Skip to main content
Top
Published in: BMC Cancer 1/2016

Open Access 01-12-2016 | Research article

Overexpression of KCNJ3 gene splice variants affects vital parameters of the malignant breast cancer cell line MCF-7 in an opposing manner

Authors: S. Rezania, S. Kammerer, C. Li, B. Steinecker-Frohnwieser, A. Gorischek, T. T. J. DeVaney, S. Verheyen, C. A. Passegger, N. Ghaffari Tabrizi-Wizsy, H. Hackl, D. Platzer, A. H. Zarnani, E. Malle, S. W. Jahn, T. Bauernhofer, W. Schreibmayer

Published in: BMC Cancer | Issue 1/2016

Login to get access

Abstract

Background

Overexpression the KCNJ3, a gene that encodes subunit 1 of G-protein activated inwardly rectifying K+ channel (GIRK1) in the primary tumor has been found to be associated with reduced survival times and increased lymph node metastasis in breast cancer patients.

Methods

In order to survey possible tumorigenic properties of GIRK1 overexpression, a range of malignant mammary epithelial cells, based on the MCF-7 cell line that permanently overexpress different splice variants of the KCNJ3 gene (GIRK1a, GIRK1c, GIRK1d and as a control, eYFP) were produced. Subsequently, selected cardinal neoplasia associated cellular parameters were assessed and compared.

Results

Adhesion to fibronectin coated surface as well as cell proliferation remained unaffected. Other vital parameters intimately linked to malignancy, i.e. wound healing, chemoinvasion, cellular velocities / motilities and angiogenesis were massively affected by GIRK1 overexpression. Overexpression of different GIRK1 splice variants exerted differential actions. While GIRK1a and GIRK1c overexpression reinforced the affected parameters towards malignancy, overexpression of GIRK1d resulted in the opposite. Single channel recording using the patch clamp technique revealed functional GIRK channels in the plasma membrane of MCF-7 cells albeit at very low frequency.

Discussion

We conclude that GIRK1d acts as a dominant negative constituent of functional GIRK complexes present in the plasma membrane of MCF-7 cells, while overexpression of GIRK1a and GIRK1c augmented their activity. The core component responsible for the cancerogenic action of GIRK1 is apparently presented by a segment comprising aminoacids 235–402, that is present exclusively in GIRK1a and GIRK1c, but not GIRK1d (positions according to GIRK1a primary structure).

Conclusions

The current study provides insight into the cellular and molecular consequences of KCNJ3 overexpression in breast cancer cells and the mechanism upon clinical outcome in patients suffering from breast cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Luescher C, Slesinger PA. Emerging roles for G protein-gated inwardly rectifying potassium (GIRK) channels in health and disease. Nat Rev Neurosci. 2010;11:301–15.CrossRef Luescher C, Slesinger PA. Emerging roles for G protein-gated inwardly rectifying potassium (GIRK) channels in health and disease. Nat Rev Neurosci. 2010;11:301–15.CrossRef
2.
go back to reference Iwanir S, Reuveny E. Adrenaline-induced hyperpolarization of mouse pancreatic islet cells is mediated by G protein-gated inwardly rectifying potassium (GIRK) channels. Pflugers Arch - Eur J Physiol. 2008;456:1097–108.CrossRef Iwanir S, Reuveny E. Adrenaline-induced hyperpolarization of mouse pancreatic islet cells is mediated by G protein-gated inwardly rectifying potassium (GIRK) channels. Pflugers Arch - Eur J Physiol. 2008;456:1097–108.CrossRef
3.
go back to reference Smith PA, Sellers LA, Humphrey PPA. Somatostatin activates two types of inwardly rectifying K+ channels in MIN-6 cells. J Physiol Lond. 2001;532:127–42.CrossRefPubMedPubMedCentral Smith PA, Sellers LA, Humphrey PPA. Somatostatin activates two types of inwardly rectifying K+ channels in MIN-6 cells. J Physiol Lond. 2001;532:127–42.CrossRefPubMedPubMedCentral
4.
go back to reference Shankar H, et al. G-protein-gated inwardly rectifying potassium channels regulate ADP-induced cPLA(2) activity in platelets through Src family kinases. Blood. 2006;108:3027–34.CrossRefPubMedPubMedCentral Shankar H, et al. G-protein-gated inwardly rectifying potassium channels regulate ADP-induced cPLA(2) activity in platelets through Src family kinases. Blood. 2006;108:3027–34.CrossRefPubMedPubMedCentral
5.
go back to reference Shankar H, et al. Role of G protein-gated inwardly rectifying potassium channels in P2Y(12) receptor-mediated platelet functional responses. Blood. 2004;104:1335–43.CrossRefPubMed Shankar H, et al. Role of G protein-gated inwardly rectifying potassium channels in P2Y(12) receptor-mediated platelet functional responses. Blood. 2004;104:1335–43.CrossRefPubMed
8.
go back to reference Stringer BK, Cooper AG, Shepard SB. Overexpression of the G-protein inwardly rectifying potassium channel 1 (GIRK1) in primary breast carcinomas correlates with axillary lymph node metastasis. Can Res. 2001;61:582–8. Stringer BK, Cooper AG, Shepard SB. Overexpression of the G-protein inwardly rectifying potassium channel 1 (GIRK1) in primary breast carcinomas correlates with axillary lymph node metastasis. Can Res. 2001;61:582–8.
9.
go back to reference Brevet M, et al. Expression of K+ channels in normal and cancerous human breast. Histol Histopathol. 2008;23:965–72.PubMed Brevet M, et al. Expression of K+ channels in normal and cancerous human breast. Histol Histopathol. 2008;23:965–72.PubMed
10.
go back to reference Kammerer S, et al. GIRK1 overexpression correlates with ER positive breast cancer subtypes and is associated with poor prognosis. Ann Oncol. 2015;26(Supplement 3):iii15–24. Kammerer S, et al. GIRK1 overexpression correlates with ER positive breast cancer subtypes and is associated with poor prognosis. Ann Oncol. 2015;26(Supplement 3):iii15–24.
11.
go back to reference Plummer HK, Yu Q, Cakir Y, Schuller HM. Expression of inwardly rectifying potassium channels (GIRKs) and beta-adrenergic regulation of breast cancer cell lines. BMC Cancer. 2004;4:93.CrossRefPubMedPubMedCentral Plummer HK, Yu Q, Cakir Y, Schuller HM. Expression of inwardly rectifying potassium channels (GIRKs) and beta-adrenergic regulation of breast cancer cell lines. BMC Cancer. 2004;4:93.CrossRefPubMedPubMedCentral
12.
go back to reference Wagner V, et al. Cloning and characterisation of GIRK1 variants resulting from alternative RNA editing of the KCNJ3 gene transcript in a human breast cancer cell line. J Cell Biochem. 2010;110:598–608.CrossRefPubMed Wagner V, et al. Cloning and characterisation of GIRK1 variants resulting from alternative RNA editing of the KCNJ3 gene transcript in a human breast cancer cell line. J Cell Biochem. 2010;110:598–608.CrossRefPubMed
13.
14.
go back to reference Glebov OO, Nichols BJ. Lipid raft proteins have a random distribution during localized activation of the T-cell receptor. Nat Cell Biol. 2004;6:238–43.CrossRefPubMed Glebov OO, Nichols BJ. Lipid raft proteins have a random distribution during localized activation of the T-cell receptor. Nat Cell Biol. 2004;6:238–43.CrossRefPubMed
15.
go back to reference Ward TH, Brandizzi F. Dynamics of proteins in Golgi membranes: comparisons between mammalian and plant cells highlighted by photobleaching techniques. Cell Mol Life Sci. 2004;61:172–85.CrossRefPubMed Ward TH, Brandizzi F. Dynamics of proteins in Golgi membranes: comparisons between mammalian and plant cells highlighted by photobleaching techniques. Cell Mol Life Sci. 2004;61:172–85.CrossRefPubMed
17.
go back to reference Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9:671–5.CrossRefPubMed Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9:671–5.CrossRefPubMed
18.
go back to reference Deryugina EI, Quigley JP. Chapter 2. Chick embryo chorioallantoic membrane models to quantify angiogenesis induced by inflammatory and tumor cells or purified effector molecules. Methods Enzymol. 2008;444:21–41.CrossRefPubMedPubMedCentral Deryugina EI, Quigley JP. Chapter 2. Chick embryo chorioallantoic membrane models to quantify angiogenesis induced by inflammatory and tumor cells or purified effector molecules. Methods Enzymol. 2008;444:21–41.CrossRefPubMedPubMedCentral
19.
go back to reference Ribatti D, Nico B, Vacca A, Presta M. The gelatin sponge-chorioallantoic membrane assay. Nat Protoc. 2006;1:85–91.CrossRefPubMed Ribatti D, Nico B, Vacca A, Presta M. The gelatin sponge-chorioallantoic membrane assay. Nat Protoc. 2006;1:85–91.CrossRefPubMed
20.
go back to reference Prevarskaya N, Skryma R, Shuba Y. Ion channels and the hallmarks of cancer. Trends Mol Med. 2010;16:107–21.CrossRefPubMed Prevarskaya N, Skryma R, Shuba Y. Ion channels and the hallmarks of cancer. Trends Mol Med. 2010;16:107–21.CrossRefPubMed
21.
22.
go back to reference Arwert EN, Hoste E, Watt FM. Epithelial stem cells, wound healing and cancer. Nat Rev Cancer. 2012;12:170–80.CrossRefPubMed Arwert EN, Hoste E, Watt FM. Epithelial stem cells, wound healing and cancer. Nat Rev Cancer. 2012;12:170–80.CrossRefPubMed
23.
go back to reference Liang CC, Park AY, Guan JL. In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc. 2007;2:329–33.CrossRefPubMed Liang CC, Park AY, Guan JL. In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc. 2007;2:329–33.CrossRefPubMed
24.
go back to reference Falasca M, Raimondi C, Maffucci T. Boyden Chamber. In: Wells CM, Parsons M, editors. Cell migration: developmental methods and protocols, vol. 769. 2nd ed. 2011. p. 87–95.CrossRef Falasca M, Raimondi C, Maffucci T. Boyden Chamber. In: Wells CM, Parsons M, editors. Cell migration: developmental methods and protocols, vol. 769. 2nd ed. 2011. p. 87–95.CrossRef
25.
go back to reference Kim MR, et al. Enhancement of vascular endothelial growth factor-mediated angiogenesis in tamoxifen-resistant breast cancer cells: role of Pin1 overexpression. Mol Cancer Ther. 2009;8:2163–71.CrossRefPubMed Kim MR, et al. Enhancement of vascular endothelial growth factor-mediated angiogenesis in tamoxifen-resistant breast cancer cells: role of Pin1 overexpression. Mol Cancer Ther. 2009;8:2163–71.CrossRefPubMed
26.
go back to reference Schoots O, Voskoglou T, VanTol HHM. Genomic organization and promoter analysis of the human G-protein-coupled K+ channel Kir3.1 (KCNJ3/HGIRK1). Genomics. 1997;39:279–88.CrossRefPubMed Schoots O, Voskoglou T, VanTol HHM. Genomic organization and promoter analysis of the human G-protein-coupled K+ channel Kir3.1 (KCNJ3/HGIRK1). Genomics. 1997;39:279–88.CrossRefPubMed
27.
go back to reference Mirshahi T, Logothetis DE. Molecular determinants responsible for differential cellular distribution of G protein-gated inwardly rectifying K+ channels. J Biol Chem. 2004;279:11890–7.CrossRefPubMed Mirshahi T, Logothetis DE. Molecular determinants responsible for differential cellular distribution of G protein-gated inwardly rectifying K+ channels. J Biol Chem. 2004;279:11890–7.CrossRefPubMed
28.
go back to reference Kennedy ME, Nemec J, Clapham DE. Localization and interaction of epitope-tagged GIRK1 and CIR inward rectifier K+ channel subunits. Neuropharmacology. 1996;35:831–9.CrossRefPubMed Kennedy ME, Nemec J, Clapham DE. Localization and interaction of epitope-tagged GIRK1 and CIR inward rectifier K+ channel subunits. Neuropharmacology. 1996;35:831–9.CrossRefPubMed
29.
go back to reference Kubo Y, Iizuka M. Identification of domains of the cardiac inward rectifying K+ channel, CIR, involved in the heteromultimer formation and in the G-protein gating. Biochem Biophys Res Commun. 1996;227:240–7.CrossRefPubMed Kubo Y, Iizuka M. Identification of domains of the cardiac inward rectifying K+ channel, CIR, involved in the heteromultimer formation and in the G-protein gating. Biochem Biophys Res Commun. 1996;227:240–7.CrossRefPubMed
30.
go back to reference Kennedy ME, Nemec J, Corey S, Wickman K, Clapham DE. GIRK4 confers appropriate processing and cell surface localization to G-protein-gated potassium channels. J Biol Chem. 1999;274:2571–82.CrossRefPubMed Kennedy ME, Nemec J, Corey S, Wickman K, Clapham DE. GIRK4 confers appropriate processing and cell surface localization to G-protein-gated potassium channels. J Biol Chem. 1999;274:2571–82.CrossRefPubMed
31.
go back to reference Fernandez-Alacid L, et al. Subcellular compartment-specific molecular diversity of pre- and post-synaptic GABA-activated GIRK channels in Purkinje cells. J Neurochem. 2009;110:1363–76.CrossRefPubMedPubMedCentral Fernandez-Alacid L, et al. Subcellular compartment-specific molecular diversity of pre- and post-synaptic GABA-activated GIRK channels in Purkinje cells. J Neurochem. 2009;110:1363–76.CrossRefPubMedPubMedCentral
32.
go back to reference Chung HJ, Qian X, Ehlers M, Jan YN, Jan LY. Neuronal activity regulates phosphorylation-dependent surface delivery of G protein-activated inwardly rectifying potassium channels. Proc Natl Acad Sci U S A. 2009;106:629–34.CrossRefPubMed Chung HJ, Qian X, Ehlers M, Jan YN, Jan LY. Neuronal activity regulates phosphorylation-dependent surface delivery of G protein-activated inwardly rectifying potassium channels. Proc Natl Acad Sci U S A. 2009;106:629–34.CrossRefPubMed
33.
34.
go back to reference Chung HJ, et al. G protein-activated inwardly rectifying potassium channels mediate depotentiation of long-term potentiation. Proc Natl Acad Sci U S A. 2009;106:635–40.CrossRefPubMed Chung HJ, et al. G protein-activated inwardly rectifying potassium channels mediate depotentiation of long-term potentiation. Proc Natl Acad Sci U S A. 2009;106:635–40.CrossRefPubMed
35.
go back to reference Karapetyan AR, Buiting C, Kuiper RA, Coolen MW. Regulatory roles for long ncRNA and mRNA. Cancer. 2013;5:462–90.CrossRef Karapetyan AR, Buiting C, Kuiper RA, Coolen MW. Regulatory roles for long ncRNA and mRNA. Cancer. 2013;5:462–90.CrossRef
36.
go back to reference Shen Z, Yang Q, You QD. Researches toward potassium channels on tumor progressions. Curr Top Med Chem. 2009;9:322–9.CrossRefPubMed Shen Z, Yang Q, You QD. Researches toward potassium channels on tumor progressions. Curr Top Med Chem. 2009;9:322–9.CrossRefPubMed
38.
40.
41.
go back to reference Afrasiabi E, et al. Expression and significance of HERG (KCNH2) potassium channels in the regulation of MDA-MB-435S melanoma cell proliferation and migration. Cell Signal. 2010;22:57–64.CrossRefPubMed Afrasiabi E, et al. Expression and significance of HERG (KCNH2) potassium channels in the regulation of MDA-MB-435S melanoma cell proliferation and migration. Cell Signal. 2010;22:57–64.CrossRefPubMed
43.
go back to reference Voloshyna I, et al. TREK-1 is a novel molecular target in prostate cancer. Cancer Res. 2008;68:1197–203.CrossRefPubMed Voloshyna I, et al. TREK-1 is a novel molecular target in prostate cancer. Cancer Res. 2008;68:1197–203.CrossRefPubMed
44.
go back to reference Muller A, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410:50–6.CrossRefPubMed Muller A, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410:50–6.CrossRefPubMed
45.
go back to reference Mundy GR. Metastasis to bone: causes, consequences and therapeutic opportunities. Nat Rev Cancer. 2002;2:584–93.CrossRefPubMed Mundy GR. Metastasis to bone: causes, consequences and therapeutic opportunities. Nat Rev Cancer. 2002;2:584–93.CrossRefPubMed
47.
go back to reference Smrcka AV. Molecular targeting of G alpha and G beta gamma subunits: a potential approach for cancer therapeutics. Trends Pharmacol Sci. 2013;34:290–8.CrossRefPubMedPubMedCentral Smrcka AV. Molecular targeting of G alpha and G beta gamma subunits: a potential approach for cancer therapeutics. Trends Pharmacol Sci. 2013;34:290–8.CrossRefPubMedPubMedCentral
Metadata
Title
Overexpression of KCNJ3 gene splice variants affects vital parameters of the malignant breast cancer cell line MCF-7 in an opposing manner
Authors
S. Rezania
S. Kammerer
C. Li
B. Steinecker-Frohnwieser
A. Gorischek
T. T. J. DeVaney
S. Verheyen
C. A. Passegger
N. Ghaffari Tabrizi-Wizsy
H. Hackl
D. Platzer
A. H. Zarnani
E. Malle
S. W. Jahn
T. Bauernhofer
W. Schreibmayer
Publication date
01-12-2016
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2016
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2664-8

Other articles of this Issue 1/2016

BMC Cancer 1/2016 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