当前位置: 首页 > 期刊 > 《干细胞学杂志》 > 2005年第10期 > 正文
编号:11357738
Electrophysiological Properties of Pluripotent Human and Mouse Embryonic Stem Cells
http://www.100md.com 《干细胞学杂志》
     a Department of Medicine, University of Hong Kong, Hong Kong, China;

    b Departments of Medicine and

    c Cellular and

    d Molecular Medicine and Institute for Cell Engineering, Johns Hopkins University, Baltimore, Maryland, USA

    Key Words. Embryonic stem cells ? Electrophysiology ? Pluripotency ? Ion channels

    Correspondence: Ronald Li, Ph.D., Johns Hopkins University, 720 Rutland Avenue, Ross 1165, Baltimore, Maryland 21205, USA. Telephone: 410-614-0035; Fax: 410-502-2802; e-mail: ronaldli@jhmi.edu; and Hung-Fat Tse, M.D., University of Hong Kong, Cardiology Division, Queen Mary Hospital, Pokfulam, Hong Kong. e-mail: hftse@hkucc.hku.hk

    ABSTRACT

    Embryonic stem cells (ESCs) are derived from the inner cell mass of blastocysts. Because ESCs can propagate indefinitely in culture while maintaining their pluripotency to differentiate into all cell types, they may therefore provide an unlimited supply of specialized cells such as cardiomyocytes and neurons for cell-based therapies. For instance, direct injection of pluripotent ESCs after myocardial infarction has been suggested as a means to repair the damaged heart . However, transplantation of cells with undesirable electrical properties into the heart can predispose patients to lethal electrical disorders (arrhythmias) . Therefore, it is critical to understand the electrophysiological profile of undifferentiated ESCs, which has not been characterized. In this study, we hypothesize that ion channels are functionally expressed in mouse (m) and human (h) ESCs, although the specific encoding genes (and/or their isoforms) and their expression levels might differ. We discovered that several specialized ion channels are differentially expressed in pluripotent mESCs and hESCs. Collectively, our experiments reveal further similarities and differences between the two species. We discuss these results in relation to the physiological function of ion channels in hESC biology as well as practical considerations for potential therapeutic applications of hESCs.

    MATERIALS AND METHODS

    Ionic Currents in Pluripotent mESCs

    Figure 1A shows that undifferentiated mESC colonies were homogenously immunostained for the pluripotency markers Oct-4 and SSEA-1 . In 159 of 304 (52.3%) undifferentiated mESCs, depolarization-activated time-dependent noninactivating outward currents that increased progressively with positive voltages could be recorded (8.6 ± 0.9 pA/pF at +40 mV; Figs. 1B, 1C). These outwardly rectifying currents resemble the delayed-rectifier K+ currents (IKDR) and could be dose-dependently inhibited by the known K+ channel blocker TEA+ (IC50 = 1.2 ± 0.3 mM, n = 13; Figs. 1B, 1D). IKDR in mESCs was also sensitive to 4-AP (IC50 = 0.5 ± 0.1 mM, n = 17), a more potent K channel blocker than TEA+ , and the Ca2+-activated large-conductance K+ current (IKCa) blocker IBTX (100nM) (current inhibition = 33.2% ± 12.7%, n = 3) (Figs. 1B, 1E). As shown in Figures 1D and 1E, increasing TEA or 4-AP to 30 mM could not lead to complete current inhibition. Indeed, even combined application of 30 mM TEA and 30 mM 4-AP also could not completely eliminate the IKDR (30 mM 4-AP, 30 mM TEA, and 30 mM TEA + 30 mM 4-AP reduced the IKDR to 45.8% ± 2.1%, 39.3% ± 7.6%, and 45.1% ± 8.1%, respectively; p > .05). The currents remaining after combined TEA/4-AP blockade were also not sensitive to 1 mM BaCl2, implicating the presence of some background current.

    Figure 1. (A): Images of pluripotent mESCs immunostained for SSEA-1 and Oct-4. (B): Representative current tracings recorded from undifferentiated mESCs before (left panels) and after (right panels) blockade by TEA, 4-AP, and IBTX, as indicated. The electrophysiological protocol used for eliciting currents is also given. (C): Current-voltage relationship of IKDR of mESCs. Dose–response relationships for (D) TEA and (E) 4-AP block of IKDR of mESCs. Abbreviations: 4-AP, 4-aminopyridine; IBTX, iberiotoxin; mESC, mouse embryonic stem cell; TEA, tetraethylammonium.

    Although voltage-gated Na+ (Nav) and Ca2+ (Cav) currents were completely absent in all pluripotent mESCs tested (n > 200), whether IKDR was present (Fig. 1B) or not (Fig. 2A), a modest yet detectable hyperpolarization-activated inward current (Ih, encoded by the hyperpolarization-activated cyclic nucleotide-modulated nonselective or HCN ion channel family ; –2.2 ± 0.4 pA/pF at –120 mV) was detected in 79 of 270 cells (29.3%; Fig. 2B). Ih in mESCs was reversibly blocked by the HCN inhibitor Cs+ . Application of 1 μM isoproterenol altered neither the kinetics nor amplitude of Ih recorded in mESCs. Inwardly rectifying K currents (IK1) responsible for stabilizing the resting membrane potential were also not present.

    Figure 2. (A): Stimulation protocol and representative current traces demonstrating the absence of Nav or Cav currents in an mESC that lacks IKDR. (B): CsCl2-sensitive hyperpolarization-activated currents could be recorded from pluripotent mESCs but not hESCs. (C): Steady-state current-voltage relationships of hyperpolarization-activated currents of mESCs and hESCs. Abbreviations: hESC, human embryonic stem cell; mESC, mouse embryonic stem cell.

    To obtain insights into the molecular identities of the ionic currents identified, total RNA was isolated from pluripotent mESCs for RT-PCR. Figure 3A shows that Kv1.1, 1.2, 1.3, 1.4, 1.6, 4.2, and BK (or Maxi-K) transcripts but not Kv1.5, 2.1, 3.1, 3.2, and 4.3 were detected. Consistent with the presence of Ih, HCN2 and HCN3 transcripts were also expressed. Inhibition by 4-AP and insensitivity to extracellular TEA ions is a pharmacological hallmark of A-type currents . However, neither TEA-subtracted nor 4-AP–subtracted current traces from IKDR blockade revealed any transient outward current with the typical rapid inactivation feature (data not shown). Therefore, although Kv1.1, 1.2, 1.6, and BK channels might underline the delayed rectifier current recorded, we conclude that Kv1.4- and Kv4.2-encoded transient outward K+ currents were not functionally expressed.

    Figure 3. (A): Expression of ion channel transcripts in mESCs probed by semiquantitative reverse transcription–polymerase chain reaction. Dose–response relationships of (B) TEA, (C) 4-AP, and (D) IBTX for inhibition of mESC proliferation assessed by BrdU incorporation (open squares) and cytotoxic effects by MTT (solid squares). Abbreviations: 4-AP, 4-aminopyridine; BrdU, bromodeoxyurindine; IBTX, iberiotoxin; mESC, mouse embryonic stem cell; TEA, tetra-ethylammonium.

    Effects of Ion Channel Blockers

    To investigate possible physiological roles of the ionic currents identified, we next studied the functional consequences of their pharmacological blockade by assessing the effects of extracellular application of K+ channel blockers on cell proliferation. Specifically, we measured DNA synthesis as an index for replication by quantifying BrdU incorporation into genomic DNA during the S phase of the cell cycle, which is proportional to the rate of cell division . Application of TEA+ significantly inhibited the proliferation of mESCs in a dose-dependent manner (Fig. 3B, open squares). The EC50 was 20.1 ± 3.7 mM (n = 3), approximately 20-fold higher than the IC50 for IKDR inhibition. Similarly, 4-AP (EC50 = 2.7 ± 0.2 mM; Fig. 3C, open squares) and IBTX (EC50 = 133.9 ± 25.9 nM; Fig. 3D, open squares) also dose-dependently reduced cell proliferation. Of note, the rank orders of these agents to inhibit proliferation follow the trend of their potencies to block IKDR (i.e., IBTX > 4-AP > TEA). To assess the cytotoxic effect of K+ channel blockers, a colorimetric MTT kit was used to examine for changes in metabolism. Notably, the EC50 values for inhibition of metabolic activity by TEA (62.7 ± 9.0 mM; Fig. 3B, solid squares), 4-AP (4.6 ± 0.5 mM; Fig. 3C, solid squares), and IBTX (333.9 ± 64.6 nM; Fig. 3D, solid squares) were significantly higher than those for inhibiting cell proliferation (p < .05). These results implicate that the metabolic influences of these blockers were relatively insignificant at concentrations at which they effectively exert their inhibitory effects on ESC proliferation, presumably by K+ channel blockade.

    Electrophysiological Properties of hESCs: Similarities and Differences

    Although hESCs and mESCs share several similarities, significant differences are known to exist between the two species . Therefore, we also examined the previously unexplored electrophysiological properties of hESCs. Pluripotent hESCs were positive for markers such as alkaline phosphatase, Oct4, SSEA4, and TRA-60 (Fig. 4A), consistent with previous reports . Similar to mESCs, TEA+-sensitive IKDR (IC50 = 2.1 ± 0.2 mM; Fig. 4D) was also detected in hESCs (~100%), but the current density was approximately sixfold higher (47.5 ± 7.9 pA/pF at +40 mV, n = 12, p < .05) (Figs. 4B, 4C). Similar to mESCs, application of TEA+ dose-dependently inhibited hESC proliferation as assayed by BrdU incorporation, with an EC50 (11.6 ± 2.0 mM) (Fig. 4D). Unlike mESCs, however, there was no measurable Ih in all hESCs tested (n = 30; Fig. 2B). Same as mESCs, neither Nav nor Cav currents could be detected in hESCs.

    Figure 4. (A): Pluripotent hESCs were positive for alkaline phosphatase, SSEA-4, and TRA-1-60. (B): Representative current tracings recorded from undifferentiated hESCs. The same electrophysiological from Figure 1 was used. (C): Steady-state current-voltage relationship of IKDR in hESCs. (D): IC50 for blockade of IKDR and EC50 for proliferation inhibition by tetraethylammonium in hESCs. Abbreviation: hESC, human embryonic stem cell.

    Microarray Analysis of Ion Channel Genes in hESCs

    Using Affymetrix U133A chips, we performed microarray analysis of pluripotent hESCs to examine the expression of ion channels at the transcriptomic level. Figure 5A shows the expression profile of all genes tested in undifferentiated hESCs. For voltage-gated ion channels, a total of 36, 19, and 49 genes on the U133A chips were identified as Cav, Nav, or Kv channel genes, respectively. For inspection, their expression profile is extracted in Figure 5A and further summarized in Figures 5B and 5C by normalizing signals to the average expression level of the entire microarray in a manner similar to that of cytokines and their receptors in hESCs, as recently reported by Dvash et al. . Our data indicate that among the total 104 voltage-gated ion channel genes mentioned above, only the transcripts of 3, 1, and 5 Cav, Nav, and Kv genes were significantly expressed, as defined by Affymetrix. The corresponding gene products were CACNA1A (Cav2.1), CACNA2D2 (Cav 2/ subunit 2), CACNB3 (Cav ?3 subunit), SCN11A (Nav1.9), KCNB1 (Kv2.1), KCND2 (Kv4.2), KCNQ2 (Kv7.2), KCNS3 (Kv9.3), and KCNH2 (Kv11.1) (note, however, that ICa, INa, and Kv4.2-encoded transient outward K+ currents could not be electrophysiologically recorded, like mESCs). Of note, KCNQ2 and KCNH2, which underlie the noninactivating, slowly deactivating M-current and the rapid component of the cardiac delayed rectifier (IKr) , were relatively highly expressed. Similarly, KCNB1 and KCNS3, which encode for the delayed rectifier Kv2.1 channels and the silent modulatory -subunit Kv9.3 that heteromerizes with Kv2.1 subunits , respectively, were also expressed. Collectively, these ion channel genes could underlie the KDR current identified, although further experiments will be needed to confirm and dissect their molecular identities. By contrast, no HCN transcript was expressed in pluripotent hESCs. RT-PCR confirmed the array results for five of nine channels (Fig. 5D).

    Figure 5. (A): Microarray analysis of pluripotent hESCs for the transcript expression of all genes tested using Affymetrix U133A microarrays (see Materials and Methods). (B): Left, 104 voltage-gated Cav, Nav, and Kv channel genes are clustered. The same expression scale bar shown in (A) was used. Right, same as the left panel, except only transcripts that were defined to be expressed, as defined by Affymetrix, are shown. (C): Bar graph of normalized transcript levels of the expressed ion channel genes. Data were normalized to the expression level of the 50th percentile of the entire microarray. (D): Expression of ion channel transcripts in hESCs probed by semiquantitative reverse transcription–polymerase chain reaction. Abbreviation: hESC, human embryonic stem cell.

    DISCUSSION

    K.W. and T.X. contributed equally to this study. This work was supported by grants from the National Institutes of Health (R01 HL-52768 and R01 HL-72857 to R.A.L.), the Blaustein Pain Research Centre (to R.A.L.), and the Hong Kong Research Grant Council (HKU 7459/04M to H.F.T., R.A.L., G.R.L., and C.P.L.). S.Y.T. was supported by a postdoctoral fellowship award from the Croucher Foundation. We would also like to thank Dr. Guibin Chen at Johns Hopkins for helpful discussion in microarray analysis.

    DISCLOSURES

    The authors indicate no potential conflicts of interest.

    REFERENCES

    Hodgson DM, Behfar A, Zingman LV et al. Stable benefit of embryonic stem cell therapy in myocardial infarction. Am J Physiol Heart Circ Physiol 2004;287:H471–H479.

    Menasche P, Hagege AA, Vilquin JT et al. Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction. J Am Coll Cardiol 2003;41:1078–1083.

    Smits PC, van Geuns RJ, Poldermans D et al. Catheter-based intramyocardial injection of autologous skeletal myoblasts as a primary treatment of ischemic heart failure: clinical experience with six-month follow-up. J Am Coll Cardiol 2003;42:2063–2069.

    Nagy A, Rossant J, Nagy R et al. Derivation of completely cell culture-derived mice from early-passage embryonic stem cells. Proc Natl Acad Sci U S A 1993;90:8424–8428.

    Doetschman TC, Eistetter H, Katz M et al. The in vitro development of blastocyst-derived embryonic stem cell lines: formation of visceral yolk sac, blood islands and myocardium. J Embryol Exp Morphol 1985;87: 27–45.

    Thomson JA, Itskovitz-Eldor J, Shapiro SS et al. Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145–1147.

    Xue T, Cho HC, Akar FG et al. Functional integration of electrically active cardiac derivatives from genetically engineered human embryonic stem cells with quiescent recipient ventricular cardiomyocytes: insights into the development of cell-based pacemakers. Circulation 2005;111:11–20.

    Moore JC, van Laake LW, Braam SR et al. Human embryonic stem cells: genetic manipulation on the way to cardiac cell therapies. Reprod Toxicol 2005;20:377–391.

    Trono D. Lentiviral Vectors. New York: Spring-Verlag, 2002.

    Zufferey R, Dull T, Mandel RJ et al. Self-inactivating lentivirus vector for safe and efficient in vivo gene delivery. J Virol 1998;72:9873–9880.

    Moore JC, van Laake LW, Braahm SR et al. Human embryonic stem cells: genetic manipulation on the way to cardiac cell therapies. Reprod Toxicol 2005;20:377–391.

    Solter D, Knowles BB. Monoclonal antibody defining a stage-specific mouse embryonic antigen (ssea-1). Proc Natl Acad Sci U S A 1978;75:5565–5569.

    Niwa H, Miyazaki J, Smith AG. Quantitative expression of oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet 2000;24:372–376.

    Hille B. The selective inhibition of delayed potassium currents in nerve by tetraethylammonium ion. J Gen Physiol 1967;50:1287–1302.

    Kirsch GE, Taglialatela M, Brown AM. Internal and external tea block in single cloned K+ channels. Am J Physiol 1991;261:C583–C590.

    Mathie A, Wooltorton JRA, Watkins CS. Voltage-activated potassium channels in mammalian neurons and their block by novel pharmacological agents. Gen Pharmacol 1998;30:13–24.

    Baker M, Howe JR, Ritchie JM. Two types of 4-aminopyridine-sensitive potassium current in rabbit schwann cells. J Physiol 1993;464:321–342.

    Pape H-C. Queer current and pacemaker: the hyperpolarization-activated cation current in neurons. Annu Rev Physiol 1996;58:299–327.

    Champigny G, Bois P, Lenfant J. Characterization of the ionic mechanism responsible for the hyperpolarization-activated current in frog sinus venosus. Pflugers Arch 1987;410:159–164.

    McCormick DA, Pape HC. Properties of a hyperpolarization-activated cation current and its role in rhythmic oscillation in thalamic relay neurones. J Physiol 1990;431:291–318.

    Amberg GC, Koh SD, Imaizumi Y et al. A-type potassium currents in smooth muscle. Am J Physiol Cell Physiol 2003;284:C583–C595.

    Yu CC, Woods AL, Levison DA. The assessment of cellular proliferation by immunohistochemistry: a review of currently available methods and their applications. Histochem J 1992;24:121–131.

    Boheler KR, Czyz J, Tweedie D et al. Differentiation of pluripotent embryonic stem cells into cardiomyocytes. Circ Res 2002;91:189–201.

    Dvash T, Mayshar Y, Darr H et al. Temporal gene expression during differentiation of human embryonic stem cells and embryoid bodies. Hum Reprod 2004;19:2875–2883.

    Cooper EC, Jan LY. M-channels: neurological diseases, neuromodulation, and drug development. Arch Neurol 2003;60:496–500.

    Pond AL, Nerbonne JM. Erg proteins and functional cardiac I(kr) channels in rat, mouse, and human heart. Trends Cardiovasc Med 2001;11:286–294.

    Kerschensteiner D, Monje F, Stocker M. Structural determinants of the regulation of the voltage-gated potassium channel kv2.1 by the modulatory alpha-subunit kv9.3. J Biol Chem 2003;278:18154–18161.

    Kerschensteiner D, Stocker M. Heteromeric assembly of kv2.1 with kv9.3: effect on the state dependence of inactivation. Biophys J 1999;77:248–257.

    Stocker M, Kerschensteiner D. Cloning and tissue distribution of two new potassium channel alpha-subunits from rat brain. Biochem Biophys Res Commun 1998;248:927–934.

    Heubach JF, Graf EM, Leutheuser J et al. Electrophysiological properties of human mesenchymal stem cells. J Physiol 2004;554:659–672.

    Xue T, Marban E, Li RA. Dominant-negative suppression of hcn1- and hcn2-encoded pacemaker currents by an engineered hcn1 construct: Insights into structure-function relationships and multimerization. Circ Res 2002;90:1267–1273.

    Parihar AS, Coghlan MJ, Gopalakrishnan M et al. Effects of intermediate-conductance Ca2+-activated k+ channel modulators on human prostate cancer cell proliferation. Eur J Pharmacol 2003;471:157–164.

    Vaur S, Bresson-Bepoldin L, Dufy B et al. Potassium channel inhibition reduces cell proliferation in the gh3 pituitary cell line. J Cell Physiol 1998;177:402–410.

    Wonderlin WF, Strobl JS. Potassium channels, proliferation and g1 progression. J Membr Biol 1996;154:91–107.

    Platoshyn O, Golovina VA, Bailey CL et al. Sustained membrane depolarization and pulmonary artery smooth muscle cell proliferation. Am J Physiol Cell Physiol 2000;279:C1540–C1549.

    Vautier F, Belachew S, Chittajallu R et al. Shaker-type potassium channel subunits differentially control oligodendrocyte progenitor proliferation. Glia 2004;48:337–345.

    van Kempen MJA, van Ginneken A, de Grijs I et al. Expression of the electrophysiological system during murine embryonic stem cell cardiac differentiation. Cell Physiol Biochem 2003;13:263–270.

    van der Heyden MA, van Kempen MJ, Tsuji Y et al. P19 embryonal carcinoma cells: a suitable model system for cardiac electrophysiological differentiation at the molecular and functional level. Cardiovasc Res 2003;58:410–422.

    Mummery C, Ward-van Oostwaard D, Doevendans P et al. Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells. Circulation 2003;107:2733–2740.

    Valiunas V, Doronin S, Valiuniene L et al. Human mesenchymal stem cells make cardiac connexins and form functional gap junctions. J Physiol 2004;555:617–626.

    Sohl G, Theis M, Hallas G et al. A new alternatively spliced transcript of the mouse connexin32 gene is expressed in embryonic stem cells, oocytes, and liver. Exp Cell Res 2001;266:177–186.

    Oyamada Y, Komatsu K, Kimura H et al. Differential regulation of gap junction protein (connexin) genes during cardiomyocytic differentiation of mouse embryonic stem cells in vitro. Exp Cell Res 1996;229:318–326.

    Satin J, Kehat I, Caspi O et al. Mechanism of spontaneous excitability in human embryonic stem cell derived cardiomyocytes. J Physiol 2004;559:479–496.

    Kehat I, Kenyagin-Karsenti D, Snir M et al. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest 2001;108:407–414.

    He JQ, Ma Y, Lee Y et al. Human embryonic stem cells develop into multiple types of cardiac myocytes: action potential characterization. Circ Res 2003;93:32–39.

    Stieber J, Herrmann S, Feil S et al. The hyperpolarization-activated channel HCN4 is required for the generation of pacemaker action potentials in the embryonic heart. Proc Natl Acad Sci U S A 2003;100:15235–15240.

    Smart SL, Bosma MM, Tempel BL. Identification of the delayed rectifier potassium channel, Kv1.6, in cultured astrocytes. Glia 1997;20:127–134.

    Schilling T, Quandt FN, Cherny VV et al. Upregulation of Kv1.3 K(+) channels in microglia deactivated by TGF-beta. Am J Physiol Cell Physiol 2000;279:C1123–C1134.

    Koh JT, Jeong BC, Kim JH et al. Changes underlying arrhythmia in the transgenic heart overexpressing Refsum disease gene-associated protein. Biochem Biophys Res Commun 2004;313:156–162.

    Amberg GC, Koh SD, Hatton WJ et al. Contribution of Kv4 channels toward the A-type potassium current in murine colonic myocytes. J Physiol 2002;544:403–415.

    Eghbali M, Toro L, Stefani E. Diminished surface clustering and increased perinuclear accumulation of large conductance Ca2+-activated K+ channel in mouse myometrium with pregnancy. J Biol Chem 2003;278:45311–45317.

    Platoshyn O, Remillard CV, Fantozzi I et al. Diversity of voltage-dependent k+ channels in human pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2004;287:L226–L238.(Kai Wanga, Tian Xueb, Suk)