.CrossRefPubMedPubMedCentral Han C, Zhou Y, An Q, Li F, Li D, Zhang X, et al. Microrna-1 (mir-1) inhibits gastric cancer cell proliferation and migration by targeting met. Tumour Biol. 2015;36:6715–23.CrossRefPubMedPubMedCentral
2.
go back to reference Arita T, Ichikawa D, Konishi H, Komatsu S, Shiozaki A, Shoda K, et al. Circulating long non-coding rnas in plasma of patients with gastric cancer. Anticancer research. 2013;33:3185–93.PubMed Arita T, Ichikawa D, Konishi H, Komatsu S, Shiozaki A, Shoda K, et al. Circulating long non-coding rnas in plasma of patients with gastric cancer. Anticancer research. 2013;33:3185–93.PubMed
3.
go back to reference Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in globocan 2012. International journal of cancer Journal international du cancer. 2015;136:E359–386.CrossRefPubMed Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in globocan 2012. International journal of cancer Journal international du cancer. 2015;136:E359–386.CrossRefPubMed
4.
go back to reference Lian G, Wei C, Wang D, Cui M, Wang Z, Liu X, et al. Protein profiling of Helicobacter pylori-associated gastric cancer. The American journal of pathology. 2014;184:1343–54.CrossRefPubMed Lian G, Wei C, Wang D, Cui M, Wang Z, Liu X, et al. Protein profiling of Helicobacter pylori-associated gastric cancer. The American journal of pathology. 2014;184:1343–54.CrossRefPubMed
5.
go back to reference Kosaka T, Davydova J, Ono HA, Akiyama H, Hirai S, Ohno S, et al. Imaging and antitumoral effect of a cyclo-oxygenase 2-specific replicative adenovirus for small metastatic gastric cancer lesions. Anticancer research. 2015;35:5201–10.PubMed Kosaka T, Davydova J, Ono HA, Akiyama H, Hirai S, Ohno S, et al. Imaging and antitumoral effect of a cyclo-oxygenase 2-specific replicative adenovirus for small metastatic gastric cancer lesions. Anticancer research. 2015;35:5201–10.PubMed
6.
go back to reference Li W, Zhou Y, Yang J, Zhang X, Zhang H, Zhang T, et al. Gastric cancer-derived mesenchymal stem cells prompt gastric cancer progression through secretion of interleukin-8. Journal of experimental & clinical cancer research : CR. 2015;34:52.CrossRefPubMedCentral Li W, Zhou Y, Yang J, Zhang X, Zhang H, Zhang T, et al. Gastric cancer-derived mesenchymal stem cells prompt gastric cancer progression through secretion of interleukin-8. Journal of experimental & clinical cancer research : CR. 2015;34:52.CrossRefPubMedCentral
8.
go back to reference Song IS, Oh NS, Kim HT, Ha GH, Jeong SY, Kim JM, et al. Human znf312b promotes the progression of gastric cancer by transcriptional activation of the k-ras gene. Cancer Res. 2009;69:3131–9.CrossRefPubMed Song IS, Oh NS, Kim HT, Ha GH, Jeong SY, Kim JM, et al. Human znf312b promotes the progression of gastric cancer by transcriptional activation of the k-ras gene. Cancer Res. 2009;69:3131–9.CrossRefPubMed
9.
go back to reference Wakasugi T, Izumi H, Uchiumi T, Suzuki H, Arao T, Nishio K, et al. Znf143 interacts with p73 and is involved in cisplatin resistance through the transcriptional regulation of DNA repair genes. Oncogene. 2007;26:5194–203.CrossRefPubMed Wakasugi T, Izumi H, Uchiumi T, Suzuki H, Arao T, Nishio K, et al. Znf143 interacts with p73 and is involved in cisplatin resistance through the transcriptional regulation of DNA repair genes. Oncogene. 2007;26:5194–203.CrossRefPubMed
10.
go back to reference Izumi H, Wakasugi T, Shimajiri S, Tanimoto A, Sasaguri Y, Kashiwagi E, et al. Role of znf143 in tumor growth through transcriptional regulation of DNA replication and cell-cycle-associated genes. Cancer Sci. 2010;101:2538–45.CrossRefPubMed Izumi H, Wakasugi T, Shimajiri S, Tanimoto A, Sasaguri Y, Kashiwagi E, et al. Role of znf143 in tumor growth through transcriptional regulation of DNA replication and cell-cycle-associated genes. Cancer Sci. 2010;101:2538–45.CrossRefPubMed
11.
go back to reference Myslinski E, Gerard MA, Krol A, Carbon P. A genome scale location analysis of human staf/znf143-binding sites suggests a widespread role for human staf/znf143 in mammalian promoters. The Journal of biological chemistry. 2006;281:39953–62.CrossRefPubMed Myslinski E, Gerard MA, Krol A, Carbon P. A genome scale location analysis of human staf/znf143-binding sites suggests a widespread role for human staf/znf143 in mammalian promoters. The Journal of biological chemistry. 2006;281:39953–62.CrossRefPubMed
12.
go back to reference Kawatsu Y, Kitada S, Uramoto H, Zhi L, Takeda T, Kimura T, et al. The combination of strong expression of znf143 and high mib-1 labelling index independently predicts shorter disease-specific survival in lung adenocarcinoma. Br J Cancer. 2014;110:2583–92.CrossRefPubMedPubMedCentral Kawatsu Y, Kitada S, Uramoto H, Zhi L, Takeda T, Kimura T, et al. The combination of strong expression of znf143 and high mib-1 labelling index independently predicts shorter disease-specific survival in lung adenocarcinoma. Br J Cancer. 2014;110:2583–92.CrossRefPubMedPubMedCentral
14.
go back to reference Han R, Xiong J, Xiao R, Altaf E, Wang J, Liu Y, et al. Activation of beta-catenin signaling is critical for doxorubicin-induced epithelial-mesenchymal transition in bgc-823 gastric cancer cell line. Tumour Biol. 2013;34:277–84.CrossRefPubMed Han R, Xiong J, Xiao R, Altaf E, Wang J, Liu Y, et al. Activation of beta-catenin signaling is critical for doxorubicin-induced epithelial-mesenchymal transition in bgc-823 gastric cancer cell line. Tumour Biol. 2013;34:277–84.CrossRefPubMed
15.
go back to reference Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90.CrossRefPubMed Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90.CrossRefPubMed
16.
go back to reference Xu W, Yang Z, Lu N. A new role for the pi3k/akt signaling pathway in the epithelial-mesenchymal transition. Cell adhesion & migration. 2015;9:317–24.CrossRef Xu W, Yang Z, Lu N. A new role for the pi3k/akt signaling pathway in the epithelial-mesenchymal transition. Cell adhesion & migration. 2015;9:317–24.CrossRef
17.
go back to reference Xu Q, Ma J, Lei J, Duan W, Sheng L, Chen X, et al. Alpha-mangostin suppresses the viability and epithelial-mesenchymal transition of pancreatic cancer cells by downregulating the pi3k/akt pathway. BioMed research international. 2014;2014:546353.PubMedPubMedCentral Xu Q, Ma J, Lei J, Duan W, Sheng L, Chen X, et al. Alpha-mangostin suppresses the viability and epithelial-mesenchymal transition of pancreatic cancer cells by downregulating the pi3k/akt pathway. BioMed research international. 2014;2014:546353.PubMedPubMedCentral
18.
go back to reference Zong D, Yin L, Zhong Q, Guo WJ, Xu JH, Jiang N, et al. Znf 488 enhances the invasion and tumorigenesis in nasopharyngeal carcinoma via the wnt signaling pathway involving epithelial mesenchymal transition. Cancer research and treatment : official journal of Korean Cancer Association. 2015. Zong D, Yin L, Zhong Q, Guo WJ, Xu JH, Jiang N, et al. Znf 488 enhances the invasion and tumorigenesis in nasopharyngeal carcinoma via the wnt signaling pathway involving epithelial mesenchymal transition. Cancer research and treatment : official journal of Korean Cancer Association. 2015.
19.
go back to reference Lupo A, Cesaro E, Montano G, Zurlo D, Izzo P, Costanzo P. Krab-zinc finger proteins: a repressor family displaying multiple biological functions. Current genomics. 2013;14:268–78.CrossRefPubMedPubMedCentral Lupo A, Cesaro E, Montano G, Zurlo D, Izzo P, Costanzo P. Krab-zinc finger proteins: a repressor family displaying multiple biological functions. Current genomics. 2013;14:268–78.CrossRefPubMedPubMedCentral
20.
go back to reference Zhang L, Wang X, Lai M. Modulation of epithelial-to-mesenchymal cancerous transition by natural products. Fitoterapia. 2015. Zhang L, Wang X, Lai M. Modulation of epithelial-to-mesenchymal cancerous transition by natural products. Fitoterapia. 2015.
21.
go back to reference De Craene B, Berx G. Regulatory networks defining emt during cancer initiation and progression. Nature reviews Cancer. 2013;13:97–110.CrossRefPubMed De Craene B, Berx G. Regulatory networks defining emt during cancer initiation and progression. Nature reviews Cancer. 2013;13:97–110.CrossRefPubMed
22.
go back to reference Graff JR, Gabrielson E, Fujii H, Baylin SB, Herman JG. Methylation patterns of the e-cadherin 5′ cpg island are unstable and reflect the dynamic, heterogeneous loss of e-cadherin expression during metastatic progression. The Journal of biological chemistry. 2000;275:2727–32.CrossRefPubMed Graff JR, Gabrielson E, Fujii H, Baylin SB, Herman JG. Methylation patterns of the e-cadherin 5′ cpg island are unstable and reflect the dynamic, heterogeneous loss of e-cadherin expression during metastatic progression. The Journal of biological chemistry. 2000;275:2727–32.CrossRefPubMed
23.
go back to reference Lombaerts M, van Wezel T, Philippo K, Dierssen JW, Zimmerman RM, Oosting J, et al. E-cadherin transcriptional downregulation by promoter methylation but not mutation is related to epithelial-to-mesenchymal transition in breast cancer cell lines. Br J Cancer. 2006;94:661–71.PubMedPubMedCentral Lombaerts M, van Wezel T, Philippo K, Dierssen JW, Zimmerman RM, Oosting J, et al. E-cadherin transcriptional downregulation by promoter methylation but not mutation is related to epithelial-to-mesenchymal transition in breast cancer cell lines. Br J Cancer. 2006;94:661–71.PubMedPubMedCentral
24.
go back to reference Izumi H, Yasuniwa Y, Akiyama M, Yamaguchi T, Kuma A, Kitamura N, et al. Forced expression of znf143 restrains cancer cell growth. Cancers (Basel). 2011;3:3909–20.CrossRef Izumi H, Yasuniwa Y, Akiyama M, Yamaguchi T, Kuma A, Kitamura N, et al. Forced expression of znf143 restrains cancer cell growth. Cancers (Basel). 2011;3:3909–20.CrossRef
25.
go back to reference Nieto MA. The ins and outs of the epithelial to mesenchymal transition in health and disease. Annual review of cell and developmental biology. 2011;27:347–76.CrossRefPubMed Nieto MA. The ins and outs of the epithelial to mesenchymal transition in health and disease. Annual review of cell and developmental biology. 2011;27:347–76.CrossRefPubMed
26.
go back to reference Frisch SM. The epithelial cell default-phenotype hypothesis and its implications for cancer. BioEssays : news and reviews in molecular, cellular and developmental biology. 1997;19:705–9.CrossRef Frisch SM. The epithelial cell default-phenotype hypothesis and its implications for cancer. BioEssays : news and reviews in molecular, cellular and developmental biology. 1997;19:705–9.CrossRef
27.
go back to reference Wang SC, Chai DS, Chen CB, Wang ZY, Wang L. Hpip promotes thyroid cancer cell growth, migration and emt through activating pi3k/akt signaling pathway. Biomedicine & pharmacotherapy = Biomedicine & Pharmacotherapy. 2015;75:33–9.CrossRef Wang SC, Chai DS, Chen CB, Wang ZY, Wang L. Hpip promotes thyroid cancer cell growth, migration and emt through activating pi3k/akt signaling pathway. Biomedicine & pharmacotherapy = Biomedicine & Pharmacotherapy. 2015;75:33–9.CrossRef
28.
go back to reference Grille SJ, Bellacosa A, Upson J, Klein-Szanto AJ, van Roy F, Lee-Kwon W, et al. The protein kinase akt induces epithelial mesenchymal transition and promotes enhanced motility and invasiveness of squamous cell carcinoma lines. Cancer Res. 2003;63:2172–8.PubMed Grille SJ, Bellacosa A, Upson J, Klein-Szanto AJ, van Roy F, Lee-Kwon W, et al. The protein kinase akt induces epithelial mesenchymal transition and promotes enhanced motility and invasiveness of squamous cell carcinoma lines. Cancer Res. 2003;63:2172–8.PubMed
Metadata
Title
ZNF143 enhances metastasis of gastric cancer by promoting the process of EMT through PI3K/AKT signaling pathway
Authors
Song Wei
Linjun Wang
Lei Zhang
Bowen Li
Zheng Li
Qun Zhang
Jiwei Wang
Liang Chen
Guangli Sun
Qing Li
Hao Xu
Diancai Zhang
Zekuan Xu
Publication date
01-09-2016
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 9/2016
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-016-5239-z

Other articles of this Issue 9/2016

Tumor Biology 9/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine