Open access peer-reviewed chapter

Role of an Alternatively Spliced KCNMA1 Variant in Glioma Growth

Written By

Divya Khaitan, Nagendra Ningaraj and Lincy B. Joshua

Submitted: 13 December 2017 Reviewed: 26 January 2018 Published: 22 August 2018

DOI: 10.5772/intechopen.74509

From the Edited Volume

Brain Tumors - An Update

Edited by Amit Agrawal, Luis Rafael Moscote-Salazar

Chapter metrics overview

1,075 Chapter Downloads

View Full Metrics

Abstract

Gliomas develop genetic traits to rapidly form aggressive phenotypes. Hence, management of gliomas is complicated and difficult. Besides genetic aberrations such as oncogenic copy number variation and mutations, alternative mRNA splicing triggers prooncogenic episodes in many cancers. In gliomas, we found alternative splicing at the KCNMA transcription process. KCNMA1 encodes the pore forming α-subunit of large-conductance calcium-activated voltage-sensitive potassium (BKCa) channels. These channels play critical role in glioma invasion and proliferation. We identified a novel KCNMA1 mRNA splice variant with a deletion of 108 base pairs (KCNMA1v) mostly overexpressed in high grade gliomas. We found that KCNMA1 alternative pre-mRNA splicing enhanced glioma growth, progression and diffusion. The role of KCNMA1 and its splicing as a critical posttranscriptional regulator of BKCa channel expression is presented in this chapter. Our research implies that high grade gliomas express KCNMA1v and BKCa channel isoform to accelerate growth and transformation to glioblastoma multiforme (GBM). We demonstrated that tumors hardly develop in mice injected with KCNMA1v transfected cell line expressing short-hairpin RNA (shRNA) compared to mice injected with KCNMA1v transected glioma cells. We conclude that targeting the KCNMA1 variants may be a clinically beneficial strategy to prevent or at least slow down glioma transformation to GBM.

Keywords

  • KCNMA1
  • BKCa channels
  • gliomas
  • tumorigenicity
  • potassium channels

1. Introduction

1.1. KCNMA1-encoded BKCa channels in glioma

Brain tumors are the most common type of solid tumors. In the United States, an estimated 20,000 new primary brain tumor cases are reported [1]. The most common form of malignant glioma is glioblastoma multiforme (GBM). The treatment of brain tumors is highly complicated due to their highly aggressive phenotypic and genotypic changes [2]. The median survival among GBM patients is only 15 months or less [3]. GBM contains heterogeneous subpopulations of glioma and other mixed supporting cells that are cancerous cells. They have the intrinsic ability that adapt in the brain tumor microenvironment and invade the normal brain. Gene expression profiling studies have identified many genes that have distinct expression patterns among different histological types and grades of gliomas [4]. The response of “normal cells” to malignant transformation involves changes in gene expression and is thought to be regulated by transcription [5]. The potassium ion channels are implicated in the malignant transformation to a higher grade in several cancers [5, 6, 7]. For example, we reported that low-grade gliomas might undergo certain epigenetic changes to develop into a GBM [8].

The physiological features of BKCa channels also known as maxi K or BK channels are well described [6, 7, 8, 9]. These channels are unique since its activity is triggered by depolarization and enhanced by an increase in μM range of cytosolic calcium (Figure 1). The BKCa channels provide a crucial link between the metabolic and electrical states of cells. The BKCa channel overexpression was observed in biopsies of patients with malignant gliomas compared with nonmalignant human cortical tissues and the level of expression correlated positively with increased malignancy [7]. Studies have shown the importance of BKCa channels in brain tumor biology [5]. Lastly, BKCa currents in glioma cells are more sensitive to intracellular [Ca2+] compared to BKCa channels in healthy glial cells [9, 10].

Figure 1.

BKCa channel is a tetramer of four monomeric pore-forming alpha-subunits encoded by KCNMA1. The seven transmembrane channels S0 to S6 are voltage-sensing domains, S1 to S4 channels form pore domain, S5 is a selectivity filter, and S6 is a extracellular N-terminal segment. The cytoplasmic C-terminal domain has RCK1 and RCK2 (with calcium bowl) segments.

Advertisement

2. Diverse role of KCNMA1 in glioma

KCNMA1-encoded BKCa channel plays a pivotal role in cancer cell proliferation. Amplification of KCNMA1 was observed in breast, ovarian, and endometrial cancer with the highest prevalence in invasive ductal breast cancers and serous carcinoma of ovary and endometrium (3–7%) and gliomas. KCNMA1 amplification was significantly associated with high tumor stage, high-grade, high tumor cell proliferation, and poor prognosis. Due to the large number of protein interactions and activating factors influencing BKCa channel function, intracellular Ca2+, membrane voltage, pH, shear stress, carbon monoxide, phosphorylation states, and steroid hormones, it is generally difficult to predict its direct role in a given tissue. However, in many diseases including cancers, defective regulation and/or expression of BKCa channels have repeatedly been associated with altered cell cycle progression [11], cell proliferation [11], and cell migration [11]. These altered cell functions are implicated in development of malignancy [11].

Advertisement

3. KCNMA1: STRING analysis

In order to understand the possible interactions of KCNMA1 with other genes and molecules, we used the tool STRING 9.1. It is a database consisting of known and possible protein–protein interactions with a gene of interest. The gene may have a direct (physical) or indirect (functional) association with other molecules. With this tool we can easily identify possible interaction of KCNMA1 with other associated molecules. We can derive detailed information of the protein being investigated as well as its associated molecules, crystal structure of the proteins with its PDB ID, and combined score [confidence score, neighborhood score, fusion score, homology score] on the basis of some parameters like experimental results, text-mining, co expression, databases, and co-occurrence (Figure 2).

Figure 2.

STRING 9.1 software-derived possible association of KCNMA1 with top 20 most interacting genes.

Advertisement

4. Possible KEGG pathway following activation and suppression of KCNMA1 in glioma cells

Glioma cell line U-87 MG was obtained from the American Type Culture Collection (Manassas, VA) and cultured in MEM supplemented with 10% FBS and 0.1 mM nonessential amino acids. Cells were maintained at 37°C in 5% CO2. In order to study the biological significance of up- and down-regulation of KCNMA1 on associated genes, we performed microarray using the Affymetrix Human Genome U133 Plus 2.0. Array analyses of U-87 MG cell lines where KCNMA1 was either overexpressed or suppressed showed significant changes in genes involved in cell proliferation, angiogenesis, cell cycle, and invasion (Figure 3). Class comparison tests indicated significant changes in global expression patterns. Twenty genes highly downregulated by suppression but upregulated by overexpression of KCNMA1 or vice versa are shown in Figure 3. This data support our rationale that KCNMA1 plays a critical role in the above cellular processes.

Figure 3.

High-grade glioma cells, U-87 MG were transfected to make stable cell line over-expressing KCNMA1. In addition, we transfected U-87 MG cells with shKCNMA1; this suppressed basal KCNMA1 expression. A microarray was performed on these cells. The heat map shows the differential expression of genes that were directly or indirectly affected by upregulation or down regulation of KCNMA1. We found 8102 and 7259 significant features at p < 0.05, respectively, for overexpression and suppression of KCNMA1. From these, features having at least a signal value of 255 were selected to reduce false positives (false discovery rate < 0.0079).

Array analyses of U-87 MG cell lines where KCNMA1 was either overexpressed or suppressed showed significant changes in genes involved in cell proliferation, angiogenesis, cell cycle, and invasion (Figure 4; see arrows). Cluster analysis of 476 transcripts that are altered in opposite directions by expressing KCNMA1 gene up and down in U-87 cells. These transcripts were identified from the significantly altered genes by ANOVA at p < 0.05, and the fold-change thresholds such that one of KCNMA1 up or down altered the gene expression by twofold and the other altered it at least by 1.5-fold in the opposite direction.

Figure 4.

Cluster analysis of 62 genes altered by twofold in opposite directions by expressing KCNMA1 gene up and down in U-87 cells. These transcripts were identified from the significantly altered genes by ANOVA at p < 0.05.

Advertisement

5. KCNMA1 splicing in glioma

The KCNMA1 encodes the pore-forming α-subunits of large-conductance Ca2+-activated K+ (BKCa) channels. More than 20 variants of this gene are associated with alternative splicing at ten or more different sites [12, 13], while majority of the splice sites are located in the large cytoplasmic domain. This domain is called the C-terminal half of the channel that contains multiple Ca2+ binding sites [14, 15, 16]. Gating properties and kinetics with regard to the voltage and Ca2+ dependence of gating are altered by alternative splicing in these regions [17, 18, 19]. Expressions of different BKCa isoforms have been implicated in auditory processing [20] and alter the sensitivity of BKCa to modulation by phosphorylation [21] and other processes [22]. However, the role of BKCa isoforms in cancer is now being investigated [23]. More specifically, KCNMA1 is altered in a wide variety of cancers, and their overexpression liked to increased malignancy in gliomas [4, 5, 6, 7]. The BKCa protein isoform transcribed by its alternatively spliced mRNA in cancer cells is known as likely to respond differently to changes in intracellular calcium ([Ca2+]i) and membrane potential. We and others have demonstrated that BKCa channels are overexpressed in gliomas [4, 5, 6, 7, 8, 9] and play an important role in glioma invasion and migration [24, 25].

BKCa channels show a variety of electrophysiological properties due to alternative splicing of their α-subunits. In glioma cells, Liu et al. [6] reported that BKCa channels exhibit distinct electrophysiological properties due to alternate splicing of its α-subunits. These BKCa variants showed higher Ca2+ sensitivity in glioma cells compared to BKCa channels present in normal glial cells. The amplified sensitivity to intracellular [Ca2+i] was shown in a novel splice isoform (gBK) of hSlo, the gene that encodes the α-subunits, specifically expressed in glioma [6]. We have recently shown (submitted for review) that KCNMA1 that encodes α-subunit (pore forming) of BKCa channel undergoes specific splicing at mRNA to form a variant (KCNMA1v) that encodes for a novel BKCa channel isoform only in glioblastoma multiforme (GBM). Other types of Ca2+-activated K+ channels such as intermediate (IKCa) and small (SKCa) [10] have been characterized in human glioma cells, but their roles in brain tumor biology are yet to be explored.

The alternative RNA splicing might increase protein expression levels and functions. In cancer, it was shown that abnormal mRNA splicing often leads to tumor-promoting splice variants that are translated into activated oncogenes or inactivated tumor suppressors [26, 27]. Interestingly, the brain appears to have maximum alternative splicing of exons [28]. The present knowledge suggests that alternative or aberrant pre-mRNA splicing results in oncoproteins with diverse functions in the development, progression, and dispersal of glioma cells [29, 30]. Further, genomic studies have shown that gliomas often have splice isoforms than in normal brain [30]. For instance, KCNMA1 was shown to undergo alternative pre-mRNA splicing at several sites in humans and mice [31, 32] to generate physiologically diverse BKCa channels. These altered BKCa channels respond differently to calcium/voltage changes. Often, these channels show abnormal regulation of cellular signaling pathways in glioma cells [13, 19]. Hence, the cause–effect of KCNMA1 splicing in functional modification of BKCa channels in brain tumors is a matter of great interest.

We have described an unknown KCNMA1 mRNA splice variant with a deletion of 108 base pairs of exon 22 (KCNMA1v) between the S9 and S10 protein subunits (C-terminus) overexpressed in high-grade gliomas. This serendipitous finding prompted to study the role of KCNMA1v as a critical posttranscriptional regulator of BKCa channel isoform expression and altered channel function in gliomas (submitted for review). The complex interaction between various ions and their respective ion channels at the invadopodia of the malignant gliomas is speculated to explain some of the invasive properties of gliomas [24, 25]. The role of various ions and their respective ion channels in glioma is recently well documented [33]. Among many ion channels, BKCa channels have many known spliced variants. Liu et al. have initially described a spliced variant, glioma BK (gBK), channel in human glioma cells [6]. Inherited and acquired changes in pre-mRNA splicing have been shown to play a significant role in human disease development (pre-mRNA splicing and human disease [29]. Venables et al. [34] showed that alternative splicing of pre-mRNA increases the diversity of protein functions in ovarian and breast cancer samples. Specifically, they found that expression of FOX2 was downregulated in ovarian cancer and its splicing is altered in breast cancer samples affecting cell proliferation.

However, studies on the association of changes in gene splicing pattern and malignancy are rare. However, few studies have shown the presence of BKCa channels at the invadopodia of the malignant gliomas that lead to speculation that these channels may help the invasive properties of gliomas. A recent study found a clinical relevance where the investigators found T cells derived from GBM patients who were sensitized to the gBK peptide could also kill target cells expressing gBK. This study shows that peptides derived from cancer-associated ion channels maybe useful targets for T-cell-mediated immunotherapy [23]. Several sites of alternative pre-mRNA splicing of KCNMA1 have been described, and majority of them are located within the intracellular C-terminal domain of the channel [19]. In the past a novel splice variant of KCNMA1 (gBK) with an additional 34-amino-acid exon at splice site 2 in the C-terminal has already been described in gliomas [6].

In addition to the above studies, we present herein the cloning, functional characterization, and splicing of a novel KCNMA1 splice variant. KCNMA1 encodes the alpha-subunit of human BKCa channels and is known to form BKCa channel isoforms. Here, we report hitherto unknown KCNMA1 splice variant, which has a 108-base-pair deletion at the splice site on one of its exons, which we termed as KCNMA1v. More importantly, KCNMA1v expression correlates positively with the relative degree of malignancy of the glioma cell lines (under publication). Moreover, we found that KCNMA1v was expressed only in high-grade glioma samples and not in normal brain tissues as evidenced by examination of human biopsy specimens (under publication). Expression of KCNMA1v in HEK (null type) revealed that the pharmacological and biophysical properties of the variant were consistent with the properties of wild-type KCNMA1 gene in glioma cells suggesting that KCNMA1v is likely to encode the principal wild-type BKCa channels (under publication). Although we have not separated wild-type and splice variant isoform for sequence and structure analysis, the biological properties of both wild-type and isoform protein appear to be similar. However, when overexpressed in glioma cell line (under publication), the variant showed distinct biological properties such as enhanced Ca2+ sensitivity at physiologically relevant [Ca2+]i levels (under publication).

Progression of brain tumor from localized, slow-growing tumors to more aggressive brain tumors capable of invading the surrounding brain most likely involves a series of stepwise biological events [35]. For example, miR-182 was found to be a valuable marker of glioma progression and that high miR-182 expression is associated with poor prognosis [36]. Such a multistep process of tumorigenesis has been proposed to involve a series of mutational events which ultimately lead to development and progression of neoplasia [35]. Aberrant pre-mRNA splicing is an important factor in tumor progression and has been proposed to result in the loss of a normal pathway of differentiation, which could lead to tumor progression. Several studies have implicated BKCa channel expression to oncogenic cell transformation [37, 38]. Increased activity of BKCa channels appeared to be required for the mitogenic stimulation of non-transformed cells and may play a role in cell proliferation [39]. Consistent with the above studies, we show that KCNMA1v-induced effects promote proliferation in glioma cell lines when the variant was overexpressed. The upregulation of KCNMA1v in glioma cell lines provides an opportunity to determine variant-specific changes that enhance gliomagenesis in vivo. The overexpression of KCNMA1v resulted in increased proliferation in glioma cell lines. It has also been suggested that cell invasion into narrow brain spaces may require tumor cells to shrink and squeeze through tight interstitial space [40]. Cell shrinkage requires the efflux of K+ and Cl ions [41], and BKCa channels may serve as pathway for regulated K+ efflux [42]. Consistent with these findings, the overexpression of KCNMA1v increased the invasion potential of glioma cells (under publication). The role of BKCa channels in cell migration was already described [43]. The changes in proliferation and migration of cells over-expressing KCNMA1v were mostly attributed to increased levels of KCNMA1 and BKCa channel protein expression in transfected cells. Additionally, overexpression of KCNMA1v in glioma cells may assist them to diffusely invade the normal brain. Due to this phenomenon, GBM patients typically show high propensity to recur as the cancer cells expressing KCNMA1v might survive surgical and therapeutic treatment. The xenograft tumors in mice likewise demonstrated increased growth, which correlated well with Ki-67 expression (under publication). The overexpression of KCNMA1v resulted in increased angiogenesis in the tumor xenografts, supporting the angiogenic role of KCNMA1v. The observation that the overexpression of KCNMA1v in human gliomas correlates with increased angiogenesis in high-grade gliomas further supports that KCNMA1 splicing event is an important biological process for glioma progression. Consistent with this observation, we found that glioma cells over-expressing KCNMA1v secreted significantly the high level of angiogenic factor VEGF (under publication).

Advertisement

6. Conclusion

Further investigation into the mechanisms and cellular events caused by KCNMA1 splicing may lead to the development of future therapies for this highly deadly disease. Splice variants that are found in high-grade gliomas have clear diagnostic and prognostic values besides providing potential targets for anticancer drug development. Clinical outcome of KCNMA1v expression in high-grade glioma is expected to reveal the variants’ clinical importance. This analysis is being performed in our laboratory. In conclusion, the results presented here might suggest that quantifying the levels of KCNMA1v could be useful to identify biological process that increases the malignancy and affect prognosis of high-grade glioma patients.

Advertisement

Acknowledgments

The authors thank the Scintilla Group, Bangalore, India; Anderson Cancer Institute and Mercer University Medical Center, Savannah, GA, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Cedars-Sinai Medical Center, Los Angeles, CA, USA; American Cancer Society, USA; Georgia Cancer Coalition, Atlanta, GA, USA; and NIH for providing opportunity and research grant support. We also thank Dr. Nagendra of MVIT, Bangalore, for assisting us with the STRING software for the analysis and Michigan State University Research Center, Grand Rapids, MI, USA, for generating KKEG pathway using Affymetrix analysis data.

References

  1. 1. Rich JN, Bigner DD. Development of novel targeted therapies in the treatment of malignant glioma. Nature Reviews. Drug Discovery. 2004 May 1;3(5):430-446
  2. 2. Claes A, Idema AJ, Wesseling P. Diffuse glioma growth: A guerilla war. Acta Neuropathologica. 2007 Nov 1;114(5):443-458
  3. 3. Krex D, Klink B, Hartmann C, von Deimling A, Pietsch T, Simon M, Sabel M, Steinbach JP, Heese O, Reifenberger G, Weller M. Long-term survival with glioblastoma multiforme. Brain. 2007 Sep 4;130(10):2596-2606
  4. 4. Liang Y, Diehn M, Watson N, Bollen AW, Aldape KD, Nicholas MK, Lamborn KR, Berger MS, Botstein D, Brown PO, Israel MA. Gene expression profiling reveals molecularly and clinically distinct subtypes of glioblastoma multiforme. Proceedings of the National Academy of Sciences of the United States of America. 2005 Apr 19;102(16):5814-5819
  5. 5. Weaver AK, Bomben VC, Sontheimer H. Expression and function of calcium-activated potassium channels in human glioma cells. GLIA. 2006 Aug 15;54(3):223-233
  6. 6. Liu X, Chang Y, Reinhart PH, Sontheimer H. Cloning and characterization of glioma BK, a novel BK channel isoform highly expressed in human glioma cells. The Journal of Neuroscience. 2002 Mar 1;22(5):1840-1849
  7. 7. Weaver AK, Liu X, Sontheimer H. Role for calcium-activated potassium channels (BK) in growth control of human malignant glioma cells. Journal of Neuroscience Research. 2004 Oct 15;78(2):224-234
  8. 8. Khaitan D, Reddy PL, Ningaraj NS. Differential epigenetic inactivation of genes in Gliomas. EC Pharmacology and Toxicology (3.6). 2017:198-207
  9. 9. Ningaraj NS. Drug delivery to brain tumours: Challenges and progress. Expert Opinion on Drug Delivery. 2006 Jul 1;3(4):499-509
  10. 10. Ningaraj NS, Rao M, Black KL. Calcium-dependent potassium channels as a target protein for modulation of the blood-brain tumor barrier. Drug News & Perspectives. 2003 Jun 1;16(5):291-298
  11. 11. Chaudhary MI, editor. Frontiers in Drug Design & Discovery: Volume: 7. Bentham Science Publishers; 2016 May. p. 238
  12. 12. Meera P, Wallner M, Toro L. Molecular biology of high-conductance, Ca2+-activated potassium channels. In: Potassium channels in cardiovascular biology. Boston, MA: Springer; 2001. pp. 49-70
  13. 13. Shipston MJ. Alternative splicing of potassium channels: A dynamic switch of cellular excitability. Trends in Cell Biology. 2001 Sep 1;11(9):353-358
  14. 14. Xia XM, Zeng X, Lingle CJ. Multiple regulatory sites in large-conductance calcium-activated potassium channels. Nature. 2002 Aug 22;418(6900):880-884
  15. 15. Zeng XH, Xia XM, Lingle CJ. Divalent cation sensitivity of BK channel activation supports the existence of three distinct binding sites. The Journal of general physiology. 2005 Mar 1;125(3):273-286
  16. 16. Krishnamoorthy G, Shi J, Sept D, Cui J. The NH 2 terminus of RCK1 domain regulates Ca 2+−dependent BK Ca channel gating. The Journal of general physiology. 2005 Sep 1;126(3):227-241
  17. 17. Adelman JP, Shen KZ, Kavanaugh MP, Warren RA, Wu YN, Lagrutta A, Bond CT, North RA. Calcium-activated potassium channels expressed from cloned complementary DNAs. Neuron. 1992 Aug 31;9(2):209-216
  18. 18. Saito M, Nelson C, Salkoff L, Lingle CJ. A cysteine-rich domain defined by a novel exon in aSlo variant in rat adrenal chromaffin cells and PC12 cells. The Journal of Biological Chemistry. 1997 May 2;272(18):11710-11717
  19. 19. Chen L, Tian L, MacDonald SH, McClafferty H, Hammond MS, Huibant JM, Ruth P, Knaus HG, Shipston MJ. Functionally diverse complement of large conductance calcium-and voltage-activated potassium channel (BK) α-subunits generated from a single site of splicing. The Journal of Biological Chemistry. 2005 Sep 30;280(39):33599-33609
  20. 20. Fettiplace R, Fuchs PA. Mechanisms of hair cell tuning. Annual Review of Physiology. 1999 Mar;61(1):809-834
  21. 21. Tian L, Duncan RR, Hammond MS, Coghill LS, Wen H, Rusinova R, Clark AG, Levitan IB, Shipston MJ. Alternative splicing switches potassium channel sensitivity to protein phosphorylation. The Journal of Biological Chemistry. 2001 Mar 16;276(11):7717-7720
  22. 22. Erxleben C, Everhart AL, Romeo C, Florance H, Bauer MB, Alcorta DA, Rossie S, Shipston MJ, Armstrong DL. Interacting effects of N-terminal variation and strex exon splicing on slo potassium channel regulation by calcium, phosphorylation, and oxidation. The Journal of Biological Chemistry. 2002 Jul 26;277(30):27045-27052
  23. 23. Ge L, Hoa NT, Cornforth AN, Bota DA, Mai A, Kim DI, Chiou SK, Hickey MJ, Kruse CA, Jadus MR. Glioma big potassium channel expression in human cancers and possible T cell epitopes for their immunotherapy. The Journal of Immunology. 2012 Sep 1;189(5):2625-2634
  24. 24. Sontheimer H. Ion channels and amino acid transporters support the growth and invasion of primary brain tumors. Molecular Neurobiology. 2004 Feb;29(1, 1):61-71
  25. 25. Sontheimer H. An unexpected role for ion channels in brain tumor metastasis. Experimental Biology and Medicine. 2008 Jul;233(7):779-791
  26. 26. Chen J, Weiss WA. Alternative splicing in cancer: Implications for biology and therapy. Oncogene. 2015 Jan 2;34(1):1-4
  27. 27. Yeo G, Holste D, Kreiman G, Burge CB. Variation in alternative splicing across human tissues. Genome Biology. 2004 Sep 13;5(10):R74
  28. 28. Xu Q, Modrek B, Lee C. Genome-wide detection of tissue-specific alternative splicing in the human transcriptome. Nucleic Acids Research. 2002 Sep 1;30(17):3754-3766
  29. 29. Faustino NA, Cooper TA. Pre-mRNA splicing and human disease. Genes & Development. 2003 Feb 15;17(4):419-437
  30. 30. Cheung HC, Baggerly KA, Tsavachidis S, Bachinski LL, Neubauer VL, Nixon TJ, Aldape KD, Cote GJ, Krahe R. Global analysis of aberrant pre-mRNA splicing in glioblastoma using exon expression arrays. BMC Genomics. 2008 May 12;9(1):216
  31. 31. Beisel KW, Rocha-Sanchez SM, Ziegenbein SJ, Morris KA, Kai C, Kawai J, Carninci P, Hayashizaki Y, Davis RL. Diversity of Ca2+-activated K+ channel transcripts in inner ear hair cells. Gene. 2007 Jan 15;386(1):11-23
  32. 32. Fleishman SJ, Yifrach O, Ben-Tal N. An evolutionarily conserved network of amino acids mediates gating in voltage-dependent potassium channels. Journal of Molecular Biology. 2004 Jul;340(2, 2):307-318
  33. 33. Ningaraj NS, Khaitan D. Targeting ion channels for drug delivery to brain tumors. In: Rahman A-U, Choudhary I, editors. Frontiers in Anti-Cancer Drug Discovery. Vol. 5. University of Cambridge, UK: Bentham Science Publishers Ltd; 2015. pp. 231-246
  34. 34. Venables JP, Klinck R, Koh C, Gervais-Bird J, Bramard A, Inkel L, Durand M, Couture S, Froehlich U, Lapointe E, Lucier JF. Cancer-associated regulation of alternative splicing. Nature Structural & Molecular Biology. 2009 Jun 1;16(6):670-676
  35. 35. McNamara MG, Sahebjam S, Mason WP. Emerging biomarkers in glioblastoma. Cancer. 2013 Aug 22;5(3):1103-1119
  36. 36. Jiang L, Mao P, Song L, Wu J, Huang J, Lin C, Yuan J, Qu L, Cheng SY, Li J. miR-182 as a prognostic marker for glioma progression and patient survival. The American Journal of Pathology. 2010 Jul 31;177(1):29-38
  37. 37. Huang X, Jan LY. Targeting potassium channels in cancer. The Journal of Cell Biology. 2014 Jul 21;206(2):151-162
  38. 38. Pardo LA, Stühmer W. The roles of K+ channels in cancer. Nature Reviews. Cancer. 2014 Jan;14(1, 1):39-48
  39. 39. Si H, Grgic I, Heyken WT, Maier T, Hoyer J, Reusch HP, Köhler R. Mitogenic modulation of Ca2+-activated K+ channels in proliferating A7r5 vascular smooth muscle cells. British Journal of Pharmacology. 2006 Aug 1;148(7):909-917
  40. 40. Pardo LA, del Camino D, Sánchez A, Alves F, Brüggemann A, Beckh S, Stühmer W. Oncogenic potential of EAG K+ channels. The EMBO Journal. 1999 Oct 15;18(20):5540-5547
  41. 41. Blackiston DJ, McLaughlin KA, Levin M. Bioelectric controls of cell proliferation: Ion channels, membrane voltage and the cell cycle. Cell Cycle. 2009 Nov 1;8(21):3527-3536
  42. 42. Kondratskyi A, Kondratska K, Skryma R, Prevarskaya N. Ion channels in the regulation of apoptosis. Biochimica et Biophysica Acta (BBA) - Biomembranes. 2015 Oct 31;1848(10):2532-2546
  43. 43. Sciaccaluga M, Fioretti B, Catacuzzeno L, Pagani F, Bertollini C, Rosito M, Catalano M, D'Alessandro G, Santoro A, Cantore G, Ragozzino D. CXCL12-induced glioblastoma cell migration requires intermediate conductance Ca 2+−activated K+ channel activity. American Journal of Physiology-Cell Physiology. 2010 July 1;299(1):C175-C184

Written By

Divya Khaitan, Nagendra Ningaraj and Lincy B. Joshua

Submitted: 13 December 2017 Reviewed: 26 January 2018 Published: 22 August 2018