Skip to main content
Top
Published in: BMC Cancer 1/2018

Open Access 01-12-2018 | Research article

BKCa participates in E2 inducing endometrial adenocarcinoma by activating MEK/ERK pathway

Authors: Fenfen Wang, Qin Chen, Genping Huang, Xuedong Guo, Na Li, Yang Li, Baohua Li

Published in: BMC Cancer | Issue 1/2018

Login to get access

Abstract

Background

The large-conductance, voltage-gated, calcium (Ca (2+))-activated potassium channel (BKCa) plays an important role in regulating Ca (2+) signaling and cell physiological function, and is aberrantly expressed in some types of cancers. The present study focuses on identifying the oncogenic potential and clinical significance of BKCa in endometrial adenocarcinoma, as well as exploring the mechanistic relevance by 17β -estradiol (E2) inducing aberrant activation of MEK1/2 and ERK1/2 via BKCa.

Methods

The expression of BKCa, ERK1/2 and p-ERK1/2 were examined by immunohistochemical staining in 263 cases, including 185 primary types I endometrial cancer tissues, 38 atypical endometrial hyperplasia tissues and 40 normal endometrium tissues. Cell growth, cycle, apoptosis rate, migration and invasion was separately tested in Ishikawa cells using siRNA-BKCa and/or E2 treatment, as well as the expression of these interested proteins by western blot analysis.

Results

We showed that expression of BKCa is significantly elevated in 185 types I endometrial adenocarcinoma tissues compared to those of the normal endometrium and atypical endometrial hyperplasia tissues. Furthermore, in vitro observations revealed that down-regulation of BKCa expression inhibited cell growth by both enhancing apoptosis and blocking G1/S transition, suppressed cell migration and invasion in Ishakiwa cells, and decreased the expression of p-MEK1/2 and p-ERK1/2. Additionally, RNAi-mediated knockdown of BKCa attenuated the increased cellular growth and invasion, as well as the elevated expression of p-MEK1/2 and p-ERK1/2 proteins, induced by E2 stimulation. More importantly, the aberrant expression of BKCa and p-ERK1/2 were closely related with poor prognostic factors in type I endometrial cancer, and up-regulated expression of p-ERK1/2 was significantly associated with shorter disease-free survival (DFS) and overall survival (OS) and was an independent prognostic factor in type I endometrial cancer patients.

Conclusion

Our results demonstrated that BKCa and the key downstream effectors p-ERK1/2 could be involved in important signaling pathways in initiation and development of endometrial adenocarcinoma and may provide a new therapeutic approach for women with endometrial cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Morice P, Leary A, Creutzberg C, Abu-Rustum N, Darai E. Endometrial cancer. Lancet. 2016;387:1094–108.CrossRef Morice P, Leary A, Creutzberg C, Abu-Rustum N, Darai E. Endometrial cancer. Lancet. 2016;387:1094–108.CrossRef
2.
go back to reference Siufi DF, Siufi Neto J, Abrao MS, Favero G. Lymphadenectomy in early stage endometrial cancer: a critical review of the current literature. Tumori. 2014;100:477–85.CrossRef Siufi DF, Siufi Neto J, Abrao MS, Favero G. Lymphadenectomy in early stage endometrial cancer: a critical review of the current literature. Tumori. 2014;100:477–85.CrossRef
3.
go back to reference Tong JS, Zhang QH, Wang ZB, Li S, Yang CR, Fu XQ, et al. ER-alpha36, a novel variant of ER-alpha, mediates estrogen-stimulated proliferation of endometrial carcinoma cells via the PKCdelta/ERK pathway. PLoS One. 2010;5:e15408.CrossRef Tong JS, Zhang QH, Wang ZB, Li S, Yang CR, Fu XQ, et al. ER-alpha36, a novel variant of ER-alpha, mediates estrogen-stimulated proliferation of endometrial carcinoma cells via the PKCdelta/ERK pathway. PLoS One. 2010;5:e15408.CrossRef
4.
go back to reference Koo KH, Jeong WJ, Cho YH, Park JC, Min do S, Choi KY. K-Ras stabilization by estrogen via PKCdelta is involved in endometrial tumorigenesis. Oncotarget. 2015;6:21328–40.PubMedPubMedCentral Koo KH, Jeong WJ, Cho YH, Park JC, Min do S, Choi KY. K-Ras stabilization by estrogen via PKCdelta is involved in endometrial tumorigenesis. Oncotarget. 2015;6:21328–40.PubMedPubMedCentral
5.
go back to reference Su T, Qu JJ, Wang K, Li BL, Zhao D, Zhu YP, et al. Cross-talk between p21-activated kinase 4 and ERalpha signaling triggers endometrial cancer cell proliferation. Oncotarget. 2017;8:68083–94.PubMedPubMedCentral Su T, Qu JJ, Wang K, Li BL, Zhao D, Zhu YP, et al. Cross-talk between p21-activated kinase 4 and ERalpha signaling triggers endometrial cancer cell proliferation. Oncotarget. 2017;8:68083–94.PubMedPubMedCentral
6.
go back to reference Hou S, Heinemann SH, Hoshi T. Modulation of BKCa channel gating by endogenous signaling molecules. Physiology (Bethesda). 2009;24:26–35. Hou S, Heinemann SH, Hoshi T. Modulation of BKCa channel gating by endogenous signaling molecules. Physiology (Bethesda). 2009;24:26–35.
7.
go back to reference Lu R, Alioua A, Kumar Y, Eghbali M, Stefani E, Toro L. MaxiK channel partners: physiological impact. J Physiol. 2006;570:65–72.CrossRef Lu R, Alioua A, Kumar Y, Eghbali M, Stefani E, Toro L. MaxiK channel partners: physiological impact. J Physiol. 2006;570:65–72.CrossRef
8.
go back to reference Hu XQ, Dasgupta C, Chen M, Xiao D, Huang X, Han L, et al. Pregnancy reprograms large-conductance Ca2+-activated K+ channel in uterine arteries: roles of ten-eleven translocation Methylcytosine dioxygenase 1-mediated active demethylation. Hypertension. 2017;69(6):1181-191.CrossRef Hu XQ, Dasgupta C, Chen M, Xiao D, Huang X, Han L, et al. Pregnancy reprograms large-conductance Ca2+-activated K+ channel in uterine arteries: roles of ten-eleven translocation Methylcytosine dioxygenase 1-mediated active demethylation. Hypertension. 2017;69(6):1181-191.CrossRef
9.
go back to reference Bloch M, Ousingsawat J, Simon R, Schraml P, Gasser TC, Mihatsch MJ, et al. KCNMA1 gene amplification promotes tumor cell proliferation in human prostate cancer. Oncogene. 2007;26:2525–34.CrossRef Bloch M, Ousingsawat J, Simon R, Schraml P, Gasser TC, Mihatsch MJ, et al. KCNMA1 gene amplification promotes tumor cell proliferation in human prostate cancer. Oncogene. 2007;26:2525–34.CrossRef
10.
go back to reference Cambien B, Rezzonico R, Vitale S, Rouzaire-Dubois B, Dubois JM, Barthel R, et al. Silencing of hSlo potassium channels in human osteosarcoma cells promotes tumorigenesis. Int J Cancer. 2008;123:365–71.CrossRef Cambien B, Rezzonico R, Vitale S, Rouzaire-Dubois B, Dubois JM, Barthel R, et al. Silencing of hSlo potassium channels in human osteosarcoma cells promotes tumorigenesis. Int J Cancer. 2008;123:365–71.CrossRef
11.
go back to reference Oeggerli M, Tian Y, Ruiz C, Wijker B, Sauter G, Obermann E, et al. Role of KCNMA1 in breast cancer. PLoS One. 2012;7:e41664.CrossRef Oeggerli M, Tian Y, Ruiz C, Wijker B, Sauter G, Obermann E, et al. Role of KCNMA1 in breast cancer. PLoS One. 2012;7:e41664.CrossRef
12.
go back to reference Ma G, Liu H, Hua Q, Wang M, Du M, Lin Y, et al. KCNMA1 cooperating with PTK2 is a novel tumor suppressor in gastric cancer and is associated with disease outcome. Mol Cancer. 2017;16:46.CrossRef Ma G, Liu H, Hua Q, Wang M, Du M, Lin Y, et al. KCNMA1 cooperating with PTK2 is a novel tumor suppressor in gastric cancer and is associated with disease outcome. Mol Cancer. 2017;16:46.CrossRef
13.
go back to reference Shi JH, Jin L, Leng JH, Lang JH. Expression of potassium channels in uterine smooth muscle cells from patients with Adenomyosis. Chin Med J. 2016;129:200–5.CrossRef Shi JH, Jin L, Leng JH, Lang JH. Expression of potassium channels in uterine smooth muscle cells from patients with Adenomyosis. Chin Med J. 2016;129:200–5.CrossRef
14.
go back to reference Lorca RA, Prabagaran M, England SK. Functional insights into modulation of BKCa channel activity to alter myometrial contractility. Front Physiol. 2014;5:289.CrossRef Lorca RA, Prabagaran M, England SK. Functional insights into modulation of BKCa channel activity to alter myometrial contractility. Front Physiol. 2014;5:289.CrossRef
15.
go back to reference Gao L, Cong B, Zhang L, Ni X. Expression of the calcium-activated potassium channel in upper and lower segment human myometrium during pregnancy and parturition. Reprod Biol Endocrinol. 2009;7:27.CrossRef Gao L, Cong B, Zhang L, Ni X. Expression of the calcium-activated potassium channel in upper and lower segment human myometrium during pregnancy and parturition. Reprod Biol Endocrinol. 2009;7:27.CrossRef
16.
go back to reference Li N, Liu L, Li G, Xia M, Du C, Zheng Z. The role of BKCa in endometrial cancer HEC-1-B cell proliferation and migration. Gene. 2018;655:42–7.CrossRef Li N, Liu L, Li G, Xia M, Du C, Zheng Z. The role of BKCa in endometrial cancer HEC-1-B cell proliferation and migration. Gene. 2018;655:42–7.CrossRef
17.
go back to reference Wang F, Li Y, Zhou J, Xu J, Peng C, Ye F, et al. miR-375 is down-regulated in squamous cervical cancer and inhibits cell migration and invasion via targeting transcription factor SP1. Am J Pathol. 2011;179:2580–8.CrossRef Wang F, Li Y, Zhou J, Xu J, Peng C, Ye F, et al. miR-375 is down-regulated in squamous cervical cancer and inhibits cell migration and invasion via targeting transcription factor SP1. Am J Pathol. 2011;179:2580–8.CrossRef
18.
go back to reference Li B, Shi H, Wang F, Hong D, Lv W, Xie X, et al. Expression of E-, P- and N-cadherin and its clinical significance in cervical squamous cell carcinoma and precancerous lesions. PLoS One. 2016;11:e0155910.CrossRef Li B, Shi H, Wang F, Hong D, Lv W, Xie X, et al. Expression of E-, P- and N-cadherin and its clinical significance in cervical squamous cell carcinoma and precancerous lesions. PLoS One. 2016;11:e0155910.CrossRef
19.
go back to reference Du C, Zheng Z, Li D, Chen L, Li N, Yi X, et al. BKCa promotes growth and metastasis of prostate cancer through facilitating the coupling between alphavbeta3 integrin and FAK. Oncotarget. 2016;7:40174–88.PubMedPubMedCentral Du C, Zheng Z, Li D, Chen L, Li N, Yi X, et al. BKCa promotes growth and metastasis of prostate cancer through facilitating the coupling between alphavbeta3 integrin and FAK. Oncotarget. 2016;7:40174–88.PubMedPubMedCentral
20.
go back to reference Schickling BM, England SK, Aykin-Burns N, Norian LA, Leslie KK, Frieden-Korovkina VP. BKCa channel inhibitor modulates the tumorigenic ability of hormone-independent breast cancer cells via the Wnt pathway. Oncol Rep. 2015;33:533–8.CrossRef Schickling BM, England SK, Aykin-Burns N, Norian LA, Leslie KK, Frieden-Korovkina VP. BKCa channel inhibitor modulates the tumorigenic ability of hormone-independent breast cancer cells via the Wnt pathway. Oncol Rep. 2015;33:533–8.CrossRef
21.
go back to reference Khaitan D, Sankpal UT, Weksler B, Meister EA, Romero IA, Couraud PO, et al. Role of KCNMA1 gene in breast cancer invasion and metastasis to brain. BMC Cancer. 2009;9:258.CrossRef Khaitan D, Sankpal UT, Weksler B, Meister EA, Romero IA, Couraud PO, et al. Role of KCNMA1 gene in breast cancer invasion and metastasis to brain. BMC Cancer. 2009;9:258.CrossRef
22.
go back to reference Du C, Chen L, Zhang H, Wang Z, Liu W, Xie X, et al. Caveolin-1 limits the contribution of BKCa channel to MCF-7 breast cancer cell proliferation and invasion. Int J Mol Sci. 2014;15:20706–22.CrossRef Du C, Chen L, Zhang H, Wang Z, Liu W, Xie X, et al. Caveolin-1 limits the contribution of BKCa channel to MCF-7 breast cancer cell proliferation and invasion. Int J Mol Sci. 2014;15:20706–22.CrossRef
23.
go back to reference Mound A, Rodat-Despoix L, Bougarn S, Ouadid-Ahidouch H, Matifat F. Molecular interaction and functional coupling between type 3 inositol 1,4,5-trisphosphate receptor and BKCa channel stimulate breast cancer cell proliferation. Eur J Cancer. 2013;49:3738–51.CrossRef Mound A, Rodat-Despoix L, Bougarn S, Ouadid-Ahidouch H, Matifat F. Molecular interaction and functional coupling between type 3 inositol 1,4,5-trisphosphate receptor and BKCa channel stimulate breast cancer cell proliferation. Eur J Cancer. 2013;49:3738–51.CrossRef
24.
go back to reference Eritja N, Yeramian A, Chen BJ, Llobet-Navas D, Ortega E, Colas E, et al. Endometrial carcinoma: specific targeted pathways. Adv Exp Med Biol. 2017;943:149–207.CrossRef Eritja N, Yeramian A, Chen BJ, Llobet-Navas D, Ortega E, Colas E, et al. Endometrial carcinoma: specific targeted pathways. Adv Exp Med Biol. 2017;943:149–207.CrossRef
25.
go back to reference He YY, Cai B, Yang YX, Liu XL, Wan XP. Estrogenic G protein-coupled receptor 30 signaling is involved in regulation of endometrial carcinoma by promoting proliferation, invasion potential, and interleukin-6 secretion via the MEK/ERK mitogen-activated protein kinase pathway. Cancer Sci. 2009;100:1051–61.CrossRef He YY, Cai B, Yang YX, Liu XL, Wan XP. Estrogenic G protein-coupled receptor 30 signaling is involved in regulation of endometrial carcinoma by promoting proliferation, invasion potential, and interleukin-6 secretion via the MEK/ERK mitogen-activated protein kinase pathway. Cancer Sci. 2009;100:1051–61.CrossRef
26.
go back to reference Li Y, Jia Y, Che Q, Zhou Q, Wang K, Wan XP. AMF/PGI-mediated tumorigenesis through MAPK-ERK signaling in endometrial carcinoma. Oncotarget. 2015;6:26373–87.PubMedPubMedCentral Li Y, Jia Y, Che Q, Zhou Q, Wang K, Wan XP. AMF/PGI-mediated tumorigenesis through MAPK-ERK signaling in endometrial carcinoma. Oncotarget. 2015;6:26373–87.PubMedPubMedCentral
27.
go back to reference Marampon F, Gravina GL, Popov VM, Scarsella L, Festuccia C, La Verghetta ME, et al. Close correlation between MEK/ERK and Aurora-B signaling pathways in sustaining tumorigenic potential and radioresistance of gynecological cancer cell lines. Int J Oncol. 2014;44:285–94.CrossRef Marampon F, Gravina GL, Popov VM, Scarsella L, Festuccia C, La Verghetta ME, et al. Close correlation between MEK/ERK and Aurora-B signaling pathways in sustaining tumorigenic potential and radioresistance of gynecological cancer cell lines. Int J Oncol. 2014;44:285–94.CrossRef
28.
go back to reference Coiret G, Borowiec AS, Mariot P, Ouadid-Ahidouch H, Matifat F. The antiestrogen tamoxifen activates BK channels and stimulates proliferation of MCF-7 breast cancer cells. Mol Pharmacol. 2007;71:843–51.CrossRef Coiret G, Borowiec AS, Mariot P, Ouadid-Ahidouch H, Matifat F. The antiestrogen tamoxifen activates BK channels and stimulates proliferation of MCF-7 breast cancer cells. Mol Pharmacol. 2007;71:843–51.CrossRef
29.
go back to reference Ambroisine ML, Favre J, Oliviero P, Rodriguez C, Gao J, Thuillez C, et al. Aldosterone-induced coronary dysfunction in transgenic mice involves the calcium-activated potassium (BKCa) channels of vascular smooth muscle cells. Circulation. 2007;116:2435–43.CrossRef Ambroisine ML, Favre J, Oliviero P, Rodriguez C, Gao J, Thuillez C, et al. Aldosterone-induced coronary dysfunction in transgenic mice involves the calcium-activated potassium (BKCa) channels of vascular smooth muscle cells. Circulation. 2007;116:2435–43.CrossRef
30.
go back to reference Kundu P, Alioua A, Stefani E, Toro L. Regulation of mouse Slo gene expression: multiple promoters, transcription start sites, and genomic action of estrogen. J Biol Chem. 2007;282:27478–92.CrossRef Kundu P, Alioua A, Stefani E, Toro L. Regulation of mouse Slo gene expression: multiple promoters, transcription start sites, and genomic action of estrogen. J Biol Chem. 2007;282:27478–92.CrossRef
31.
go back to reference Jamali K, Naylor BR, Kelly MJ, Ronnekleiv OK. Effect of 17beta-estradiol on mRNA expression of large- conductance, voltage-dependent, and calcium-activated potassium channel alpha and beta subunits in Guinea pig. Endocrine. 2003;20:227–37.CrossRef Jamali K, Naylor BR, Kelly MJ, Ronnekleiv OK. Effect of 17beta-estradiol on mRNA expression of large- conductance, voltage-dependent, and calcium-activated potassium channel alpha and beta subunits in Guinea pig. Endocrine. 2003;20:227–37.CrossRef
32.
go back to reference Nishimura I, Ui-Tei K, Saigo K, Ishii H, Sakuma Y, Kato M. 17beta-estradiol at physiological concentrations augments Ca(2+) -activated K+ currents via estrogen receptor beta in the gonadotropin-releasing hormone neuronal cell line GT1-7. Endocrinology. 2008;149:774–82.CrossRef Nishimura I, Ui-Tei K, Saigo K, Ishii H, Sakuma Y, Kato M. 17beta-estradiol at physiological concentrations augments Ca(2+) -activated K+ currents via estrogen receptor beta in the gonadotropin-releasing hormone neuronal cell line GT1-7. Endocrinology. 2008;149:774–82.CrossRef
33.
go back to reference Korovkina VP, Brainard AM, Ismail P, Schmidt TJ, England SK. Estradiol binding to maxi-K channels induces their down-regulation via proteasomal degradation. J Biol Chem. 2004;279:1217–23.CrossRef Korovkina VP, Brainard AM, Ismail P, Schmidt TJ, England SK. Estradiol binding to maxi-K channels induces their down-regulation via proteasomal degradation. J Biol Chem. 2004;279:1217–23.CrossRef
34.
go back to reference Mizumoto Y, Kyo S, Mori N, Sakaguchi J, Ohno S, Maida Y, et al. Activation of ERK1/2 occurs independently of KRAS or BRAF status in endometrial cancer and is associated with favorable prognosis. Cancer Sci. 2007;98:652–8.CrossRef Mizumoto Y, Kyo S, Mori N, Sakaguchi J, Ohno S, Maida Y, et al. Activation of ERK1/2 occurs independently of KRAS or BRAF status in endometrial cancer and is associated with favorable prognosis. Cancer Sci. 2007;98:652–8.CrossRef
35.
go back to reference Zhao Y, Yang Y, Trovik J, Sun K, Zhou L, Jiang P, et al. A novel wnt regulatory axis in endometrioid endometrial cancer. Cancer Res. 2014;74:5103–17.CrossRef Zhao Y, Yang Y, Trovik J, Sun K, Zhou L, Jiang P, et al. A novel wnt regulatory axis in endometrioid endometrial cancer. Cancer Res. 2014;74:5103–17.CrossRef
36.
go back to reference Mhawech-Fauceglia P, Kesterson J, Wang D, Akers S, DuPont NC, Clark K, et al. Expression and clinical significance of the transforming growth factor-beta signalling pathway in endometrial cancer. Histopathology. 2011;59:63–72.CrossRef Mhawech-Fauceglia P, Kesterson J, Wang D, Akers S, DuPont NC, Clark K, et al. Expression and clinical significance of the transforming growth factor-beta signalling pathway in endometrial cancer. Histopathology. 2011;59:63–72.CrossRef
Metadata
Title
BKCa participates in E2 inducing endometrial adenocarcinoma by activating MEK/ERK pathway
Authors
Fenfen Wang
Qin Chen
Genping Huang
Xuedong Guo
Na Li
Yang Li
Baohua Li
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2018
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-5027-9

Other articles of this Issue 1/2018

BMC Cancer 1/2018 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