ArticlePDF Available

Functional expression of Ca2+ signaling pathways in mouse embryonic stem cells

Authors:

Abstract

Mouse embryonic stem (mES) cells have the potential to differentiate into all types of cells, but the physiological properties of undifferentiated mES cells, including Ca2+ signaling systems, are not fully understood. In this study, we investigated Ca2+ signaling pathways in mES cells by using confocal Ca2+ imaging systems, patch clamp techniques and RT-PCR. The stimulations with ATP and histamine (His) induced a transient increase of intracellular Ca2+ concentration ([Ca2+]i), which were prevented by the pretreatment of 2-amino-ethoxydiphenyl borate (2-APB), a blocker for inositol-1,4,5-triphosphate receptors (InsP3Rs). The application of caffeine (Caff) or ryanodine (Ry) did not change [Ca2+]i. When stores were depleted with Ca2+ -ATPase blocker, thapsigargin (TG), or histamine, the capacitative Ca2+ entry (CCE) was observed. In whole cell patch clamp mode, store-operated Ca2+ currents could be recorded in cells treated with histamine and thapsigargin. On the other hand, voltage-operated Ca2+ channels (VOCCs) could not be elicited. The application of blockers for plasma membrane Ca2+ pump (PMCAs) (carboxeosin or caloxin2A1) induced a large increase of [Ca2+]i. When the Na+/Ca2+ exchangers (NCXs) were blocked by Na+ free solution or KBR7943, [Ca2+]i was also elevated. Using RT-PCR, mRNAs for InsP3Rs type-1, -2, and -3, PMCA-1 and -4, NCX-1, -2, and -3 could be detected. From these results, we conclude that Ca2+ release from ER is mediated by InsP3Rs in mES cells before differentiation and Ca2+ entry through plasma membrane is mainly mediated by the store-operated Ca2+ channels (SOCs). For the Ca2+ extrusion systems, both NCXs and PMCAs play important roles for maintaining the low level of [Ca2+]i.
Cell Calcium 36 (2004) 135–146
Functional expression of Ca2+signaling pathways in
mouse embryonic stem cells
Eri Yanagidaa,b,1, Satoshi Shojic,1, Yoshiyuki Hirayamaa,d, Fumio Yoshikawac,
Keishi Otsua,b, Hiroshi Uematsub, Masayasu Hiraokaa,
Teiichi Furuichic, Seiko Kawanoa,
aDepartment of Cardiovascular Diseases, Medical Research Institute, Tokyo Medical and Dental University,
1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
bDepartment of Gerodontolgy, Graduated School, Tokyo Medical and Dental University, Tokyo, Japan
cBrain Science Institute, RIKEN, Wako, Japan
dFirst Department of Medicine, Nippon Medical School, Tokyo, Japan
Received 20 September 2003; received in revised form 21 December 2003; accepted 16 January 2004
Abstract
Mouse embryonic stem (mES) cells have the potential to differentiate into all types of cells, but the physiological properties of undif-
ferentiated mES cells, including Ca2+signaling systems, are not fully understood. In this study, we investigated Ca2+signaling pathways
in mES cells by using conforcal Ca2+imaging systems, patch clamp techniques and RT-PCR. The stimulations with ATP and his-
tamine (His) induced a transient increase of intracellular Ca2+concentration ([Ca2+]i), which were prevented by the pretreatment of
2-amino-ethoxydiphenyl borate (2-APB), a blocker for inositol-1,4,5-triphosphate receptors (InsP3Rs). The application of caffeine (Caff)
or ryanodine (Ry) did not change [Ca2+]i. When stores were depleted with Ca2+-ATPase blocker, thapsigargin (TG), or histamine, the ca-
pacitative Ca2+entry (CCE) was observed. In whole cell patch clamp mode, store-operated Ca2+currents could be recorded in cells treated
with histamine and thapsigargin. On the other hand, voltage-operated Ca2+channels (VOCCs) could not be elicited. The application of
blockersforplasmamembraneCa2+pump(PMCAs)(carboxeosinorcaloxin2A1)induceda large increase of [Ca2+]i. When the Na+/Ca2+
exchangers(NCXs) were blockedby Na+free solution orKBR7943, [Ca2+]iwas alsoelevated.UsingRT-PCR,mRNAsfor InsP3Rstype-1,
-2, and -3, PMCA-1 and -4, NCX-1, -2, and -3 could be detected. From these results, we conclude that Ca2+release from ER is mediated
by InsP3Rs in mES cells before differentiation and Ca2+entry through plasma membrane is mainly mediated by the store-operated Ca2+
channels (SOCs). For the Ca2+extrusion systems, both NCXs and PMCAs play important roles for maintaining the low level of [Ca2+]i.
© 2004 Elsevier Ltd. All rights reserved.
Keywords: Mouse ES cell; Ca2+; InsP3receptors; Store-operated Ca2+channels; Na+/Ca2+exchange; Plasma membrane Ca2+pump; Patch clamp
1. Introduction
Embryonic stem (mES) cells have the potential to dif-
ferentiate into all types of the cells [1]. Recently, the
development of research activity on ES cells is rapidly
progressing in the areas of both the basic science and
clinical medicine [1,2]. ES cells have been established as
permanent lines of undifferentiated pluripotent cells from
early mouse embryos [3]. During differentiation or pro-
liferation, many processes are modulated by varieties of
Corresponding author. Tel.: +81-3-5803-5832; fax: +81-3-5684-6295.
E-mail address: seiko.card@mri.tmd.ac.jp (S. Kawano).
1Both authors equally contributed to this work.
hormones, cytokines, and genes [4]. Calcium ion (Ca2+)
is known to play an important role for the differentiation
and proliferation as a biological signal [5,6]. Nevertheless,
the physiological Ca2+signaling systems in mES cells are
not fully characterized, including Ca2+stores, Ca2+entry
systems, or Ca2+extrusion systems. In most cells, two
main Ca2+sources, Ca2+release from internal stores and
Ca2+entry across the plasma membrane are utilized for
generating signals [5,6]. In internal stores, two function-
ally different Ca2+release channels have been identified,
namely inositol-1,4,5-trisphosphate receptors (InsP3Rs) and
ryanodine receptors (RyRs) [5,7]. On the other hand, two
distinct Ca2+entry pathways across the plasma membrane
are recognized, voltage-operated Ca2+channels (VOCCs)
0143-4160/$ – see front matter © 2004 Elsevier Ltd. All rights reserved.
doi:10.1016/j.ceca.2004.01.022
136 E. Yanagida et al. / Cell Calcium 36 (2004) 135–146
and store-operated Ca2+channels (SOCs) [8]. In most of
non-excitable cells examined, the existence of capacitative
Ca2+entry (CCE) has been demonstrated [9]. Before dif-
ferentiation, mES cells belong to non-excitable ones, but
less is known about Ca2+entry pathways, as to whether
VOCCs or SOCs function. In addition, it has not been well
understood what kinds of Ca2+extrusion systems are work-
ing to maintain the low level of Ca2+concentration in the
cytosole.
In the present study, we used mES cells (D3 cell line) for
the functional studies and investigated the types and func-
tions of Ca2+channels and transporters in plasma membrane
and internal stores. To achieve this, we performed Ca2+
imagings and patch clamp experiments and the molecular
studies.
The results indicate that the stimulation of G-protein
coupled receptors (GPCRs) induces Ca2+release from ER
mediated via InsP3Rs but ryanodine receptors do not func-
tion in mES cells. Ca2+entry through plasma membrane is
mainly mediated by SOCs but not by VOCCs. Both plasma
membrane Ca2+-ATPase (PMCA) and Na+/Ca2+ex-
changer (NCX) contribute to the extrusion of Ca2+from the
cytosole.
2. Experimental procedures
2.1. Cell cultures
Pluripotent mouse embryonic stem cells (ES-D3) were
purchased from America Type Culture Collection (ATCC)
(Manassas, VA, USA). Cells were maintained in the un-
differentiated state by culturing onto feeder layers of my-
tomycin C treated STO cells in Vitacell Dulbecco’s Mod-
ified Eagle’s Medium containing 10% fetal bovine serum,
0.1mM 2-mercaptoethanol at 37C in a humidified atmo-
sphere of 95% air, 5% CO2. The stem cell growth medium
also contained leukemia inhibitory factor (LIF) (1000U/ml)
to maintain the undifferentiated state [10]. A marker for un-
differentiating, OCT-4 gene was detected in mES cells in
this study (Fig. 3A)[11]. Therefore, we used these cells as
undifferentiated ones in each experiment. Murine leukemia
inhibitory factor (ESGRO) was purchased from Chemicon
International (Temecula, CA, USA). For PCR experiments,
we used ES cells before two passages to minimize the con-
tamination of spontaneous differentiated cells.
2.2. [Ca2+]imeasurements
The intracellular free Ca2+concentration was monitored
in mES cells by using confocal microscopy (Olympus
FV500; Tokyo, Japan). The cells were loaded with 20 M
fluo-3-AM for 30min at 37C. Standard bath solution con-
tained (in mM): NaCl, 140; KCl, 5.0; MgCl2, 2; glucose,
5; HEPES, 10; CaCl2, 0, 2 or 4mM. pH was adjusted to
7.3 by NaOH. Fluo-3 was excited at 488nm and emission
was detected at >505nm. Fluo-3-AM was purchased from
Molecular Probes Inc. (Eugene, OR, USA).
2.3. Electrophysiology
Patch clamp experiments were performed as reported pre-
viously [12,13]. Briefly, using a patch-clamp amplifier (Ax-
opatch 2A and pCLAMP8, Axon Instruments, Foster City,
CA, USA), whole cell membrane currents were recorded.
Recording electrodes were made from borosilicate glass,
coated with Sylgard (Dow Corning Corp., Midland, MI,
USA) and fire polished to a resistance of 7–12M, when
filled with internal pipette solutions. A 3 M KCl–agar bridge
connected a reference electrode, containing the same solu-
tion as the patch electrode, to the bath solution were nulled
before forming a gigaseal. Data were stored on the hard disk
digitized at 10kHz and low-pass filtered at 1kHz by a filter
with Bessel characteristics (octave attenuation, 48dB) and
analyzed off-line on a computer (VZ-6000, Epson, Tokyo,
Japan). All experiments were done at 22C. The input re-
sistance and membrane capacity were always checked at the
beginning and end of experiments. We have omitted the data
where the clamp was inadequate and membrane resistance
or capacity changed during experiments.
2.3.1. Solutions
For patch clamp experiments to record membrane cur-
rents and potentials, we used the standard bath solution
same as in [Ca2+]iimaging experiments. Standard pipette
solution contains (in mM): KCl, 120; K2ATP, 5; K2CP, 5;
MgCl2, 5; HEPES, 5; EGTA, 1; CaCl20.05; pH was ad-
justed to 7.3 by Tris–OH. The bath solution for recording
store-operated Ca2+currents contained (in mM): CaCl2,
110; CsCl, 4; MgCl2, 1; glucose, 10; HEPES, 10; EDTA,
2. pH was adjusted to 7.4 by CsOH. For recording the ion
selectivity, 110 mM CaCl2in the bath solution was replaced
with equimolar BaCl2or SrCl2. Internal solution contained
(in mM): Cs-aspartate, 140; MgCl2, 1; CaCl2, 1; EGTA, 10;
HEPES, 10. pH was adjusted to 7.2 by CsOH. Free ionized
Ca2+was calculated to be 0.018M according to a pro-
gram by Fabiato [14]. We chose the above ionic conditions
to isolate Ca2+currents from other currents. Outward K+
currents were eliminated by internal Cs+and inward Cl
currents were minimized by internal aspartate.
2.4. Drugs
Caffeine (Caff) was purchased from Wako, Inc. (Wako,
Osaka, Japan), acetylcholine (ACh), thapsigargin (TG), his-
tamine (His) and LaCl3were purchased from Sigma Chemi-
cal Co. (St. Louis, MO, USA). 2-Amino-ethoxydiphenyl bo-
rate (2-APB) was a gift from Dr. Mikoshiba (Tokyo Univer-
sity, Tokyo, Japan). Carboxyeosin diacetate was purchased
from Molecular Probes Inc. Caloxin2A1 (H–VSNSNWPSF-
PSSGGG–NH2) was synthesized [15]. KBR7943 was a gift
from Dr. Iwamoto (Fukuoka University, Fukuoka, Japan).
E. Yanagida et al. / Cell Calcium 36 (2004) 135–146 137
2.5. Reverse transcription-PCR
Total RNA was extracted from cultured mES cells us-
ing the RNeasy kit (QIAGEN), and treated with RQ1
RNase-free DNase (Promega) followed by the incubation at
65C for 15 min to obtain DNA-free total RNA. First-strand
cDNA was synthesized from 10g of the RNA using Su-
perScript II reverse transcriptase (Invitrogen) and random
hexamers. PCR was performed using an ExTaq polymerase
kit (Takara Shuzo) and primer sets listed in the Table 1.
The thermal cycling conditions were as follows: 95C for
Table 1
Gene References Primer Sequences (53)
IP3R-1 [44] Forward AGTTTGGCCAACGATTTCCTG
Reverse GTTGACATTCATGTGAGGAGG
IP3R-2 [44] Forward GTTCCCACTATGACCTTAACT
Reverse ATGGTTCTCATGGGGTGTGTT
IP3R-3 [44] Forward CCACACGGAGCTGCCACATTT
Reverse TCAGCGGCTCATGCAGTTCTG
OCT-4 [40] Forward GAGGAAGCCGACAACAATGAGAACCTTCAG
Reverse TTCTGGCGCCGGTTACAGAACCATACTCGA
MTRP-1 [43] Forward CAAGATTTTGGGAAATTTCTGG
Reverse TTTATCCTCATGATTTGCTAT
MTRP-2 [41] Forward GAGATCTAGATCCGGTTCATGTTCATCCT
Reverse GTTCGAATTCGAGCGAGCAAACTTCCACTC
MTRP-3 [43] Forward TGACTTCCGTTGTGCTCAAATATG
Reverse CCTTCTGAAGCCTTCTCCTTCTGC
MTRP-4 [43] Forward TCTGCAGATATCTCTGGGAAGGATGC
Reverse AAGCTITGTTCGAGCAAATTTCCATTC
MTRP-5 [43] Forward ATCTACTGCCTAGTACTACTGGCT
Reverse CAGCATGATCGGCAATGAGCTG
MTRP-6 [43] Forward AAAGATATCTTCAAATTCATGGTC
Reverse CACGTCCGCATCATCCTCAATTTC
MTRP-7 [41] Forward CGTGCTGTATGGGGTTTATAATGTTCACC
Reverse ATGCGGCCGCTTTGGAATGCTGTTAGAC
PMCA-1 [42] Forward TGGCAAACAACTCAGTTGCATATAGTGG
Reverse TCCTGTTCAATTCGACTCTGCAAGCCTCG
PMCA-2 [42] Forward TCTGGTGAGGGTGTACTGAGGACA
Reverse GAGCGTCACGTCCTGTAGTGC
PMCA-3 [42] Forward GAAGACCTCACCCACAGAGG
Reverse TCTGCTCCTGCTCAATTCGG
PMCA-4 [42] Forward AAGAAGATGATGAAGGACAACAAC
Reverse GTTGCGTACCATATTGTCTCGGTC
NCX-1 [45] Forward TGAGAGGGACCAAGATGATGAGGAA
Reverse TGACCCAAGACAAGCAATTGAAGAA
NCX-2 [45] Forward CGATGATGAGGACGATGGTGCCAGT
Reverse AGCTCAATGAAGAAGTTGTCCTTCT
NCX-3 [45] Forward TTTGGAAGGAAAGGAGGTAGAGATGAA
Reverse AGGCAAAGGAAGACTATTGAGTATT
RyR-1 Forward GAGGGCGATGAAGATGAGAAC
Reverse ATTTGCGGAAGAAGTTGAAGG
RyR-2 Forward GCTGGCCTTGTTTGTTG
Reverse TTGCCATTATGGGTAACTGAG
RyR-3 Forward CTGTGTGGTGGGCTATTACTG
Reverse AGCTGTTTGCCATTGTGAGT
Ca2+chanel L-type Forward CGAGTTTGGTTGAGCATCAC
Reverse CTCGTGGGACAGAAAAATGC
T-type Forward GGCGTGGTGGTGGAGAACTT
Reverse GATGATGGTGGGGTTGAT
P/Q-type Forward GAGATGATGGCCATTTGGCCCAAC
Reverse TCAGAGATGGTACTGAGGTCA
5min, 35 cycles of 94C (30 s), 58 C (30 s), and 72 C
(30s). The PCR products were separated on 2% agarose gel
and visualized under UV transilluminator (BioRad) after
staining with ethidium bromide. We also performed the se-
quencing to identify PCR products. The designs of primers
used in this study are listed in the table.
2.6. Statistics
Data were expressed as mean ±S.D.or S.E. indicated
in the text. Student’s paired t-test or unpaired t-test was
138 E. Yanagida et al. / Cell Calcium 36 (2004) 135–146
used to assess statistical significance. Values of <0.05 was
considered significantly different.
3. Results
3.1. Ca2+release from endoplasmic reticulum in mES cells
We investigated Ca2+release function from internal stores
by using fluo-3 confocal imaging techniques to monitor
[Ca2+]iin mES cells. First, to test the function of InsP3re-
ceptors (InsP3Rs), various agonists for G-protein coupled re-
ceptors were used. The application of 5M ATP, an agonist
for P2 receptor [16], evoked a transient elevation of [Ca2+]i
Fig. 1. ATP induced Ca2+release in mES cells. Cells were loaded with fluo-3-AM for 30 min. Fluo-3 fluorescence was recorded every 10s with the
conforcal microscopy. (A) In the upper pictures, the images of [Ca2+]iwere taken before (a), immediately after (b), and 27min after application of
5M ATP (c), as indicated in the lower graph. By the stimulation with ATP, a [Ca2+]itransient was induced. The time course of [Ca2+]iwas plotted in
the lower graph. (B) In the presence of a cell-permeable inositol-1,4,5-trisphosphate receptor blocker, 2-amino-ethoxydiphenyl borate (75M),anATP
induced [Ca2+]itransient was markedly inhibited. The time course of [Ca2+]iwas plotted in the bottom. The images were taken before (a), immediate
after (b) and 19min after application of 5 M ATP (c). We used pseudo-ratio δF/F0=(F Fbase)/Fbase, where Fis the measured fluorescence intensity
of fluo-3; Fbase, the fluorescence intensity of fluo-3 in the cell before agonist stimulation.
in all 18 cells (Fig. 1A). When cells were pretreated with
the cell-permeable InsP3receptor blocker, 2APB (75M)
[17], the effects of ATP were inhibited (Fig. 1B). The ap-
plication of 20M histamine, an agonist for H2 receptor,
evoked a large [Ca2+]itransient (18 cells) (Fig. 2A) and
these effects were blocked by the pretreatment with 2-APB
(Fig. 2B). By the stimulation of muscarinic receptors with
1M ACh, no significant changes of [Ca2+]iwere observed
(16 cells) (Fig. 2C). The mean amplitude of [Ca2+]itran-
sient was 1.01 ±0.79 (n=18), 1.77 ±0.26 (n=18) and
0.047 ±0.014 (n=16) (δF/F0; mean ±S.E.) after the ap-
plication of ATP, histamine, and ACh, respectively (Fig. 2F).
Therefore, GPCRs appeared to be involved in the release of
Ca2+from ER mediated via InsP3receptors, so called InsP3
E. Yanagida et al. / Cell Calcium 36 (2004) 135–146 139
Fig. 2. Ca2+release from endoplasmic reticulum in mES cells. (A) The
application of 20M histamine, an agonist for H2 receptors, induced a
large [Ca2+]itransient. (B) In the presence of 2-APB, a histamine-induced
[Ca2+]itransient was blocked. (C) Acetylcholine, an agonist for mus-
carinic receptors, did not affect [Ca2+]i. (D, E) The stimulation with
10mM caffeine (D), or 10 M ryanodine (E), modulators for ryanodine
receptors, did not affect [Ca2+]i. (F) Quantitative analysis of agonist ef-
fects: ATP (n=18), His (n=18), ACh (n=16), Caff (n=8) and Ry
(n=8). Data are expressed as δF/F0in control (a), the peak amplitude
of [Ca2+]ievoked by each stimulation in the absence (b) or presence of
2-APB (c). Mean ±S.E.Student’s t-test was applied to determine dif-
ferences between mean values. A P-value of <0.05 was taken as the
statistically significance level. P-value of <0.01.
induced Ca2+release (IICR). We examined which subtypes
of InsP3receptors participated in IICR using RT-PCR and
found that mRNAs for InsP3R-1, -2, and -3 genes could be
detected (Fig. 3B). On the other hand, stimulation of ryan-
odine receptors by caffeine (10mM) or 10 M ryanodine
did not change [Ca2+]i(Fig. 2D and E), indicating no func-
tion of ryanodine receptors in mES cells. Furthermore, the
expression of mRNAs for ryanodine receptors (1, 2, and 3)
could not be detected by RT-PCR (data not shown). From
these results it is concluded that Ca2+release from ER is
Fig. 3. The expression of mRNA. Using RT-PCR, OCT-4, InsP3Rs type-1,
-2, and -3 were examined. OCT-4 (230bp) (upper panel), InsP3R type-1
(779 bp), type-2 (770bp) and type-3 (773 bp) (lower panel) were expressed
at detectable levels. The sequences are 100% indentical in InsP3R type-3
and 99.8–99.9% indentical in InsP3R type-1 and -2.
mainly mediated via InsP3receptors but not ryanodine re-
ceptors in mES cells.
3.2. Ca2+entry pathways on plasma membrane in mES
cells
Next, we examined the Ca2+entry pathways on the
plasma membrane. In most non-excitable cells, a capacita-
tive Ca2+entry (CCE), which is activated by store depletion,
is supposed to work as a physiological Ca2+entry path-
way [7,9]. While undifferentiated ES cells should belong
to non-excitable cells, the existence of CCE has not been
demonstrated. To elucidate this, we designed the following
experiments. In Ca2+imaging experiments, we used a Ca2+
pump blocker, thapsigargin (TG) and further applied 20M
histamine in the Ca2+free bath solution to deplete the
stores. When the bath solution was changed to 4mM Ca2+
containing solution, a slow increase of [Ca2+]iwas ob-
served (7/7 cells) (Fig. 4A). These increases of [Ca2+]iwere
blocked by the application of 0.1mM La3+, an inhibitor for
CCE (7/7 cells). When the stores were not depleted in the
absence of TG or histamine, the increase of [Ca2+]iwas not
observed by changing to 4mM Ca2+containing solution
(8/8 cells) (Fig. 4B). The above results indicate that CCE
functions as Ca2+entry pathway in mES cells.
In contrast, voltage-operated Ca2+channels are well
known to play a central role for Ca2+entry across the
plasma membrane in electrically excitable cells and may
contribute to differentiation or proliferation. However, in
most non-excitable cells including mES cells, the functions
of VOCCs are not well understood. To examine whether
VOCCs were expressed and function in mES cells, the cells
were depolarized by high K+external solution in Ca2+
imaging experiments. The elevation of [Ca2+]iwas never
observed (Fig. 4C)(n=14), suggesting no function of
VOCCs in mES cells.
140 E. Yanagida et al. / Cell Calcium 36 (2004) 135–146
Fig. 4. Ca2+entry through plama membrane. (A) When the stores were
depleted with the 1M TG and 20 M histamine in the Ca2+free bath
solution, [Ca2+]iwas strikingly decreased (a) following a large increase
of [Ca2+]i. When [Ca2+]owas increased to 4mM, [Ca2+]igradually
increased (b, c). The application of 0.1mM LaCl3markedly decreased
[Ca2+]i(d). (B) In cells without pretreatment to deplete the stores, Ca2+
entry was not observed at 4mM [Ca2+]o. (C) When the bath solution
was changed from 5 to 145mM K+, the increase of [Ca2+]iwas not
observed. (D) Quantitative analysis of [Ca2+]ichanges. As indicated in
the graph (A), depletion of store (a), 10min (b), and 50 min (c). after
adding of 4mM Ca2+to the bath (n=7) (d); 5 min after application
of La3+. Mean ±S.E.Pvalue of <0.01 by paired Student’s t-test. (B)
0mM Ca2+(a), and 4mM Ca2+(b) (n=8). (C) 5 mM K+(a) and
145mM K+(b) (n=14).
3.3. Direct recording of store operated Ca2+current ‘ISOC
To verify the above results, we performed the whole cell
patch clamp experiments. In pooled mES cells, the mean
membrane potential was 7±4.3 mV (mean±S.D.,n=11)
and the membrane capacity was 10.9±3.6 pF (mean±S.D.,
n=48). To record ISOC without contamination by other
currents, most K+ions were replaced with Cs+and all intra-
cellular Clions were replaced with aspartate. Ca2+stores
were depleted by the pretreatment with 1M TG and ap-
Table 2
Erev nPermeability ratio
Ca2+>+55.8 ±9.3 6 1.00
Ba2++3.8 ±7.4 6 0.016
Sr2++20 ±3.5 6 0.059
plication of 20M histamine and the cells were dialyzed
with a pipette solution containing 10mM EGTA. The time
course of membrane currents held at 80mV was contin-
uously monitored. When cells were bathed with 110mM
Ca2+containing bath solution, inward currents slowly devel-
oped. These inward currents were not inactivated at 80mV
and were blocked by the application of 0.1mM La3+(6/6
cells). The current–voltage relationships of ISOC were ob-
tained by applying ramp pulses from 120 to +60mV for
200-ms (Fig. 5A(a)). The La3+sensitive currents were in-
wardly rectifying without a clear reversal potential up to
+60mV (Fig. 5A(b))(n=6), indicating ISOC.
3.3.1. Ion selectivity of SOC
To test the ionic selectivity of SOC channels in mES
cells, current-voltage relationships were obtained with Ba2+
and Sr2+. While Ca2+was replaced with equimolar Ba2+
or Sr2+in the bath solutions, the reversal potentials of
La3+sensitive currents were obtained to determine the rel-
ative permeabilities of these ions. As shown in Fig. 5B and
C,Ba
2+currents and Sr2+currents reversed at around 0
and +20mV, respectively (Table 2). The permeability ratios
were calculated from the Goldman–Hodgkin–Katz equation:
Erev =RT
ZF lnPX[X]o
PCa[Ca2+]i
where [X]oand [Ca2+]iare the bath and intracellular con-
centrations of divalent cation, respectively. The values of
Erev and the corresponding permeability ratios are summa-
rized in Table 2. The order of divalent cation permeability
through ISOC was determined to be Ca2+>Sr2+>Ba2+.
Thus, ISOC in mES cell is particularly selective for Ca2+.
As indicated above, our data showed the existence of ISOC
in mES cells (Figs. 4 and 5). Although the molecular iden-
tity of SOCs has not been determined, it is reported that
certain members of the TRP family of cation channels dis-
play properties intriguingly similar to SOCs [18]. Therefore,
we examined the expression of mRNA of several subtypes
of TRPC. As shown in Fig. 6, only mRNA TRPC-1 and -2
were detected.
3.3.2. VOCCs
To investigate the functional expression of VOCCs, whole
cell membrane currents were recorded. When voltage-steps
were applied to potentials between 70 and +100mV from
80mV holding potentials, inward currents could not be
observed (Fig. 5D) (20/20 cells). We also examined the ex-
pression of mRNAs for several kinds of VOCCs in mES
E. Yanagida et al. / Cell Calcium 36 (2004) 135–146 141
Fig. 5. Direct recording of ISOC induced by 20M histamine. Ramp clamp pulses (200 ms) were applied from 120 to +60 mV at every 2 s. Bath
solutions contained 110mM Ca2+(A), Ba2+(B), or Sr2+(C). Small inward currents were induced by the application of histamine, which were blocked
by 0.1mM La3+. (a) The representative ramp clamp currents at 10 min after application of histamine (1) and after the application of 0.1 M La3+(2)
in each trace (A–C). (b) ISOC was obtained as a La3+sensitive current ((1) – (2)) in trace (A–C). (D) Superimposed current traces elicited by 500ms
depolarizing pulses to voltages between 60 and +50mV in 10 mV steps at 0.5Hz from a holding potential of 80 mV. The standard bath solution and
the standard pipette solution were used. No inward Ca2+currents were observed.
Fig. 6. The expression of mRNA. Using RT-PCR, the TRPC-1–7 mRNAs were examined. Both TRPC-1 (372bp) and -2 (345 bp) genes were expressed
at detectable levels. The sequences are 99.8–99.9% indentical in TRPC-1 and -2.
142 E. Yanagida et al. / Cell Calcium 36 (2004) 135–146
cells. None of them included L-, T-, P/Q-type Ca2+channels
could not be detected in our experiments (data not shown).
Therefore, it is concluded that VOCCs do not function in
mES cells before differentiation.
3.4. Ca2+extrusion systems in mES cells
3.4.1. Plasma membrane Ca2+pump-ATPase
A membrane Ca2+pump (PMCA) has been identi-
fied as a major contributor to the cell Ca2+extrusion.
Therefore, we examined the function of PMCA in mES
Fig. 7. The function of plasma membrane Ca2+pump. (A) The appli-
cation of 5M carboxyeosin induced the elevation of the resting level
of [Ca2+]i. (B) Caloxin2A1 (2mM), a synthesized peptide of PMCA
blocker, markedly increased [Ca2+]ifollowing a large [Ca2+]itransient.
(C) Quantitative analysis of carboxeosin (n=8) and caloxin2A1 (n=12)
effects. Data are expressed as δF/F0before (a) and after (b) application
of blockers indicated in the graph. Mean ±S.E.Student’s paired t-test
was applied to determine differences between mean values. ∗∗Pvalue
of <0.001 in (A) and Pvalue of <0.01 in (B). (D) Using RT-PCR
Ca2+pump-ATPase (PMCA) genes were examined. The mRNA PMCA-1
(550bp) and -4 (563 bp) were expressed at detectable levels but not
PMCA-2 or -3. The sequences are 100% identical in PMCA-1 and -4.
cells. We tested two different types of Ca2+pump block-
ers, namely carboxyeosin and caloxin2A1. Application of
carboxyeosin (5M), which is a cell-permeable fluores-
cein analogue and one of the most potent blocker [19],
markedly increased basal [Ca2+]i(8/8 cells) (Fig. 7A and
C). We also tested a specific PMCA blocker, caloxin2A1,
which is a synthesized peptide obtained by screening a
random peptide phage display library for binding to the
second extacellular domain (residues 401–413) sequence
of PMCA and selected to bind the second putative extra-
cellular domain to inhibit the Ca2+pump function [15].
Application of 2mM caloxin2A1 transiently induced an
elevation of [Ca2+]i(11/12 cells) (Fig. 7B and C). From
these results, we concluded that PMCAs functioned to ex-
trude Ca2+from the cytosole and lowered [Ca2+]i.Itis
known that there are four genes PMCA isoforms 1–4 in
mammals [35]. These isoforms are known to be expressed
in a tissue-dependent manner. Therefore, we further in-
vestigated the expression of genes for PMCA isoforms by
RT-PCR. As shown in Fig. 7D, we could detect mRNAs
for PMCA-1 and -4 in mES cells but not for PMCA-2
or -3.
3.4.2. Na+/Ca2+exchanger
As well as PMCAs, the Na+/Ca2+exchanger (NCX) of
mammalian plasma membrane assumes to play an important
role in the maintenance of the intracellular Ca2+homeosta-
sis [20,21]. But little is known about whether NCX functions
in mES cells. To examine this, several kinds of blockers for
NCX were tested in Ca2+imaging experiments. Since Na+
gradient was necessary for the activation of Na+/Ca2+ex-
changer, almost all Na+in the bath solution were eliminated
to block NCX. As shown in Fig. 8A, the basal [Ca2+]iwas
increased (n=15). A specific Na+/Ca2+exchanger blocker,
KBR7943 [21], was also tested. A low dose of KBR7943
(30M) did not affect [Ca2+]i, however, the application of
high dose of KBR7943 (100M) increased [Ca2+]i(12/13
cells) (Fig. 8B–D). These results indicate that the Na+/Ca2+
exchangers might operate to extrude Ca2+from cytosole and
contribute to maintain low level of [Ca2+]iin mES cells. By
RT-PCR, mRNAs for NCX-1, -2, and -3 could be detected
in mES cells (Fig. 8E).
4. Discussion
It is known that there are widely different Ca2+signaling
systems to operate and control cellular functions depending
on cell types, which express unique Ca2+signaling tools
[22]. In this study we demonstrate a unique Ca2+signal-
ing system in mES cells. (1) InsP3induced Ca2+release
from ER, (2) Ca2+entry through the store-operated Ca2+
channels, (3) to maintain the low level of [Ca2+]i,, both
plasma membrane Ca2+pumps and Na+/Ca2+exchang-
ers play important roles for extrusion of Ca2+out of the
cytosol.
E. Yanagida et al. / Cell Calcium 36 (2004) 135–146 143
Fig. 8. The function of Na+/Ca2+exchanger. (A) When the function of Na+/Ca2+exchanger was blocked by Na+-free bath solution, the resting level of
[Ca2+]igradually elevated. (B) The application of 30M KBR7943 did not change [Ca2+]i, significantly. (C) By the application of 100M KBR7943,
[Ca2+]iwas increased. (D) Quantitative analysis. In the left, control (a) and Na+free solution (b) indicated in the graph A. In the right, before drug (a),
after application of 30M KBR7943 (b) and 100 M KBR7943 (c) indicated in the graph B or C. Mean ±S.E.Student’s paired t-test was applied to
determine differences between mean values in aand b, and between aand c.Pvalue of <0.01, ∗∗Pvalue of <0.001. (E). The expression of mRNA
NCX-1 (631bp), -2 (696 bp), and -3 (620 bp) by RT-PCR. All of these genes could be detected. The sequences are 100% identical in NCX-2 and NCX-3
and 99.9% identical in NCX-1.
4.1. InsP3induced Ca2+release in mES cells
It is well known that inositol-1,4,5-trisphosphate receptors
and ryanodine receptors families participate in the release of
Ca2+from the internal stores [5,7,22]. Generally, InsP3Rs
are main pathways for release of Ca2+from internal stores
in non-excitable cells [9]. In contrast, RyRs contribute to
Ca2+release in excitable cells, such as muscles and neurons
[8]. Since mES cell is a non-excitable one but has an abil-
ity to differentiate into many cell types including excitable
cells, it is feasible that both RyRs and InsP3Rs may func-
tion to release Ca2+from ER. Our results in Ca2+imaging
and patch clamp experiments, however, clearly showed the
functional expression of InsP3Rs but not RyRs in mES cells
(Figs. 2 and 5). It has been reported that InsP3R mRNA and
functional InsP3-gated Ca2+release channels are widely ex-
pressed at early stages of development in virtually all tissues
in murine embryos [23]. We clarified the expression of three
isoforms of InsP3Rs-1, -2, and -3 in mES cells (Fig. 3). On
the other hand, there are several reports to study ryanodine
receptors in mES cells derived cardomyocytes [24]. These
data show that ryanodine receptors are expressed at early
stages of differentiation but not in undifferentiated stages.
Therefore, we speculate that RyRs might develop and ex-
press the functions during later stages of differentiation to
excitable cells.
144 E. Yanagida et al. / Cell Calcium 36 (2004) 135–146
4.2. Ca2+entry pathways in mES cells
There are many different plasma membrane channels that
control Ca2+entry from the external medium in response
to stimuli that induce membrane depolarization, stretch,
noxious stimuli, extracellular agonists, intracellular mes-
sengers and the depletion of intracellular stores [22].In
general, store-operated Ca2+channels (SOCs) are believed
to be the main Ca2+entry pathways in non-excitable cells.
Before differentiation, where ES cells are non-excitable,
however, there is no evidence to show Ca2+entry path-
ways until now. In this study we first demonstrated the
function of SOCs as Ca2+entry pathways in mES cells
by Ca2+imagings and recordings of ISOC, which had a
very positive reversal potential (greater than +55 mV),
and was extremely selective for Ca2+(Figs. 4A and 5).
Recently, we have demonstrated the ISOC in human mes-
enchymal stem cells (hMSCs) [13]. The order of divalent
cation selectivity to ISOC in mES cell is almost identi-
cal to those of hMSC and other cells reported previously
[25]. Although the molecular identity of SOCs has not
been determined, TRPs have been the major candidates
for store-operated channels, because of the similar proper-
ties of cation permeability and the activation mechanisms
[18]. It is suspected that TRPC-1, TRPC-2, TRPC-4 and
TRPC-5 are regulated by store depletion [26]. TRPC-2 is
reported to be responsible for the sustained Ca2+influx in
mouse sperm during fertilization [26,27]. Our results by
RT-PCR showed the expression both TRPC-1 and TRPC-2
(Fig. 6), which might be responsible for Ca2+entry in mES
cells.
There are several reports to study VOCCs in stem cells,
but there is no report to study VOCCs in embryonic stem
cells. In hMSCs, the DHP receptor, 1A, 1H genes are
expressed but voltage-gated Ca2+currents are small at this
stage [13]. In embryonic stem cell derived neurons, the ex-
pression of all voltage-operated Ca2+channels, N-, L-type,
P/Q- and R type, have been reported [28]. In human neural
precursor cells, it has been reported that no inward Ca2+cur-
rent is observed [29]. The expression of T-type Ca2+current
has been reported in a mesodermal stem cell line C3H10T1/2
by patch clamp experiments [30]. In the multipotential cells
that could differentiate to vascular smooth muscle cells, the
functional expression of L-type Ca2+channel (dihydropy-
ridine receptor) depends on the differentiated states [31].
Similar results have been published by others [28,32,33].In
this study we confirmed no inward Ca2+currents in mES
cells by patch clamp experiments (Fig. 5) and also fluores-
cent studies. In addition we could not detect mRNAs for
VOCCs by RT-PCR. Therefore, we conclude that Ca2+entry
through plasma membrane is mainly mediated by SOCs but
not voltage-operated Ca2+channels in mES cells before dif-
ferentiation. We speculate that voltage-operated Ca2+chan-
nels would be expressed during the differentiation into the
wide variety of the cells that they can form with their array
of Ca2+signaling pathway.
4.3. Resting membrane potential in mES cells
The mean value of resting membrane potentials in mES
cells seems to be high compared with other cells. However,
previous reports show that inward rectifier K+channels,
which contribute to the resting membrane potential, are not
expressed in undifferentiated ES cells [47]. In human mes-
enchymal stem cells, we have found that the resting mem-
brane potential is around 10mV [39]. It is also reported
that the resting membrane potential in undifferentiated my-
oblasts is around 8mV and gradually hyperpolarizes to
30mV during the differentiation [48]. Therefore, we con-
clude that such a high value of resting membrane potential
is characteristic of mES cells.
4.4. Ca2+extrusion pathways in mES cells
Although all eukaryotic cells have Ca2+pumps in their
plasma membrane to pump Ca2+out of the cytosol, Ca2+
extrusion systems in mES cells have not been well under-
stood. So far, it has not been examined what kinds of Ca2+
extrusion pathway exist and function in mES cells. In this
study, we identified the functional expression of PMCAs and
NCXs in mES cells (Figs. 7 and 8) and concluded that both
types of transporters were involved in Ca2+extrusion from
the cytosole for maintaining the low level of [Ca2+]i.
Our results in the effects of Ca2+pump blockers show that
Ca2+responses to these blockers are quite different (Fig. 7A
and B). These different responses in [Ca2+]imight be due to
the different blocking mechanisms. Caloxin2A1 is a synthe-
sized peptide and selected to bind the second putative extra-
cellular domain of PMCA to inhibit the Ca2+pump function
[15]. On the other hand, carboxyeosine is a cell-permeable
fluorescein analogue and one of the most potent blocker for
Ca2+pump [19], however, the blocking mechanisms is still
not clear. Previously it is reported that carboxyeosine and
ATP do not compete for the Ca2+pump and suggested that
the binding site for carboxyeosine is separate for the ATP
site [46].
It is known that four separate genes encode mammalian
PMCAs. The expression of different PMCA isoforms is reg-
ulated in a developmental, tissue- and cell type-specific man-
ner, suggesting that these pumps are functionally adapted to
the physiological needs of particular cells and tissues [34].
It is also known that PMCA-1 and -4 are found in virtually
all tissues in the adult, whereas PMCA-2 and -3 are primar-
ily expressed in excitable cells of the nervous system and
muscles [35]. On the other hand, there are very few studies
appeared in the literature on the developmental pattern of
expression of the different PMCA isoforms. During mouse
embryonic development, PMCA-1 is ubiquitously detected
from the earliest stages [35]. Our results with RT-PCR pro-
vided the direct molecular evidence for the expression of
PMCA-1 and -4 isoforms in mES cells (Fig. 7).
Generally, cells such as muscle and nerve cells, which
make extensive use of Ca2+signaling, have an additional
E. Yanagida et al. / Cell Calcium 36 (2004) 135–146 145
Ca2+transport protein, Na+/Ca2+exchanger, in their
plasma membrane [36]. Our results in this study verify
the function of Na+/Ca2+exchanger in mES cells for the
first time. Recent studies provide evidence that Na+/Ca2+
exchangers function not only in excitable cells but also
in non-excitable cells, such as calf pulmonary artery en-
dothelial (CPAE) cells [19], human trophoblast [37], mouse
distal convoluted tubule cells [38]. In hMSCs we previously
showed the dynamic regulation of Ca2+extrusion by both
PMCA and NCX [39]. These evidences are consistent with
our results in this study.
We guess that [Ca2+]isignaling pathways in mES cells
would be modified during the differentiating processes into
the wide variety of cell types, which can form with their
array of [Ca2+]isignaling pathways. Our findings in this
study would provide the insight in relation to the function
of cellular processes and the implications with respect to the
mechanisms of differentiation.
Acknowledgements
Authors would like to thank Dr. Mikoshiba (Tokyo Uni-
versity, Tokyo, Japan) and Dr. Iwamoto (Fukuoka Univer-
sity, Hukuoka, Japan) for their generous gifts of 2-APB and
KBR7943. This work was supported by grants from the Min-
istry of Education, Science and Culture of Japan to S.K.
References
[1] P.J. Donovan, J. Gearhart, The end of the beginning for puluripotent
stem cells, Nat. Rev. 414 (2001) 92–97.
[2] P. Anversa, B. Nadal-Ginard, Myocyte renewal and ventricular re-
modeling, Nature 415 (2002) 240–243.
[3] M.J. Evans, M.H. Kaufman, Establishment in culture of pluripotential
cells from mouse embryos, Nature 292 (1981) 154–156.
[4] T. Burdon, A. Smith, P. Savatier, Signalling, cell cycle and pluripo-
tency in embryonic stem cells, Trends Cell Biol. 12 (2002) 432–438.
[5] M.J. Berridge, P. Lipp, M.D. Bootman, The versatility and universal-
ity of calcium-signaling, Nat. Rev. Mol. Cell Biol. 1 (2000) 11–21.
[6] J. Meldolesi, Calcium signalling: oscillation, activation, expression,
Nature 392 (1998) 863–866.
[7] M.J. Berridge, Inositol trisphosphate and calcium-signaling, Nature
361 (1993) 315–325.
[8] M.J. Berridge, Elementary and global aspects of calcium-signaling,
J. Physiol. 499 (1997) 291–306.
[9] A.C. Elliot, Recent developments in non-excitable cell calcium entry,
Cell Calcium 30 (2001) 73–93.
[10] R.L. Williams, D.J. Hilton, S. Pease, et al., Myeloid leukaemia
inhibitory factor maintains the developmental potential of embryonic
stem cells, Nature 336 (1988) 684–687.
[11] H. Niwa, J. Miyazaki, A.G. Smith, Quantitative expression of Oct-3/4
defines differentiation, dedifferentiation or self-renwal of ES cells,
Nat. Genet. 24 (2000) 372–376.
[12] S. Kawano, Y. Hirayama, M. Hiraoka, Activation mechanisms of
Ca2+-sensitive transient outward current in rabbit ventricular my-
ocytes, J. Physiol. 486 (1995) 593–604.
[13] S. Kawano, S. Shoji, S. Ichinose, K. Yamagata, M. Tagami, M.
Hiraoka, Characterization of Ca2+signalling pathways in human
mesenchymal stem cells, Cell Calcium 32 (2002) 165–174.
[14] A. Fabiato, Computer programs for calculating total from specified
free or free from specified total ionic concentrations in aqueous
solutions containing multiple metals and ligand, Methods Enzymol.
157 (1988) 378–417.
[15] C. Jyoti, M. Walia, M.J. Caloxin, a novel plasma membrane Ca2+
pump inhibitor, Am. J. Physiol.-Cell Ph. 280 (2001) C1027–C1030.
[16] G. Vassort, Adenosine 59-triphosphate: a P2-purinergic agonist in
the myocardium, Physiol. Rev. 81 (2001) 767–806.
[17] J. Wu, N. Kamimura, T. Takeo, et al., 2-Aminoethoxydiphenyl bo-
rate modulates kinetics of intracellular Ca2+signals mediated by
inositol-1,4,5-trisphosphate-sensitive Ca2+stores in single pancreatic
acinar cells of mouse, Mol. Pharmacol. 58 (2000) 1368–1374.
[18] K. Venkatachalam, D.B. Rossum, R.L. Patterson, H.T. Ma, D.L. Gill,
The cellular and molecular basis of store-operated calcium entry,
Nat. Cell Biol. 4 (2002) E263–E272.
[19] M. Sedova, L.A. Blatter, Dynamic regulation of [Ca2+]iby plasma
membrane Ca2+-ATPase and Na+/Ca2+exchange during capacitative
Ca2+entry in bovine vascular endothelial cells, Cell Calcium 25
(1999) 333–343.
[20] M.P. Blaustein, W.J. Lederer, Sodium/calcium exchange: its physio-
logical implications, Physiol. Rev. 79 (1999) 763–854.
[21] T. Iwamoto, A. Uehara, T.Y. Nakamura, I. Imanaga, M. Shigekawa,
Chimeric analysis of Na+/Ca2+exchangers NCX1 and NCX3 reveals
structural domains important for differential sensitivity to external
Ni2+or Li+, J. Biol. Chem. 274 (1999) 23094–23102.
[22] M.J. Berridge, M.D. Bootman, H.L. Roderick, Calcium-signaling:
dynamics, homeostasis and remodeling, Nat. Rev. Mol. Cell Biol. 4
(2003) 517–529.
[23] N. Rosemblit, M.C. Moschella, E. Ondrias/c ová, D.E. Gutstein, K.
Ondrias/c, A.R. Marks, Intracellular calcium release channel expres-
sion during embryogenesis, Dev. Biol. 206 (1999) 163–177.
[24] R. Kenneth, J.C. Boheler, D. Tweedie, T.H. Yang, S.V. Anisimov,
A.M. Wobus, Differentiation of pluripotent embryonic stem cells into
cardiomyocytes, Circ. Res. 91 (2002) 189–201.
[25] A.B. Parekh, R. Penner, Store depletion and calcium influx, Physiol.
Rev. 77 (1997) 901–930.
[26] D.E. Clapham, L.W. Runnels, C. Strübing, The TRP ion channel
family, Nat. Rev. Neurosci. 2 (2001) 387–396.
[27] M.K. Jungnickel, H. Marrero, L. Birnbaumer, J.R. Lémos, H.M.
Florman, Trp2 regulates entry of Ca2+into mouse sperm triggered
by egg ZP3, Nat. Cell Biol. 3 (2001) 499–502.
[28] S. Arnhold, C. Andressen, D. Angelov, et al., Embryonic stem
cell derived neurones express a maturation dependent pattern of
voltage-gated calcium channels and calcium-binding proteins, Int. J.
Dev. Neurosci. 18 (2000) 201–212.
[29] D.R. Piper, T. Mujtaba, M.S. Rao, M.T. Lucero, Immunocytochemical
and physiological characterization of a population of cultured human
neural precursors, J. Neurophysiol. 84 (2000) 534–548.
[30] Y. Kubo, Comparison of initial stages of muscle differentiation in
rat and mouse myoblastic and mouse mesodermal stem cell lines, J.
Physiol. 442 (1991) 743–759.
[31] M. Gollasch, H. Haase, C. Ried, C. Lindschau, I. Morano, F.C.
Luft, H. Haller, L-type calcium channel expression depends on the
differentiated state of vascular smooth muscle cells, FASEB J. 12
(1998) 593–601.
[32] M.D. Kilpatrick, S.N. Treistman, Time course of nerve growth factor
modulation of ethanol inhibition of Ca2+currents in PC12 cells,
Neurosci. Lett. 176 (1994) 101–104.
[33] D.L. Lewis, H.J. De Aizpurua, D.M. Rausch, Enhanced expression
of Ca2+channels by nerve growth factor and the v-src oncogene in
rat phaeochromocytoma cells, J. Physiol. 465 (1993) 325–342.
[34] G.R. Monteith, B.D. Roufogalis, Plasma membrane calcium pump a
physiological perspective on its regulation, Cell Calcium 18 (1995)
459–470.
[35] E.E. Strehler, D.A. Zacharias, Role of alternative splicing in gen-
erating isoform diversity among plasma membrane calcium pumps,
Phys. Rev. 81 (2001) 21–50.
146 E. Yanagida et al. / Cell Calcium 36 (2004) 135–146
[36] M. Shigekawa, T. Iwamoto, Cardiac Na+/Ca2+exchange molecular
and pharmacological aspects, Circ. Res. Rev. 88 (2001) 864–876.
[37] R. Moreau, L. Simoneau, J. Lafond, Calcium fluxes in human
trophoblast (BeWo) cells: calcium channels, calcium-ATPase, and
sodium–calcium exchanger expression, Mol. Reprod. Dev. 64 (2003)
189–198.
[38] S.N. Kip, E.E. Strehler, Characterization of PMCA isoforms and
their contribution to intracellular Ca2+flux in MDCK cells, Am. J.
Physiol.-Renal 283 (2002) F29–F40.
[39] S. Kawano, K. Otsu, S. Shoji, K. Yamagata, M. Hiraoka, Ca2+
oscillations regulated by Na+–Ca2+exchanger and plasma membrane
Ca2+pump induce fluctuations of membrane currents and potentials
in human mesenchymal stem cells, Cell Calcium 34 (2003) 145–156.
[40] C. Kimura, M.M. Shen, N. Yakeda, S. Aizawa, I. Matsuo, Comple-
mentary functions of Otx2 and cripto in initial patterning of mouse
epiblast, Dev. Biol. 235 (2001) 12–32.
[41] P. Gailly, C.V. Schoor, Involvement of trp-2 protein in store-operated
influx of calcium in fibroblasts, Cell Calcium 30 (2001) 157–165.
[42] C.E. Magyar, K.E. White, R. Rojas, G. Apodaca, P.A. Friendman,
Plasma membrane Ca2+-ATPase and NCX1 Na+/Ca2+exchanger
expression in distal convoluted tubule cells, Am. J. Physiol.-Renal
283 (2002) F29–F410.
[43] L.G. Reynaldo, W.P. Schilling, Differential expression of mammalian
TRP homologues across tissues and cell lines, Biochem. Bioph. Res.
Co. 239 (1997) 279–283.
[44] M. Hayashi, T. Monkawa, T. Yoshida, et al., Intracellular cal-
cium concentration in the inositol trisphosphate receptor type
1 knockout mouse, J. Am. Soc. Nephrol. 10 (1999) 2094–
2101.
[45] T. Iwamoto, M. Shigekawa, Differential inhibition of Na+/Ca2+
exchanger isoforms by divalent cations and isothiourea derivative,
Am. J. Physiol.-Cell Physiol. 275 (1998) C423–C430.
[46] C. Gatto, M.A. Milanick, Inhibition of the red blood cell calcium
pump by eosin and other fluorescein analogues, Am. J. Physiol.-
Cell Physiol. 264 (1993) C1577–C1586.
[47] J. Hescheler, et al., Embryonic stem cells: a model to study structural
and functional properties in cardiomyogenesis, Cardiovasc. Res. 36
(1997) 149–162.
[48] E. Cooper, A new role for ion channels in myoblastfusion, J. Cell
Biol. 153 (2001) F9–F11.
... The 340/380 nm-emission ratio was analyzed over a time course of 4 min, and no difference was found in the basal Ca 2+ levels between Dicer deleted (Dicer −/− ) and undeleted (Dicer fl/fl ) ESCs ( Figure 2). Since the endoplasmic reticulum (ER) is the major storage site for Ca 2+ in ESCs [26], we asked if perturbations in Ca 2+ transport across the ER membrane would differentially affect cytoplasmic Ca 2+ levels in Dicer −/− ESCs. Thapsigargin inhibits Ca 2+ uptake into the ER by blocking the sarcoendoplasmic reticulum Ca 2+ -ATPase (SERCA), which results in the release of Ca 2+ into the cytoplasm through ER Ca 2+ export transporters. ...
... emission ratio was analyzed over a time course of 4 min, and no difference was found in the basal Ca 2+ levels between Dicer deleted (Dicer −/− ) and undeleted (Dicer fl/fl ) ESCs ( Figure 2). Since the endoplasmic reticulum (ER) is the major storage site for Ca 2+ in ESCs [26], we asked if perturbations in Ca 2+ transport across the ER membrane would differentially affect cytoplasmic Ca 2+ levels in Dicer −/− ESCs. Thapsigargin inhibits Ca 2+ uptake into the ER by blocking the sarcoendoplasmic reticulum Ca 2+ -ATPase (SERCA), which results in the release of Ca 2+ into the cytoplasm through ER Ca 2+ export transporters. ...
... Ca 2+ release from the ER is mediated by the stimulation of ryanodine and inositol trisphosphate (IP 3 ) receptors in the ER membrane, whereas import into the ER is mediated by SERCA, as stated above. ESCs have very low expression of ryanodine receptors [26] and, consistent with this, the stimulation of ryanodine receptors by the agonist caffeine gave a minimal Ca 2+ response, which was very similar in Dicer fl/fl and Dicer −/− ESCs ( Figure 5B). In contrast, IP 3 receptors are expressed in ESCs, and one isoform, Itpr2 was found to be more highly expressed in Dicer −/− ESCs. ...
Article
Full-text available
MicroRNAs (miRNAs) are important regulators of embryonic stem cell (ESC) biology, and their study has identified key regulatory mechanisms. To find novel pathways regulated by miRNAs in ESCs, we undertook a bioinformatics analysis of gene pathways differently expressed in the absence of miRNAs due to the deletion of Dicer, which encodes an RNase that is essential for the synthesis of miRNAs. One pathway that stood out was Ca2+ signaling. Interestingly, we found that Dicer−/− ESCs had no difference in basal cytoplasmic Ca2+ levels but were hyperresponsive when Ca2+ import into the endoplasmic reticulum (ER) was blocked by thapsigargin. Remarkably, the increased Ca2+ response to thapsigargin in ESCs resulted in almost no increase in apoptosis and no differences in stress response pathways, despite the importance of miRNAs in the stress response of other cell types. The increased Ca2+ response in Dicer−/− ESCs was also observed during purinergic receptor activation, demonstrating a physiological role for the miRNA regulation of Ca2+ signaling pathways. In examining the mechanism of increased Ca2+ responsiveness to thapsigargin, neither store-operated Ca2+ entry nor Ca2+ clearance mechanisms from the cytoplasm appeared to be involved. Rather, it appeared to involve an increase in the expression of one isoform of the IP3 receptors (Itpr2). miRNA regulation of Itpr2 expression primarily appeared to be indirect, with transcriptional regulation playing a major role. Therefore, the miRNA regulation of Itpr2 expression offers a unique mechanism to regulate Ca2+ signaling pathways in the physiology of pluripotent stem cells.
... Studies suggest that Ca 2+ channels play crucial roles in CSC functioning, with dysregulated Ca 2+ signalling proteins promoting their differentiation into cancer cells and enhancing chemoresistance [113,114] . Dysregulation of various Ca 2+ channels, including VGCCs [81,115] , RyRs [116,117] , IP3R [118] , SOCE [119] , or TRPs [120,121] , has been observed in CSCs across different cancer types. Targeting these dysregulated Ca 2+ channels induces terminal differentiation of CSCs and sensitizes them to chemotherapy. ...
Article
Full-text available
Globally, cancer, as a major public health concern, poses a severe threat to people’s well-being. Advanced and specialized therapies can now cure the majority of people with early-stage cancer. However, emerging resistance to traditional and novel chemotherapeutic drugs remains a serious issue in clinical medicine. Chemoresistance often leads to cancer recurrence, metastasis, and increased mortality, accounting for 90% of chemotherapy failures. Thus, it is important to understand the molecular mechanisms of chemoresistance and find novel therapeutic approaches for cancer treatment. Among the several factors responsible for chemoresistance, calcium (Ca²⁺) dysregulation plays a significant role in cancer progression and chemoresistance. Therefore, targeting this derailed Ca²⁺ signalling for cancer therapy has become an emerging research area. Of note, the Ca²⁺ signal and its proteins are a multifaceted and potent tool by which cells achieve specific outcomes. Depending on cell survival needs, Ca²⁺ is either upregulated or downregulated in both chemosensitive and chemoresistant cancer cells. Consequently, the appropriate treatment should be selected based on Ca²⁺ signalling dysregulation. This review discusses the role of Ca²⁺ in cancer cells and the targeting of Ca²⁺ channels, pumps, and exchangers. Furthermore, we have emphasised the role of Ca²⁺ in chemoresistance and therapeutic strategies. In conclusion, targeting Ca²⁺ signalling is a multifaceted process. Methods such as site-specific drug delivery, target-based drug-designing, and targeting two or more Ca²⁺ proteins simultaneously may be explored; however, further clinical studies are essential to validate Ca²⁺ blockers’ anti-cancer efficacy.
... The aberrant expression of IP3R, which is responsible for calcium release from the endoplasmic reticulum, was reported in different cancer types (Cui et al., 2017). The expression of these channels was found to be conserved in different types of stem cells and their implication in cell proliferation and differentiation was confirmed (Cui et al., 2017;Kotova et al., 2014;Yanagida et al., 2004;Ye, 2010). So far, only one group reported an indirect link between IP3R and CSCs phenotype. ...
Thesis
Since approved by the FDA (Food and Drug Administration) in 1978, platinum doublet therapy remains the current gold standard chemotherapeutic treatment for non-small cell lung cancer (NSCLC). However, resistance to platinum salts evolves rapidly in patients with NSCLC and one of the major reasons behind this resistance is their proved ability to enrich CSC (cancer stem cell) population. Calcium is recognized as an activator of critical signaling pathways, including the ones responsible for resistance to apoptosis. Herein, the calcium channel Orai3 has been recently identified as an important element in the resistance to chemotherapy in breast cancer cells. Furthermore, Orai3 channel is overexpressed in NSCLC tissues, is correlated to a high tumor grade and constitutes a predictive marker of metastasis and survival in resectable NSCLC tumors. In-vitro, Orai3 channel controls cell proliferation in NSCLC cell lines in a calcium dependent manner, via the Akt pathway. In the present work, we investigated the role of Orai3 channel in resistance to Cisplatin in NSCLC and the signaling pathways associated. We found that Orai3 channel becomes overexpressed after treatment with Cisplatin in NSCLC tissues and cell lines. Clinically, Orai3 overexpression after chemotherapy was associated with partial or no tumor regression. In cell lines, Cisplatin treatment increased Orai3 expression in a time-dependent manner and this overexpression was accompanied by an enrichment of CSCs population demonstrated by CD133 staining and an overexpression of the CSCs markers Nanog, SOX-2 and Slug. Moreover, Orai3 silencing or the reduction of extracellular calcium concentration sensitized the cells to Cisplatin and leads to a drastic reduction in the expression of Nanog and SOX-2 and a partial reduction of Slug expression. Furthermore, Orai3 overexpression induced the overexpression of both markers Nanog and SOX-2. Interestingly, we remarked a change in the function of Orai3 upon the treatment with Cisplatin. In basal conditions, Orai3 was found to regulate basal calcium entry in control cells or cells transfected with an empty vector while upon the treatment with Cisplatin or when Orai3 is overexpressed via Orai3 vector transfection, it becomes involved in SOC entry. We also noticed that Orai3 functions as SOC channel in CSCs. Finally, we found that upon the inhibition of the signaling pathway Akt, the expression of the stemness markers didn’t increase and Cisplatin’s efficiency was enhanced. In a conclusion, Orai3 overexpression enables the channel to become a SOC channel and favors the calcium entry. Also, Orai3 channel, via the Akt pathway, promotes the enrichment of CSCs which are insensitive to Cisplatin. Taken together, our results show for the first time that Orai3 channel is able to induce chemoresistance in NSCLC cells by increasing the CSCs population. Our findings provide a new context in the understanding of NSCLC resistance to chemotherapy and present Orai3 as a promising biomarker which could help in the choice of chemotherapeutic drugs for patients with NSCLC
... Inositol 1,4,5trisphosphate receptors (IP 3 Rs), which include three subtypes (IP 3 R1, IP 3 R2, and IP 3 R3), and ryanodine receptors, are the two types of Ca 2+ release channels located on the ER membrane. However, IP 3 Rs are the predominant Ca 2+ release channels expressed in ESCs and early differentiating cells from ESCs, since ryanodine receptors are hardly detected at these stages [17,[20][21][22]. These properties of IP 3 Rs suggest that they might participate in early cell fate decisions. ...
... Inositol 1,4,5trisphosphate receptors (IP 3 Rs), which include three subtypes (IP 3 R1, IP 3 R2, and IP 3 R3), and ryanodine receptors, are the two types of Ca 2+ release channels located on the ER membrane. However, IP 3 Rs are the predominant Ca 2+ release channels expressed in ESCs and early differentiating cells from ESCs, since ryanodine receptors are hardly detected at these stages [17,[20][21][22]. These properties of IP 3 Rs suggest that they might participate in early cell fate decisions. ...
Article
Type 2 inositol 1,4,5-trisphosphate receptor (IP3R2) regulates the intracellular Ca2+ release from endoplasmic reticulum in human embryonic stem cells (hESCs), cardiovascular progenitor cells (CVPCs), and mammalian cardiomyocytes. However, the role of IP3R2 in human cardiac development is unknown and its function in mammalian cardiomyocytes is controversial. hESC-derived cardiomyocytes have unique merits in disease modeling, cell therapy, and drug screening. Therefore, understanding the role of IP3R2 in the generation and function of human cardiomyocytes would be valuable for the application of hESC-derived cardiomyocytes. In the current study, we investigated the role of IP3R2 in the differentiation of hESCs to cardiomyocytes and in the hESC-derived cardiomyocytes. By using IP3R2 knockout (IP3R2KO) hESCs, we showed that IP3R2KO did not affect the self-renewal of hESCs as well as the differentiation ability of hESCs into CVPCs and cardiomyocytes. Furthermore, we demonstrated the ventricular-like myocyte characteristics of hESC-derived cardiomyocytes. Under the α1-adrenergic stimulation by phenylephrine (10 μmol/L), the amplitude and maximum rate of depolarization of action potential (AP) were slightly affected in the IP3R2KO hESC-derived cardiomyocytes at differentiation day 90, whereas the other parameters of APs and the Ca2+ transients did not show significant changes compared with these in the wide-type ones. These results demonstrate that IP3R2 has minimal contribution to the differentiation and function of human cardiomyocytes derived from hESCs, thus provide the new knowledge to the function of IP3R2 in the generation of human cardiac lineage cells and in the early cardiomyocytes.
... [10] The expression of different isoforms and splice variants is regulated in a developmental, tissue-specific, and cell type-specific manner, signifying that these pumps are functionally adapted to the physiological needs of particular cells and tissues. [11] Transcription factor 7-like 2 (TCF7L2) belongs to a family of TCF lymphoid enhancer factor transcription factors and is a key component of the Wnt signaling pathway involved in the regulation of pancreatic beta-cell proliferation, differentiation, and insulin secretion. [12] It is involved in vascular remodeling through the regulation of smooth muscle cell proliferation and endothelial cell growth. ...
Article
Full-text available
Introduction: Cardiovascular disease (CVD) is the primary cause of morbidity and premature mortality in chronic kidney disease (CKD). The transcription factor 7-like 2 (TCF7L2) gene product TCF4 is a transcription factor that acts as a downstream effector in the canonical Wnt signaling pathway and may be important in the development of both type 2 diabetes and renal development and disease. It is, therefore, plausible that mutations in this gene could manifest themselves in reduced kidney function or kidney disease through their effects on hyperglycemia as well as independent of this mechanism. The ATP2B1 gene encodes the plasma membrane calcium ATPase isoform 1, which removes bivalent calcium ions from eukaryotic cells against very large concentration gradients and is responsible for controlling the contraction and dilation of vascular smooth muscles. Aim and objectives: The aims of this study are (1) to evaluate single-nucleotide polymorphisms (SNPs) of TCF7L2 gene as cardiovascular risk predictors in CKD and (2) to evaluate SNPs of ATP2B1 gene as cardiovascular risk predictors in CKD. Subjects and methods: Fifty clinically diagnosed CKD patients in the age group between 20 and 60 years of both genders were selected as cases and fifty healthy participants from the master health checkup department were selected as controls. Genomic DNA was extracted based on the spin column kit method. The DNA samples were stored at -20°C until analysis. Genotyping for TCF7L2 gene rs7903146 (C/T) and ATP2B1 gene rs11105354 (A/G) was carried out through polymerase chain reaction. Results: T allele frequency was observed in 12 controls and 23 cases (odds ratio [OR] = 2.2, 95% confidence interval [CI]: 1.0-4.7). CC genotype was observed in 38 controls and 27 cases and CT genotype in 22 cases and 12 controls. A allele was found in 38 cases and 23 controls (OR = 2, 95% CI: 1.1-3.8). The mean values of cholesterol, low-density lipoprotein, triglycerides, glucose, insulin, urea, and creatinine were high in cases when compared to controls. Conclusion: T allele of TCF7L2 gene rs7903146 (C/T) and A allele of ATP2B1 (A/G) gene rs11105354 (A/G) are associated with CVD in CKD patients.
Article
Muscular diseases are characterized by a wide genetic diversity and the Ca²⁺-signalling machinery is often perturbed. Its characterization is therefore pivotal and requires appropriate cellular models. Muscle biopsies are the best approach but are invasive for the patient and difficult to justify if the biopsy is not for diagnostic purposes. To circumvent this, interest is mounting in urine-derived stem cells that can be differentiated into skeletal muscle cells. In the present study, we isolated stem cells from urine (USC) samples of healthy donors and differentiated them by MyoD lentiviral vector transduction into skeletal muscle cells (USC-SkMC). As expected, USCs and USC-SkMCs are characterized by a radically different pattern of expression of stem and skeletal muscle markers. Characterization of cells in the present manuscript focused on Ca²⁺-signalling. Undifferentiated and differentiated cells differed in the expression of key proteins involved in Ca²⁺-homeostasis and also displayed different Ca²⁺-responses to external stimuli, confirming that during differentiation there was a transition from a non-excitable to an excitable phenotype. In USCs, the main mechanism of calcium entry was IP3 dependent, suggesting a major involvement of receptor-operated Ca²⁺ entry. Indeed, U-73122 (a PLC inhibitor) significantly inhibited the Ca²⁺increase triggered by ATP both in calcium and calcium-free conditions. In USC-SkMCs both store- and receptor-operated calcium entry were active. Furthermore, a caffeine challenge led to Ca²⁺ release both in the presence or absence of extracellular calcium, which was inhibited by ryanodine, suggesting the presence and functionality of ryanodine receptors in USC-SkMCs. Lastly, the voltage-operated calcium channels are operative in USC-SkMCs, unlike in USCs, since stimulation with high concentration of KCl induced a significant calcium transient, partially reversed by verapamil. Our data therefore support the use of skeletal muscle cells derived from USCs as an easily amenable tool to investigate Ca²⁺-homeostasis, in particular in those (neuro)muscular diseases that lack valid alternative models.
Article
Pluripotent and post-natal, tissue-specific stem cells share functional features such as the capacity to differentiate into multiple lineages and to self-renew, and are endowed with specific cell maintenance mechanism as well as transcriptional and epigenetic signatures that determine stem cell identity and distinguish them from their progeny. Calcium is a highly versatile and ubiquitous second messenger that regulates a wide variety of cellular functions. Specific roles of calcium in stem cell niches and stem cell maintenance mechanisms are only beginning to be explored, however. In this review, I discuss stem cell-specific regulation and roles of calcium, focusing on its potential involvement in the intertwined metabolic and epigenetic regulation of stem cells.
Article
Cancer stem cells (CSC's) have emerged as a key area of investigation due to associations with cancer development and treatment resistance, related to their ability to remain quiescent, self-renew and terminally differentiate. Targeting CSC's in addition to the tumour bulk could ensure complete removal of the cancer, lessening the risk of relapse and improving patient survival. Understanding the mechanisms supporting the functions of CSC's is essential to highlight targets for the development of therapeutic strategies. Changes in intracellular calcium through calcium channel activity is fundamental for integral cellular processes such as proliferation, migration, differentiation and survival in a range of cell types, under both normal and pathological conditions. Here in we highlight how calcium channels represent a key mechanism involved in CSC function. It is clear that expression and or function of a number of channels involved in calcium entry and intracellular store release are altered in CSC's. Correlating with aberrant proliferation, self-renewal and differentiation, which in turn promoted cancer progression and treatment resistance. Research outlined has demonstrated that targeting altered calcium channels in CSC populations can reduce their stem properties and induce terminal differentiation, sensitising them to existing cancer treatments. Overall this highlights calcium channels as emerging novel targets for CSC therapies.
Article
Full-text available
The impact of calcium signalling on so many areas of cell biology reflects the crucial role of calcium signals in the control of diverse cellular functions. Despite the precision with which spatial and temporal details of calcium signals have been resolved, a fundamental aspect of the generation of calcium signals — the activation of 'store-operated channels' (SOCs) — remains a molecular and mechanistic mystery. Here we review new insights into the exchange of signals between the endoplasmic reticulum (ER) and plasma membrane that result in activation of calcium entry channels mediating crucial long-term calcium signals.
Article
Full-text available
Embryonic stem (ES) cells, the totipotent outgrowths of blastocysts, can be cultured and manipulated in vitro and then returned to the embryonic environment where they develop normally and can contribute to all cell lineages. Maintenance of the stem-cell phenotype in vitro requires the presence of a feeder layer of fibroblasts or of a soluble factor, differentiation inhibitory activity (DIA) produced by a number of sources; in the absence of DIA the ES cells differentiate into a wide variety of cell types. We recently noted several similarities between partially purified DIA and a haemopoietic regulator, myeloid leukaemia inhibitory factor (LIF), a molecule which induces differentiation in M1 myeloid leukaemic cells and which we have recently purified, cloned and characterized. We demonstrate here that purified, recombinant LIF can substitute for DIA in the maintenance of totipotent ES cell lines that retain the potential to form chimaeric mice.
Article
Calcium influx in nonexcitable cells regulates such diverse processes as exocytosis, contraction, enzyme control, gene regulation, cell proliferation, and apoptosis. The dominant Ca2+ entry pathway in these cells is the store-operated one, in which Ca2+ entry is governed by the Ca2+ content of the agonist-sensitive intracellular Ca2+ stores. Only recently has a Ca2+ current been described that is activated by store depletion. The properties of this new current, called Ca2+ release-activated Ca2+ current (ICRAC), have been investigated in detail using the patch-clamp technique. Despite intense research, the nature of the signal that couples Ca2+ store content to the Ca2+ channels in the plasma membrane has remained elusive. Although ICRAC appears to be the most effective and widespread influx pathway, other store-operated currents have also been observed. Although the Ca2+ release-activated Ca2+ channel has not yet been cloned, evidence continues to accumulate that the Drosophila trp gene might encode a store-operated Ca2+ channel. In this review, we describe the historical development of the field of Ca2+ signaling and the discovery of store-operated Ca2+ currents. We focus on the electrophysiological properties of the prototype store-operated current ICRAC, discuss the regulatory mechanisms that control it, and finally consider recent advances toward the identification of molecular mechanisms involved in this ubiquitous and important Ca2+ entry pathway.
Article
The Na+-Ca2+ exchanger (NCX) is one of the essential regulators of Ca2+ homeostasis in cardiomyocytes and thus an important modulator of the cardiac contractile function. The purpose of this review is to survey recent advances in cardiac NCX research, with particular emphasis on molecular and pharmacological aspects. The NCX function is thought to be regulated by a variety of cellular factors. However, data obtained by use of different experimental systems often appear to be in conflict. Where possible, we endeavor to provide a rational interpretation of such data. We also provide a summary of current work relating to the structure and function of the cardiac NCX. Recent molecular studies of the NCX protein are beginning to shed light on structural features of the ion translocation pathway in the NCX membrane domain, which seems likely to be formed, at least partly, by the phylogenetically conserved alpha -1 and alpha -2 repeat structures and their neighboring membrane-spanning segments. Finally, we discuss new classes of NCX inhibitors with improved selectivity. One of these, 2-[2-[4-(4-nitrobenzyloxy)phenyl]ethyl]isothiourea methanesulfonate (KB-R7943), appears to exhibit unique selectivity for Ca2+-influx-mode NCX activity. Data obtained with these inhibitors should provide a basis for designing more selective and clinically useful drugs targeting NCX.
Article
The Na+/Ca2+ exchanger, an ion transport protein, is expressed in the plasma membrane (PM) of virtually all animal cells. It extrudes Ca2+ in parallel with the PM ATP-driven Ca2+ pump. As a reversible transporter, it also mediates Ca2+ entry in parallel with various ion channels. The energy for net Ca2+ transport by the Na+/Ca2+ exchanger and its direction depend on the Na+, Ca2+, and K+ gradients across the PM, the membrane potential, and the transport stoichiometry. In most cells, three Na+ are exchanged for one Ca2+. In vertebrate photoreceptors, some neurons, and certain other cells, K+ is transported in the same direction as Ca2+, with a coupling ratio of four Na+ to one Ca2+ plus one K+. The exchanger kinetics are affected by nontransported Ca2+, Na+, protons, ATP, and diverse other modulators. Five genes that code for the exchangers have been identified in mammals: three in the Na+/Ca2+ exchanger family (NCX1, NCX2, and NCX3) and two in the Na+/Ca2+ plus K+ family (NCKX1 and NCKX2). Genes homologous to NCX1 have been identified in frog, squid, lobster, and Drosophila. In mammals, alternatively spliced variants of NCX1 have been identified; dominant expression of these variants is cell type specific, which suggests that the variations are involved in targeting and/or functional differences. In cardiac myocytes, and probably other cell types, the exchanger serves a housekeeping role by maintaining a low intracellular Ca2+ concentration; its possible role in cardiac excitation-contraction coupling is controversial. Cellular increases in Na+ concentration lead to increases in Ca2+ concentration mediated by the Na+/Ca2+ exchanger; this is important in the therapeutic action of cardiotonic steroids like digitalis. Similarly, alterations of Na+ and Ca2+ apparently modulate basolateral K+ conductance in some epithelia, signaling in some special sense organs (e.g., photoreceptors and olfactory receptors) and Ca2+- dependent secretion in neurons and in many secretory cells. The juxtaposition of PM and sarco(endo)plasmic reticulum membranes may permit the PM Na+/Ca2+ exchanger to regulate sarco(endo)plasmic reticulum Ca2+ stores and influence cellular Ca2+ signaling.
Article
There are remarkable changes of calcium binding proteins and voltage dependent Ca2+ channel subtypes during in vitro differentiation of embryonic stem cell derived neurons. To observe these maturation dependent changes neurones were studied using combined immunohistochemical, patch clamp and videomicroscopic time lapse techniques. Embryonic stem cell derived neuronal maturation proceeds from apolar to bi- and multipolar neurones, expressing all Ca2+ channel subtypes. There is, however, a clear shift in channel contribution to whole cell current from apolar neurones with mainly N- and L-type channel contribution in favour of P/Q- and R-type participation in bi- and multipolar cells. Expression of the calcium binding protein parvalbumin could be detected in bipolar, while calretinin and calbindin was preferentially found in multipolar neurones. Our data provides new insights into fundamental neurodevelopmental mechanisms related to Ca2+ homeostasis, and clarifies contradictory reports on the development of Ca2+ channel expression using primary cultures of neurones already committed to certain brain compartments.
Article
1. Electrophysiological and immunohistochemical properties during the early stages of muscle differentiation were studied in two myoblastic cell lines, mouse C2C12 and rat L6, and compared to those in myogenic clonal cells derived from the mouse mesodermal stem cell line C3H10T1/2, studied in the preceding paper. 2. Mouse C2C12 cells were induced to differentiate to muscle by changing from 10% fetal calf serum to 2% horse serum in the medium. Most of the C2C12 cells before serum reduction showed ATP-induced slow K+ current. Twelve per cent showed inward rectifier K+ current. They expressed fibronectin and Neural Cell Adhesion Molecule (NCAM). Small spindle-shaped cells at an early stage of muscle differentiation began to appear 24 h after serum reduction. In contrast to cells before serum reduction, only 13% of these spindle-shaped cells showed an ATP response. Most showed tetrodotoxin (TTX)-resistant Na+ current and outward K+ current. Thirty-eight per cent had inward rectifier K+ current. They expressed NCAM but not fibronectin. The T-type Ca2+ current was not observed up to the latest stage of differentiation investigated. 3. Rat L6 cells in maintaining culture medium showed only infrequent ATP responses, but already showed TTX-resistant Na+ current. No clear T-type Ca2+, inward rectifier K+ or outward K+ currents were observed. About one-third of the cells did not express fibronectin. From these results, L6 cells appear to be at a stage near to but slightly earlier than that of C2C12 cells after serum reduction. 4. The properties of the early stages of muscle differentiation in C3H10T1/2 cells, such as the disappearance of ATP-induced K+ current and fibronectin, and the appearance of NCAM, were also seen in C2C12 and L6. However, T-type Ca2+ and inward K+ currents, which were found in the initial stages of C3H10T1/2 muscle differentiation, were not clearly observed in C2C12 and L6. Instead, C2C12 and L6 showed a TTX-resistant Na+ current which was never observed in C3H10T1/2 cells. 5. The properties of the TTX-resistant Na+ current were investigated. In L6 cells, it was reduced to 60% by 1 microM-TTX. It could be evoked by depolarizations to a level above -50 mV with a maximum amplitude at around -15 mV. Steady-state inactivation was detectable with pre-pulses to -100 mV for 100 ms and reached half at pre-pulses of -78 mV. These parameters of inactivation are clearly different from those of the TTX-sensitive Na+ current observed in C3H10T1/2-derived mature muscle cells in the preceding paper.
Article
This chapter focuses on the computer programs for calculating total from specified free, or free from specified total ionic concentrations in aqueous solutions containing multiple metals and ligands. Many experiments in biology, biochemistry, biophysics, and physiology require aqueous solutions with concentrations of free cations, far below those obtainable by simply adding the corresponding salts to the solutions. Then metal buffers made up of the metallic ion (or cation) and the appropriate ligand (or anion) must be used. Similarly, living cells contain ligands, such as cation-binding proteins, that cause the total to differ from the free concentrations of metals. Ligands also bind hydrogen ions so that instead of a single complex between metal and ligand, several complexes form, corresponding to various degrees of protonation. Each has its own stability constant. Thus, apparent stability constants (Kapp) have to be calculated to characterize the multiple equilibria among one metal, one ligand, and the hydrogen ion at a given pH value. The main thrust of the present chapter is to describe a FORTRAN program named "SPECS" in such a way that it can be readily used by the largest number of investigators possible.
Article
Excerpt The level of free calcium within the cytosol regulates a range of cellular processes. Complex homeostatic mechanisms based on calcium pumps ensure that the resting level of calcium is kept low (~100 nm). A variety of external signals can trigger an increase in calcium either by promoting an influx of external calcium or by mobilizing calcium from intracellular reservoirs. The ability of many agonists (neurotransmitters, hormones, and growth factors) to generate such calcium signals is very dependent on the hydrolysis of inositol lipids (Michell 1975; Berridge and Fain 1979). This calcium-signaling pathway has a series of transduction steps. The external signal arriving at the cell surface triggers lipid hydrolysis to give inositol trisphosphate, and that then mobilizes the calcium responsible for stimulating a variety of effector systems. The first transduction event occurs at the plasma membrane, where the external signal is translated into intracellular second messengers. The agonist binds to...