J Breast Cancer.  2018 Jun;21(2):103-111. 10.4048/jbc.2018.21.2.103.

Knockdown of Chloride Channel-3 Inhibits Breast Cancer Growth In Vitro and In Vivo

Affiliations
  • 1Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. tangyb@mail.sysu.edu.cn
  • 2Department of Pharmacy, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
  • 3Department of Clinical Pharmacology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.

Abstract

PURPOSE
Chloride channel-3 (ClC-3) is a member of the chloride channel family and plays a critical role in a variety of cellular activities. The aim of the present study is to explore the molecular mechanisms underlying the antitumor effect of silencing ClC-3 in breast cancer.
METHODS
Human breast cancer cell lines MDA-MB-231 and MCF-7 were used in the experiments. Messenger RNA and protein expression were examined by quantitative real-time polymerase chain reaction and western blot analysis. Cell proliferation was measured by the bromodeoxyuridine method, and the cell cycle was evaluated using fluorescence-activated cell sorting. Protein interaction in cells was analyzed by co-immunoprecipitation. Tumor tissues were stained with hematoxylin-eosin and tumor burden was measured using the Metamorph software.
RESULTS
Breast cancer tissues collected from patients showed an increase in ClC-3 expression. Knockdown of ClC-3 inhibited the secretion of insulin-like growth factor (IGF)-1, cell proliferation, and G1/S transition in breast cancer cells. In the mouse xenograft model of human breast carcinoma, tumor growth was significantly slower in animals injected with ClC-3-deficient cells compared with the growth of normal human breast cancer cells. In addition, silencing of ClC-3 attenuated the expression of proliferating cell nuclear antigen, Ki-67, cyclin D1, and cyclin E, as well as the activation of extracellular signal-regulated protein kinases (ERK) 1/2, both in vitro and in vivo.
CONCLUSION
Together, our data suggest that upregulation of ClC-3 by IGF-1 contributes to cell proliferation and tumor growth in breast cancer, and ClC-3 deficiency suppresses cell proliferation and tumor growth via the IGF/IGF receptor/ERK pathway.

Keyword

Breast neoplasms; Cell proliferation; Chloride channel-3; Insulin-like growth factor 1

MeSH Terms

Animals
Blotting, Western
Breast Neoplasms*
Breast*
Bromodeoxyuridine
Cell Cycle
Cell Line
Cell Proliferation
Chloride Channels
Cyclin D1
Cyclin E
Cyclins
Flow Cytometry
Heterografts
Humans
Immunoprecipitation
In Vitro Techniques*
Insulin-Like Growth Factor I
Methods
Mice
Proliferating Cell Nuclear Antigen
Protein Kinases
Real-Time Polymerase Chain Reaction
RNA, Messenger
Tumor Burden
Up-Regulation
Bromodeoxyuridine
Chloride Channels
Cyclin D1
Cyclin E
Cyclins
Insulin-Like Growth Factor I
Proliferating Cell Nuclear Antigen
Protein Kinases
RNA, Messenger

Figure

  • Figure 1 Chloride channel-3 (CIC-3) expression in breast tumor tissues. (A) Chloride channels (ClC-1–7, CFTR) are expressed in human breast cancer tissues from clinical postoperative patients, as indicated by qRT-PCR mRNA analysis. The data are presented as relative fold changes in breast cancer tissues versus normal tissues. (B) Expression of ClC-3 was examined by western blotting, (C) ClC-3 protein is expressed in breast cancer tissues. The result shows a significant increase in ClC-3 expression in breast cancer tissues vs. normal tissues. CFTR=cystic fbrosis transmembrane conductance regulator; qRT-PCR=quantitative real - time polymerase chain reaction; mRNA=messenger RNA; GAPDH=glyceraldehyde 3-phosphate dehydrogenase. *p<0.01 vs. control.

  • Figure 2 Insulin-like growth factor 1 (IGF-1) and chloride channel-3 (CIC-3) expression in human breast cancer cells. (A) Effect of 30–300 ng/mL IGF-1 on the expression of ClC-3 in MDA-MB-231 cells. (B) Effect of 30–300 ng/mL IGF-1 on the expression of ClC-3 in MCF-7 cells (n=5). GAPDH=glyceraldehyde 3-phosphate dehydrogenase. *p<0.05 vs. control; †p<0.01 vs. control.

  • Figure 3 Effects of chloride channel-3 (ClC-3) knockdown on insulin-like growth factor 1 (IGF-1)-dependent cell proliferation. (A) Increase in cell growth induced by 100 ng/mL IGF-1 was determined by counting the number of cells, and was significantly inhibited by incubation with ClC-3 small interfering RNA (siRNA) for 24 hours, but not by transfection with negative siRNA. (B) The effects of ClC-3 siRNA on cell proliferation induced by IGF-1 were further determined by assaying bromodeoxyuridine (BrdU) incorporation. Incubation with ClC-3 siRNA for 24 hours also significantly decreased BrdU incorporation. (C) Quiescent MDA-MB-231cells incubated in 100 ng/mL IGF-1 were transfected with ClC-3 siRNA or negative siRNA control (IGF-1+negative siRNA) plus transfecting agent lipofectamine. The cells were trypsinized, rinsed with phosphate-buffered saline, and treated with 20 µg/mL RNase. DNA was stained with propidium iodide, and 1×106 cells were analyzed by fluorescence-activated cell sorter analysis. Bars represent mean±SD (n=5). Analysis of variance was used for comparison. *p<0.01 vs. control; †p<0.01 vs. IGF-1.

  • Figure 4 Effects of chloride channel-3 (ClC-3) knockdown on the expression of cell cycle-regulatory proteins. (A) Representative western blot images of Ki-67, proliferating cell nuclear antigen (PCNA), cyclins, and p21. (B) Densitometric analysis of the effects of ClC-3 knockdown on expression of Ki-67, PCNA, cyclin D1, cyclin E, and p21 (n=5). GAPDH=glyceraldehyde 3-phosphate dehydrogenase; IGF-1=insulin-like growth factor 1; siRNA=small interfering RNA. *p<0.01 vs. control; †p<0.01 vs. IGF-1.

  • Figure 5 Effect of insulin-like growth factor 1 (IGF-1) induced extracellular regulated protein kinases 1/2 (ERK1/2) activation and the signal-responsive interaction of chloride channel-3 (CIC-3) with insulin-like growth factor receptor (IGF-R). (A) Effects of ClC-3 knockdown on the expression of ERK1/2 and phosphorylated ERK1/2 (pERK1/2). Bars represent mean±SD (n=5). Analysis of variance was employed for comparison. (B) Co-immunoprecipitation assay of ClC-3/IGF-R interaction in MDA-MB-231 cells (n=5). Cells were treated with or without IGF-1 (100 ng/mL). Protein extracts were immunoprecipitated (IP) with anti-IGF-R or anti-ClC-3 antibody, followed by immunoblotting (IB) with anti-IGF-R or anti-ClC-3. siRNA=small interfering RNA. *p<0.01 vs. control; †p<0.01 vs. IGF-1+Neg ClC-3.

  • Figure 6 Effects of chloride channel-3 (ClC-3) knockdown on the growth of human breast tumors in vivo. (A) Representative images of tumors on day 20 from the control and ClC-3 short hairpin RNA (shRNA) groups. (B) Changes in tumor volume were calculated postinoculation every 4 days. Dots represent mean±SD. Seven mice were included in each group. Analysis of variance was employed for comparison. (C) Immunohistochemistry was performed to assess the expression of Ki-67 in tumor tissues isolated from control and ClC-3 shRNA mice, tumor tissues were cut and stained with hematoxylin-eosin, and observed with phase contrast microscope (×200). (D) Western blot analysis was performed to measure the expression of Ki-67, proliferating cell nuclear antigen (PCNA), cyclin D1, cyclin E, and phosphorylated extracellular regulated protein kinases1/2 (pERK1/2); glyceraldehyde 3-phosphate dehydrogenase (GADPH) was used as loading control (n=7). *p<0.01 vs. control.


Reference

1. Singh E, Joffe M, Cubasch H, Ruff P, Norris SA, Pisa PT. Breast cancer trends differ by ethnicity: a report from the South African National Cancer Registry (1994-2009). Eur J Public Health. 2017; 27:173–178.
Article
2. Kreiter E, Richardson A, Potter J, Yasui Y. Breast cancer: trends in international incidence in men and women. Br J Cancer. 2014; 110:1891–1897.
Article
3. Yang H, Ma L, Wang Y, Zuo W, Li B, Yang Y, et al. Activation of ClC-3 chloride channel by 17beta-estradiol relies on the estrogen receptor alpha expression in breast cancer. J Cell Physiol. 2018; 233:1071–1081.
Article
4. Xu B, Jin X, Min L, Li Q, Deng L, Wu H, et al. Chloride channel-3 promotes tumor metastasis by regulating membrane ruffling and is associated with poor survival. Oncotarget. 2015; 6:2434–2450.
Article
5. Lemonnier L, Lazarenko R, Shuba Y, Thebault S, Roudbaraki M, Lepage G, et al. Alterations in the regulatory volume decrease (RVD) and swelling-activated Cl- current associated with neuroendocrine differentiation of prostate cancer epithelial cells. Endocr Relat Cancer. 2005; 12:335–349.
Article
6. Mao J, Chen L, Xu B, Wang L, Li H, Guo J, et al. Suppression of ClC-3 channel expression reduces migration of nasopharyngeal carcinoma cells. Biochem Pharmacol. 2008; 75:1706–1716.
Article
7. Du S, Yang L. ClC-3 chloride channel modulates the proliferation and migration of osteosarcoma cells via AKT/GSK3beta signaling pathway. Int J Clin Exp Pathol. 2015; 8:1622–1630.
8. Kasinathan RS, Föller M, Lang C, Koka S, Lang F, Huber SM. Oxidation induces ClC-3-dependent anion channels in human leukaemia cells. FEBS Lett. 2007; 581:5407–5412.
Article
9. Cuddapah VA, Sontheimer H. Molecular interaction and functional regulation of ClC-3 by Ca2+/calmodulin-dependent protein kinase II (CaMKII) in human malignant glioma. J Biol Chem. 2010; 285:11188–11196.
Article
10. Hong S, Bi M, Wang L, Kang Z, Ling L, Zhao C. CLC-3 channels in cancer (review). Oncol Rep. 2015; 33:507–514.
Article
11. Wang L, Ma W, Zhu L, Ye D, Li Y, Liu S, et al. ClC-3 is a candidate of the channel proteins mediating acid-activated chloride currents in nasopharyngeal carcinoma cells. Am J Physiol Cell Physiol. 2012; 303:C14–C23.
Article
12. Tang YB, Liu YJ, Zhou JG, Wang GL, Qiu QY, Guan YY. Silence of ClC-3 chloride channel inhibits cell proliferation and the cell cycle via G/S phase arrest in rat basilar arterial smooth muscle cells. Cell Prolif. 2008; 41:775–785.
Article
13. Mao J, Chen L, Xu B, Wang L, Wang W, Li M, et al. Volume-activated chloride channels contribute to cell-cycle-dependent regulation of HeLa cell migration. Biochem Pharmacol. 2009; 77:159–168.
Article
14. Huang YY, Huang XQ, Zhao LY, Sun FY, Chen WL, Du JY, et al. ClC-3 deficiency protects preadipocytes against apoptosis induced by palmitate in vitro and in type 2 diabetes mice. Apoptosis. 2014; 19:1559–1570.
Article
15. Huang EW, Xue SJ, Zhang Z, Zhou JG, Guan YY, Tang YB. Vinpocetine inhibits breast cancer cells growth in vitro and in vivo. Apoptosis. 2012; 17:1120–1130.
Article
16. Wang M, Tang YB, Ma MM, Chen JH, Hu CP, Zhao SP, et al. TRPC3 channel confers cerebrovascular remodelling during hypertension via transactivation of EGF receptor signalling. Cardiovasc Res. 2016; 109:34–43.
Article
17. Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009; 9:153–166.
Article
18. Braun-Dullaeus RC, Mann MJ, Dzau VJ. Cell cycle progression: new therapeutic target for vascular proliferative disease. Circulation. 1998; 98:82–89.
19. Abbas T, Dutta A. p21 in cancer: intricate networks and multiple activities. Nat Rev Cancer. 2009; 9:400–414.
Article
20. Dhillon AS, Hagan S, Rath O, Kolch W. MAP kinase signalling pathways in cancer. Oncogene. 2007; 26:3279–3290.
Article
21. Roberts PJ, Der CJ. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 2007; 26:3291–3310.
Article
22. Saini KS, Loi S, de Azambuja E, Metzger-Filho O, Saini ML, Ignatiadis M, et al. Targeting the PI3K/AKT/mTOR and Raf/MEK/ERK pathways in the treatment of breast cancer. Cancer Treat Rev. 2013; 39:935–946.
Article
23. Mester J, Redeuilh G. Proliferation of breast cancer cells: regulation, mediators, targets for therapy. Anticancer Agents Med Chem. 2008; 8:872–885.
Article
Full Text Links
  • JBC
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr