Editorial Open Access
Copyright ©2011 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Stem Cells. Mar 26, 2011; 3(3): 19-24
Published online Mar 26, 2011. doi: 10.4252/wjsc.v3.i3.19
Functional ion channels in stem cells
Gui-Rong Li, Departments of Medicine and Physiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong, China
Xiu-Ling Deng, Department of Physiology and Pathophysiology, School of Medicine, Xi’an Jiaotong University, Xi’an 710049, Shaanxi Province, China
Author contributions: Li GR and Deng XL both contributed to this paper.
Supported by (in part) Grants (734703M and 8CRF09) from the Research Grant Council of Hong Kong, China
Correspondence to: Dr. Gui-Rong Li, Associate Professor, L4-59, Laboratory Block, FBM, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong, China. grli@hkucc.hku.hk
Telephone: +852-2819-9513 Fax: +852-2855-9730
Received: October 30, 2010
Revised: January 14, 2011
Accepted: January 21, 2011
Published online: March 26, 2011

Abstract

Bioelectrical signals generated by ion channels play crucial roles in excitation genesis and impulse conduction in excitable cells as well as in cell proliferation, migration and apoptosis in proliferative cells. Recent studies have demonstrated that multiple ion channels are heterogeneously present in different stem cells; however, patterns and phenotypes of ion channels are species- and/or origin-dependent. This editorial review focuses on the recent findings related to the expression of functional ion channels and the roles of these channels in regulation of cell proliferation in stem cells. Additional effort is required in the future to clarify the ion channel expression in different types of stem cells; special attention should be paid to the relationship between ion channels and stem cell proliferation, migration and differentiation.

Key Words: Stem cells, Ion channels, Proliferation



STEM CELLS

Stem cells are found in all multi-cellular organisms and are characterized by the ability to self-renew through mitotic cell division and differentiate into a diverse range of specialized cell types. There are two types of original mammalian stem cells: embryonic stem cells and adult stem cells found in adult tissues. In addition, it has recently been found that induced pluripotent stem cells (iPS) can be developed from other types of cells including fibroblasts[1].

Embryonic stem (ES) cells are derived from mammalian embryos in the blastocyst phase of development[2,3]. Adult (or somatic) stem cells were initially isolated from mouse bone marrow[4]; further studies have shown that stem cells are present in different types of tissue including brain, heart, blood vessels, skeletal muscles, skin, liver and fat tissue. Adult stem cells remain in a quiescent or non-dividing state and can be activated by disease or tissue injury. In adults, stem cells/progenitor cells act as a repair system for the body and maintain the normal turnover of regenerative organs such as blood, skin and intestinal tissues[5].

iPS cells are recently developed cells induced from somatic cells such as skin fibroblasts and B lymphocytes[6]. They were generated initially by Takahashi & Yamanaka[1] by reprogramming somatic cells by over-expressing a combination of four transcription factors: octamer 3/4 (Oct4), SRY box-containing gene 2 (Sox2), Kruppel-like factor 4 (Klf4) and c-Myc in murine fibroblasts to induce the cells enter an embryonic-like state[1,7]. The iPS cells are then produced by introducing the four transcription factor–encoding genes into human fibroblasts[7]. Two other groups produced similar iPS cells by introducing slightly different combinations of genes: POU5F1 (OCT4), SOX2, NANOG and LIN28A (LIN28)[8,9]. These iPS cells are similar to ES cells in morphology, growth properties and expression of phenotypic markers. These cells closely resemble ES cells and can differentiate into multiple types of cells in vitro and in vivo[1,6,7,9]. ES cells, adult tissue mesenchymal stem cells (MSCs) and their progenitors and iPS cells all possess potential therapeutic value in regenerative medicine.

In addition to the three major types of stem cells mentioned above that possess potential benefit for regenerative medicine, another type of stem cells is found within tumors or hematological cancers and has characteristics associated with normal stem cells, specifically the ability to give rise to all cell types found in a particular cancer sample. Cancer stem cells are tumorigenic in contrast to other non-tumorigenic cells[9] and may induce tumors through the stem cell processes: self-renewal and differentiation into multiple cell types. Moreover, cancer stem cells are believed to persist in tumors as a distinct population and cause relapse and metastasis by giving rise to new tumors. Therefore, cancer stem cells may be a target for developing specific therapies to improve survival and quality of life of cancer patients, especially sufferers of metastatic disease[10].

Although stem cells are important in regenerative medicine and/or cancer treatment, their cellular physiology and biology are not fully understood. Membrane ion channels are known to play a crucial role in proliferation, apoptosis and migration in a wide range of cells.

ION CHANNELS IN STEM CELLS

Multiple functional ion channel currents have been reported to be heterogeneously present in different types of stem cells. They include the voltage-gated delayed rectifier K+ current IKDR (encoded by different Kv genes), the Ca2+-activated K+ current KCa (including BKCa, large conductance KCa; IKCa, intermediate conductance KCa; and SKCa, small conductance KCa), the transient outward K+ current Ito (or A-type current, IA), inward rectifier K+ current (IKir), hyperpolarization-activated cyclic nucleotide-regulated cation current (Ih), chloride current (ICl), voltage-gated Na+ current (INa), L-type calcium current (ICa.L), transient receptor potential (TRP) nonselective cation currents. These currents have been found to be heterogeneously present in ES cells, mesenchymal stem cells (MSCs) from bone marrow, fat tissue and human umbilical cord vein, neural progenitor cells, cardiac progenitor cells or iPS cells derived from different species.

ION CHANNELS IN EMBRYONIC STEM CELLS

In ES cells, it has been reported that a tetraethylammonium (TEA)- and 4-aminopyridine (4-AP)-sensitive IKDR is co-present with iberiotoxin-sensitive BKCa in 52% of mouse ES cells and homogenously present in (100%) human ES cells[11]. However, phenotypes of IKDR differ between mouse and human ES cells. IKDR is encoded by Kv1.1, Kv1.2, Kv1.3 and Kv1.6 genes in mouse ES cells and by Kv7.2 and Kv9.3 in human ES cells. Interestingly, a Cs+-sensitive hyperpolarization-activated current (Ih, encoded by HCN3) is present in 23% of mouse ES cells but not in human ES cells. In addition, iberiotoxin-sensitive BKCa is encoded by MaxiK (Slo or KCa1.1) in mouse ES cells[11]. Although human ES cell and mouse ES cells share similar expression of many surface markers and intracellular signal pathways[12,13], significant differences are found in the expression of vimentin, h-III tubulin, alpha-fetoprotein, eomesodermin, HEB, ARNT and FoxD3 as well as in the expression of the LIF receptor complex LIFR/IL6ST (gp130)[12,14]. The different patterns and phenotypes of ion channel expression in human ES cells and mouse ES cells support the notion that some basic information on human ES cells can be derived from mouse ES cells; however, such information does not correspond on a one-to-one basis[14].

ION CHANNELS IN MESENCHYMAL STEM CELLS

A noise-like iberiotoxin-sensitive KCa and a 4-AP- and TEA-sensitive IKDR are detected in most human bone marrow-derived MSCs[15,16]. The noise-like KCa is encoded by MaxiK (KCa1.1 or Slo) as demonstrated by several research groups[15-17]. Our study demonstrates that IKDR shares similar characteristics with EAG channels cloned from the brain[18] which is encoded by hEAG1 (Kv10.1) in human MSCs[16]. In addition, a voltage-gated tetrodotoxin (TTX)-sensitive Na+ current (INa.TTX, encoded by hNE-Na or NaV1.7), a 4-AP-sensitive Ito (IA, encoded by Kv1.4 and Kv4.2)[16] and a nifedipine-sensitive ICa.L (encoded by CACNA1C or CaV1.2) are present in a small population (29%, 8% and 15% respectively) of human MSCs[16].

IKCa current (encoded by KCa3.1 or KCNN4), volume-sensitive Cl- current (ICl.vol, encoded by Clcn3) and IKir (encoded by Kir2.1) but not IKDR, are present in mouse bone marrow-derived MSCs[19]. The patterns and phenotypes of ion channels in mouse MSCs are different from mouse ES cells, suggesting that ion channel expression is origin-dependent.

In addition to INa.TTX (encoded by SCN2A), Ito (encoded by Kv1.4) and ICa.L (encoded by CCHL2a) recorded in a small population (16%, 10% and 8% respectively) of rat bone marrow MSCs, 4-AP sensitive IKDR (encoded by Kv1.2 and Kv2.1) is present in 91% of cells. BKCa (KCa1.1) and IKCa (KCa3.1) are co-present in 33% of rat MSCs[20]. Interestingly, IKDR (encoded by Kv1.2 and Kv2.1) is present in 78% of rabbit bone marrow MSCs, BKCa and IKCa are co-expressed with IKDR in 29% of cells, while IKir (encoded by Kir1.1) is present in 28% of cells[21]. These results demonstrate the different patterns and phenotypes of ion channels heterogeneously expressed in MSCs from mouse, rat, rabbit and human bone marrow, indicating a species-dependence of ion channel expression in bone marrow MSCs.

Interestingly, BKCa, INa.TTX, and Ito are present in 92%, 30% and 50% of MSCs from human umbilical cord vein and encoded by KCa1.1, hNE-Na, and Kv1.4 and Kv4.2 respectively[22], and Ba2+-sensitive IKir (encoded by TWIK and Kir2.1) is present in 5% of cells. However, no typical IKDR is recorded, although Kv1.1 and hEAG1 (Kv10.1) genes are detected in these cells[22]. In MSCs from human fat tissue[23], INa.TTX (encoded by hNE-Na) and 4-AP sensitive Ito are recorded in a small population (8% and 19%) of cells. In addition to 4-AP- and TEA-sensitive IKDR (likely encoded by the multiple genes Kv1.1, Kv1.5, Kv2.1, Kv7.3, Kv11.1 and Kv10.1) recorded in 73% of cells, three types of KCa currents sensitive to inhibition by the BKCa blocker iberiotoxin, IKCa blocker clotrimazole and SKCa blocker apamin are present and the corresponding channel genes (KCa1.1, KCa3.1 and KCa2.3) are detected in human fat tissue-derived MSCs[23]. These studies suggest that patterns and phenotypes of ion channel expression in MSCs are species- and/or tissue-specific dependent.

ION CHANNELS IN NEURAL STEM/PROGENITOR CELLS

In neural stem/progenitor cells, an earlier study reported that two types of K+ currents, IKDR (encoded by Kv1.2, Kv1.5 and Kv1.6) and IA (encoded by Kv1.4), were co-expressed in oligodendrocyte progenitor cells and differentiated cultured oligodendrocytes from neonatal rats[24]. Recent studies demonstrated that both Ba2+-sensitive IKir (encoded by Kir4.1 and Kir5.1) and TEA-sensitive IKDR (encoded by Kv3.1) are present in mouse neural sphere-derived progenitor cells[25,26].

Cai and colleagues demonstrated that multiple ion channels are heterogeneously expressed in rat embryonic neural stem cells, including IA and IKDR in > 80% of cells, INa (both TTX-sensitive and TTX-insensitive) and ICa.L in a small population (22% and 19%) of neural stem cells[27]. IKDR (encoded by Kv2.1) and IA (encoded by Kv4.3) are also detected by Smith et al[28] in rat embryonic neural progenitor cells. Multiple ion channels, i.e. TTX-sensitive INa, TEA-insensitive IKDR (likely encoded by Kv1.6, Kv2.1, and Kv2.2) and 4-AP-sensitive IA (encoded by Kv4.2 and Kv4.3), are co-expressed in progenitor cells from neonatal rat forebrain[29]. However, only IKDR encoded by Kv1.3 and Kv3.1 is present in adult rat neural progenitor cells[30]. Interestingly, 4-AP-sensitive IA (encoded by Kv4.2) and α-dendrotoxin-sensitive IKDR (likely encoded by Kv1.1, Kv1.6, and Kv3.1) are recently reported in human embryonic neural progenitor cells derived from aborted fetal brain tissue (12 weeks post-fertilization)[31]. Four types of ionic currents, IA, IKDR, IKir and INa.TTX, are also described by Lim et al[32] in human neural stem cells from aborted fetal cortex. In addition, a recent study reports that nifedipine-sensitive ICa.L is expressed in neural stem/progenitor cells from the brain cortex of postnatal mice[33]. Moreover, TRPC1 has been found to mediate growth factor receptor-induced Ca2+ entry in embryonic rat neural stem cells[34].

ION CHANNELS IN CARDIAC PROGENITOR CELLS AND iPS CELLS

In cardiac progenitor cells, a recent study demonstrated that IKDR (encoded by Kv1.1, Kv1.2 and Kv1.6), ICl.vol (encoded by Clcn3) and IKir (encoded by Kir1.1, Kir2.1, and Kir2.2) are present in adult mouse cardiac c-kit+ progenitor cells[35]. Only IKDR (likely encoded by KCNQ2) is expressed in human iPS cells[36]. More information on ion channel expression in cardiac progenitor cells and iPS cells from different species is required.

ION CHANNELS IN CANCER STEM CELLS

Although cancer stem cells have been described in different types of cancers[37,38], information regarding ion channels in cancer stem cells is limited. A recent study reported that hERG (Kv11.1) channels are expressed in CD34+/CD38-/CD123(high) leukemia stem cells but not in normal bone marrow CD34+ cells[39]. A high expression level of BKCa current has recently been recorded in CD133+ stem cells from SH-SY5Y neuroblastoma[40]. Additional information remains to be collected on ion channel expression in stem cells from different types of cancer.

ROLES OF ION CHANNELS IN REGULATING PROLIFERATION AND/OR DIFFERENTIATION OF STEM CELLS

The effect of voltage-gated K+ channels on cell mitogenesis was initially reported in human T lymphocytes by DeCoursey et al[41]. Great progress has been made in establishing the roles of specific channels in cell proliferation. K+ channels modulate the cell progression through G0/G1 and K+ channel expression changes with cell cycle progression.

Ion channels play an important role in controlling cell proliferation[42-44]. Kv channel blockade exhibits a significant anti-proliferative effect in numerous types of proliferative cells including glial cells, lymphocytes, endothelial cells, breast and prostate cancer cells[42,45]. These studies indicate that cell proliferation requires activity of K+ channels. In addition, inhibition of voltage-gated K+ channels and Na+ channels suppresses migration of gastrointestinal epithelial cells[46,47]. It is believed that Kv, KCa, Na+ and Cl- channels mediate cancer cell migration, proliferation, invasion and metastasis[48].

We recently demonstrated that IKDR is upregulated in early G1 phase while IKCa is increased in progressing G1 phase in rat bone marrow-derived MSCs. Silencing IKDR channels or IKCa channels with corresponding short interference RNAs (siRNAs) targeting Kv1.2 and Kv2.1 or KCa3.1 inhibits cell proliferation and accumulates cells at G0/G1 phase[49], suggesting that IKDR and IKCa are required for the regulation of cell proliferation in rat MSCs[49,50]. Blockade of IKDR by 4-AP or TEA remarkably reduces proliferation of mouse and human ES cells[11], human iPS cells[36] and human fat tissue-derived MSCs[23] but not mouse cardiac c-kit+ progenitor cells[35]. On the other hand, the inhibition of IKDR, e.g. Kv1.3 by psora-4 or Kv3.1 by TEA, promotes proliferation of adult rat neural progenitor cells[25,26,30]. Also the blockade of IKDR by α-dendrotoxin is found to increase proliferation of human neural progenitor cells[31].

Blockade of IKCa with the selective blocker clotrimazole or silencing IKCa channel expression with KCa3.1 siRNA also reduces cell proliferation in mouse bone marrow-derived MSCs by accumulating cells at G0/G1 phase[51]. However, this is not the case for human fat tissue-derived MSCs in which the IKCa inhibition by clotrimazole has no inhibitory effect on cell proliferation[23].

The regulatory effect of BKCa on cell proliferation is dependent on cell type and/or experimental conditions. BKCa inhibition or KCa1.1 silencing reduces cell proliferation in human preadipocytes[52]. Block of BKCa by the selective channel blocker iberiotoxin inhibits cell proliferation in human endothelial cells[53,54] and in mouse ES cells[11] but not in human fat tissue-derived MSCs[23]. We recently found (unpublished) that inhibition of BKCa with paxilline or silencing BKCa with lentiviral-based short hairpin RNA targeting KCa1.1 reduces cell proliferation in human bone marrow-derived MSCs.

The volume-sensitive Cl- channel (ICl.vol) has been implicated cell proliferation and apoptosis in a variety of cells[45,55,56]. We have found that ICl.vol inhibition by the blocker 5-nitro-1-(3-phenylpropylamino) benzoic acid (NPPB) or silencing ICl.vol channel with Clcn3 siRNA remarkably reduces cell proliferation in mouse MSCs by accumulating cells at G0/G1 phase, and the effect is mediated by suppressing cyclin D and cyclin E[51]. Similarly, block of ICl.vol channel with NPPB also decreases cell proliferation in mouse cardiac c-kit+ progenitor cells[35].

In proliferative cells, membrane hyperpolarization is implicated in silencing proliferation[54,55]. Membrane depolarization by the inhibition of IKir with Ba2+ or increase of extracellular K+ concentration has been demonstrated to promote cell proliferation in adult neural progenitor cells[25]. This is consistent with the observation in astrocytes in which transient membrane depolarization with a reduction of Kir channel activity is observed during cell cycle progression from G1/S checkpoint to S phase[42]. However, this mechanism does not seem to be applicable for rat oligodendrocyte precursor cells. KATP openers diazoxide and pinacidil stimulate proliferation of rat oligodendrocyte precursor cells which is believed to be related to membrane hyperpolarization induced by KATP[57].

Limited information is available in literature regarding the physiological role of Ito (or IA) in proliferative cells. We have recently found that inhibition of Ito by 4-AP or silencing Kv4.2 channel reduces cell proliferation in human preadipocytes[52]. Consistent with this observation, activation of IA (Kv4.2) is found to be a prerequisite for cell proliferation in human embryonic neural progenitor cells[31].

Cytosolic Ca2+ activity is crucial for stem/progenitor cell cycle progression and growth[58,59]. Ca2+ entry through L-type Ca2+ channel is found to strongly correlate with differentiation of neural progenitor cells derived from mouse brain cortex; since nifedipine reduces while Bay K 8644 enhances neural differentiation[33]. In addition, TRPC1-mediated Ca2+ entry promotes differentiation of rat embryonic neural stem cells[34]. Silencing TRPC5 but not TRPC6 with corresponding siRNA decreases differentiation in rat neural progenitor cells[60]. These results suggest that cytosolic Ca2+ regulation by L-type Ca2+ channel, TRPC1 or TRPC5 channel plays an important role as a switch between proliferation and neuronal differentiation in different types of neural progenitor cells. It is interesting to note that a recent study demonstrated that TRPM7 channel is critical for the survival of mouse bone marrow derived mesenchymal stem cells[61].

While it is well recognized that voltage-gated TTX-sensitive (INaTTX) and TTX-resistant (INaTTXR, NaV1.5) Na+ channels play a crucial role in generating action potential and conducting excitation impulse in excitable cells, the physiological function of INa is not fully understood in non-excitable and proliferative cells[16,27,62]. INa has been reported to regulate cell proliferation and migration in rat gastric epithelial cells[46,47] and human cancer cells[63,64]; however, blockade of INa by TTX does not affect cell proliferation in fat tissue-derived MSCs[23]. The effects of INa on proliferation, migration and/or differentiation remain to be studied in different types of stem cells.

CONCLUSION

Although multiple ion channels have been found to be heterogeneously present in different types of stem cells, it is not clear whether the heterogeneous expression of ion channels is due to different subpopulations of cells and/or different cell cycle phases. An effort has been made to study the relationship between ion channel expression and cell proliferation in different types of stem cells. It is generally believed that IKCa (KCa3.1) and ICl.vol (Clcn3) are required for stem cell proliferation. Inhibition of IKDR (encoded by Kv1.2, Kv1.3, Kv1.5, Kv1.6, Kv2.1, Kv3.1 or Kv10.1) reduces proliferation in ES cells and MSCs; however, blockade of some specific Kv channels, e.g. Kv1.3 by psora-4 or Kv3.1 by TEA in adult rat neural progenitor cells[30], Kv1.1, Kv1.6 and Kv3.1 by α-dendrotoxin in human neural progenitor cells[31], promotes cell proliferation. No effect on proliferation is observed with TEA or 4-AP inhibition of IKDR (Kv1.1, Kv1.2 and Kv1.6) in mouse cardiac c-kit+ progenitor cells[35]. Thus, the role of IKDR in the regulation of proliferation is cell origin- and/or phenotype-dependent. Ion channels are believed to provide the basis for generating bioelectric signals that control migration, proliferation and differentiation in a variety of types of cells[55,65]. The studies summarized in this editorial indicate that patterns and phenotypes of ion channel expression in stem cells are species-, origin- and/or tissue-specific dependent. How these differences affect the cellular functions needs a detailed investigation in different type of stem cells. Further study should be focused on the effects of ion channels on migration and differentiation of different stem cells to determine which type of ion channel is involved in regulating cell migration and/or differentiation. This information is important for the study of regenerative medicine. Additional effort is required to investigate ion channels in cancer stem cells to locate potential therapeutic targets.

Footnotes

Peer reviewer: Umberto Galderisi, PhD, Associate Professor, Department of Experimental Medicine, Second University of Naples, Via L. De Crecchio 7, 80138 Napoli, Italy

S- Editor Wang JL L- Editor Roemmele A E- Editor Ma WH

References
1.  Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663-676.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Martin GR. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc Natl Acad Sci USA. 1981;78:7634-7638.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981;292:154-156.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Becker AJ, McCulloch EA, Till JE. Cytological demonstration of the clonal nature of spleen colonies derived from transplanted mouse marrow cells. Nature. 1963;197:452-454.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Smart N, Riley PR. The stem cell movement. Circ Res. 2008;102:1155-1168.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Hanley J, Rastegarlari G, Nathwani AC. An introduction to induced pluripotent stem cells. Br J Haematol. 2010;151:16-24.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Takahashi K, Okita K, Nakagawa M, Yamanaka S. Induction of pluripotent stem cells from fibroblast cultures. Nat Protoc. 2007;2:3081-3089.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Park IH, Zhao R, West JA, Yabuuchi A, Huo H, Ince TA, Lerou PH, Lensch MW, Daley GQ. Reprogramming of human somatic cells to pluripotency with defined factors. Nature. 2008;451:141-166.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Yu J, Hu K, Smuga-Otto K, Tian S, Stewart R, Slukvin II, Thomson JA. Human induced pluripotent stem cells free of vector and transgene sequences. Science. 2009;324:797-801.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Dick JE. Stem cell concepts renew cancer research. Blood. 2008;112:4793-4807.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Wang K, Xue T, Tsang SY, Van Huizen R, Wong CW, Lai KW, Ye Z, Cheng L, Au KW, Zhang J. Electrophysiological properties of pluripotent human and mouse embryonic stem cells. Stem Cells. 2005;23:1526-1534.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Ginis I, Luo Y, Miura T, Thies S, Brandenberger R, Gerecht-Nir S, Amit M, Hoke A, Carpenter MK, Itskovitz-Eldor J. Differences between human and mouse embryonic stem cells. Dev Biol. 2004;269:360-380.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Hanna J, Cheng AW, Saha K, Kim J, Lengner CJ, Soldner F, Cassady JP, Muffat J, Carey BW, Jaenisch R. Human embryonic stem cells with biological and epigenetic characteristics similar to those of mouse ESCs. Proc Natl Acad Sci USA. 2010;107:9222-9227.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Koestenbauer S, Zech NH, Juch H, Vanderzwalmen P, Schoonjans L, Dohr G. Embryonic stem cells: similarities and differences between human and murine embryonic stem cells. Am J Reprod Immunol. 2006;55:169-180.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Heubach JF, Graf EM, Leutheuser J, Bock M, Balana B, Zahanich I, Christ T, Boxberger S, Wettwer E, Ravens U. Electrophysiological properties of human mesenchymal stem cells. J Physiol. 2004;554:659-672.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Li GR, Sun H, Deng X, Lau CP. Characterization of ionic currents in human mesenchymal stem cells from bone marrow. Stem Cells. 2005;23:371-382.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Kawano S, Otsu K, Shoji S, Yamagata K, Hiraoka M. Ca(2+) oscillations regulated by Na(+)-Ca(2+) exchanger and plasma membrane Ca(2+) pump induce fluctuations of membrane currents and potentials in human mesenchymal stem cells. Cell Calcium. 2003;34:145-156.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Tang CY, Bezanilla F, Papazian DM. Extracellular Mg(2+) modulates slow gating transitions and the opening of Drosophila ether-a-Go-Go potassium channels. J Gen Physiol. 2000;115:319-338.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Tao R, Lau CP, Tse HF, Li GR. Functional ion channels in mouse bone marrow mesenchymal stem cells. Am J Physiol Cell Physiol. 2007;293:C1561-C1567.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Li GR, Deng XL, Sun H, Chung SS, Tse HF, Lau CP. Ion channels in mesenchymal stem cells from rat bone marrow. Stem Cells. 2006;24:1519-1528.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Deng XL, Sun HY, Lau CP, Li GR. Properties of ion channels in rabbit mesenchymal stem cells from bone marrow. Biochem Biophys Res Commun. 2006;348:301-309.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Park KS, Jung KH, Kim SH, Kim KS, Choi MR, Kim Y, Chai YG. Functional expression of ion channels in mesenchymal stem cells derived from umbilical cord vein. Stem Cells. 2007;25:2044-2052.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Bai X, Ma J, Pan Z, Song YH, Freyberg S, Yan Y, Vykoukal D, Alt E. Electrophysiological properties of human adipose tissue-derived stem cells. Am J Physiol Cell Physiol. 2007;293:C1539-C1550.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Attali B, Wang N, Kolot A, Sobko A, Cherepanov V, Soliven B. Characterization of delayed rectifier Kv channels in oligodendrocytes and progenitor cells. J Neurosci. 1997;17:8234-8245.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Yasuda T, Bartlett PF, Adams DJ. K(ir) and K(v) channels regulate electrical properties and proliferation of adult neural precursor cells. Mol Cell Neurosci. 2008;37:284-297.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Yasuda T, Adams DJ. Physiological roles of ion channels in adult neural stem cells and their progeny. J Neurochem. 2010;114:946-959.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Cai J, Cheng A, Luo Y, Lu C, Mattson MP, Rao MS, Furukawa K. Membrane properties of rat embryonic multipotent neural stem cells. J Neurochem. 2004;88:212-226.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Smith DO, Rosenheimer JL, Kalil RE. Delayed rectifier and A-type potassium channels associated with Kv 2.1 and Kv 4.3 expression in embryonic rat neural progenitor cells. PLoS One. 2008;3:e1604.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Stewart RR, Zigova T, Luskin MB. Potassium currents in precursor cells isolated from the anterior subventricular zone of the neonatal rat forebrain. J Neurophysiol. 1999;81:95-102.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Liebau S, Propper C, Bockers T, Lehmann-Horn F, Storch A, Grissmer S, Wittekindt OH. Selective blockage of Kv1.3 and Kv3.1 channels increases neural progenitor cell proliferation. J Neurochem. 2006;99:426-437.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Schaarschmidt G, Wegner F, Schwarz SC, Schmidt H, Schwarz J. Characterization of voltage-gated potassium channels in human neural progenitor cells. PLoS One. 2009;4:e6168.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Lim CG, Kim SS, Suh-Kim H, Lee YD, Ahn SC. Characterization of ionic currents in human neural stem cells. Korean J Physiol Pharmacol. 2008;12:131-135.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  D’Ascenzo M, Piacentini R, Casalbore P, Budoni M, Pallini R, Azzena GB, Grassi C. Role of L-type Ca2+ channels in neural stem/progenitor cell differentiation. Eur J Neurosci. 2006;23:935-944.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Fiorio Pla A, Maric D, Brazer SC, Giacobini P, Liu X, Chang YH, Ambudkar IS, Barker JL. Canonical transient receptor potential 1 plays a role in basic fibroblast growth factor (bFGF)/FGF receptor-1-induced Ca2+ entry and embryonic rat neural stem cell proliferation. J Neurosci. 2005;25:2687-2701.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Han Y, Chen JD, Liu ZM, Zhou Y, Xia JH, Du XL, Jin MW. Functional ion channels in mouse cardiac c-kit(+) cells. Am J Physiol Cell Physiol. 2010;298:C1109-C1117.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Jiang P, Rushing SN, Kong CW, Fu J, Lieu DK, Chan CW, Deng W, Li RA. Electrophysiological properties of human induced pluripotent stem cells. Am J Physiol Cell Physiol. 2010;298:C486-C495.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Frank NY, Schatton T, Frank MH. The therapeutic promise of the cancer stem cell concept. J Clin Invest. 2010;120:41-50.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Park CY, Tseng D, Weissman IL. Cancer stem cell-directed therapies: recent data from the laboratory and clinic. Mol Ther. 2009;17:219-230.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Li H, Liu L, Guo L, Zhang J, Du W, Li X, Liu W, Chen X, Huang S. HERG K+ channel expression in CD34+/CD38-/CD123(high) cells and primary leukemia cells and analysis of its regulation in leukemia cells. Int J Hematol. 2008;87:387-392.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Park JH, Park SJ, Chung MK, Jung KH, Choi MR, Kim Y, Chai YG, Kim SJ, Park KS. High expression of large-conductance Ca2+-activated K+ channel in the CD133+ subpopulation of SH-SY5Y neuroblastoma cells. Biochem Biophys Res Commun. 2010;396:637-642.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  DeCoursey TE, Chandy KG, Gupta S, Cahalan MD. Voltage-gated K+ channels in human T lymphocytes: a role in mitogenesis? Nature. 1984;307:465-468.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  MacFarlane SN, Sontheimer H. Changes in ion channel expression accompany cell cycle progression of spinal cord astrocytes. Glia. 2000;30:39-48.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Wonderlin WF, Strobl JS. Potassium channels, proliferation and G1 progression. J Membr Biol. 1996;154:91-107.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Pardo LA. Voltage-gated potassium channels in cell proliferation. Physiology (Bethesda). 2004;19:285-292.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Lang F, Shumilina E, Ritter M, Gulbins E, Vereninov A, Huber SM. Ion channels and cell volume in regulation of cell proliferation and apoptotic cell death. Contrib Nephrol. 2006;152:142-160.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Wu WK, Li GR, Wong TM, Wang JY, Yu L, Cho CH. Involvement of voltage-gated K+ and Na+ channels in gastric epithelial cell migration. Mol Cell Biochem. 2008;308:219-226.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Wu WK, Li GR, Wong HP, Hui MK, Tai EK, Lam EK, Shin VY, Ye YN, Li P, Yang YH. Involvement of Kv1.1 and Nav1.5 in proliferation of gastric epithelial cells. J Cell Physiol. 2006;207:437-444.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Fraser SP, Pardo LA. Ion channels: functional expression and therapeutic potential in cancer. Colloquium on Ion Channels and Cancer. EMBO Rep. 2008;9:512-515.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Deng XL, Lau CP, Lai K, Cheung KF, Lau GK, Li GR. Cell cycle-dependent expression of potassium channels and cell proliferation in rat mesenchymal stem cells from bone marrow. Cell Prolif. 2007;40:656-670.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Wang SP, Wang JA, Luo RH, Cui WY, Wang H. Potassium channel currents in rat mesenchymal stem cells and their possible roles in cell proliferation. Clin Exp Pharmacol Physiol. 2008;35:1077-1084.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Tao R, Lau CP, Tse HF, Li GR. Regulation of cell proliferation by intermediate-conductance Ca2+-activated potassium and volume-sensitive chloride channels in mouse mesenchymal stem cells. Am J Physiol Cell Physiol. 2008;295:C1409-C1416.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Hu H, He ML, Tao R, Sun HY, Hu R, Zang WJ, Yuan BX, Lau CP, Tse HF, Li GR. Characterization of ion channels in human preadipocytes. J Cell Physiol. 2009;218:427-435.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Wiecha J, Munz B, Wu Y, Noll T, Tillmanns H, Waldecker B. Blockade of Ca2+-activated K+ channels inhibits proliferation of human endothelial cells induced by basic fibroblast growth factor. J Vasc Res. 1998;35:363-371.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Kuhlmann CR, Most AK, Li F, Munz BM, Schaefer CA, Walther S, Raedle-Hurst T, Waldecker B, Piper HM, Tillmanns H. Endothelin-1-induced proliferation of human endothelial cells depends on activation of K+ channels and Ca2+ influx. Acta Physiol Scand. 2005;183:161-169.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Blackiston DJ, McLaughlin KA, Levin M. Bioelectric controls of cell proliferation: ion channels, membrane voltage and the cell cycle. Cell Cycle. 2009;8:3519-3528.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Liu YJ, Wang XG, Tang YB, Chen JH, Lv XF, Zhou JG, Guan YY. Simvastatin ameliorates rat cerebrovascular remodeling during hypertension via inhibition of volume-regulated chloride channel. Hypertension. 2010;56:445-452.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Fogal B, McClaskey C, Yan S, Yan H, Rivkees SA. Diazoxide promotes oligodendrocyte precursor cell proliferation and myelination. PLoS One. 2010;5:e10906.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Resende RR, Adhikari A, da Costa JL, Lorencon E, Ladeira MS, Guatimosim S, Kihara AH, Ladeira LO. Influence of spontaneous calcium events on cell-cycle progression in embryonal carcinoma and adult stem cells. Biochim Biophys Acta. 2010;1803:246-260.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Ferreira-Martins J, Rondon-Clavo C, Tugal D, Korn JA, Rizzi R, Padin-Iruegas ME, Ottolenghi S, De AA, Urbanek K, Ide-Iwata N. Spontaneous calcium oscillations regulate human cardiac progenitor cell growth. Circ Res. 2009;105:764-774.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Shin HY, Hong YH, Jang SS, Chae HG, Paek SL, Moon HE, Kim DG, Kim J, Paek SH, Kim SJ. A role of canonical transient receptor potential 5 channel in neuronal differentiation from A2B5 neural progenitor cells. PLoS One. 2010;5:e10359.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Cheng H, Feng JM, Figueiredo ML, Zhang H, Nelson PL, Marigo V, Beck A. Transient receptor potential melastatin type 7 channel is critical for the survival of bone marrow derived mesenchymal stem cells. Stem Cells Dev. 2010;19:1393-1403.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Li GR, Sun HY, Chen JB, Zhou Y, Tse HF, Lau CP. Characterization of multiple ion channels in cultured human cardiac fibroblasts. PLoS One. 2009;4:e7307.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Gao R, Shen Y, Cai J, Lei M, Wang Z. Expression of voltage-gated sodium channel alpha subunit in human ovarian cancer. Oncol Rep. 2010;23:1293-1299.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Isbilen B, Fraser SP, Djamgoz MB. Docosahexaenoic acid (omega-3) blocks voltage-gated sodium channel activity and migration of MDA-MB-231 human breast cancer cells. Int J Biochem Cell Biol. 2006;38:2173-2182.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Sundelacruz S, Levin M, Kaplan DL. Role of membrane potential in the regulation of cell proliferation and differentiation. Stem Cell Rev. 2009;5:231-246.  [PubMed]  [DOI]  [Cited in This Article: ]