ArticlePDF Available

PKC-dependent coupling of calcium permeation through transient receptor potential canonical 3 (TRPC3) to calcineurin signaling in HL-1 myocytes

Authors:

Abstract and Figures

Cardiac transient receptor potential canonical (TRPC) channels are crucial upstream components of Ca(2+)/calcineurin/nuclear factor of activated T cells (NFAT) signaling, thereby controlling cardiac transcriptional programs. The linkage between TRPC-mediated Ca(2+) signals and NFAT activity is still incompletely understood. TRPC conductances may govern calcineurin activity and NFAT translocation by supplying Ca(2+) either directly through the TRPC pore into a regulatory microdomain or indirectly via promotion of voltage-dependent Ca(2+) entry. Here, we show that a point mutation in the TRPC3 selectivity filter (E630Q), which disrupts Ca(2+) permeability but preserves monovalent permeation, abrogates agonist-induced NFAT signaling in HEK293 cells as well as in murine HL-1 atrial myocytes. The E630Q mutation fully retains the ability to convert phospholipase C-linked stimuli into L-type (Ca(V)1.2) channel-mediated Ca(2+) entry in HL-1 cells, thereby generating a dihydropyridine-sensitive Ca(2+) signal that is isolated from the NFAT pathway. Prevention of PKC-dependent modulation of TRPC3 by either inhibition of cellular kinase activity or mutation of a critical phosphorylation site in TRPC3 (T573A), which disrupts targeting of calcineurin into the channel complex, converts cardiac TRPC3-mediated Ca(2+) signaling into a transcriptionally silent mode. Thus, we demonstrate a dichotomy of TRPC-mediated Ca(2+) signaling in the heart constituting two distinct pathways that are differentially linked to gene transcription. Coupling of TRPC3 activity to NFAT translocation requires microdomain Ca(2+) signaling by PKC-modified TRPC3 complexes. Our results identify TRPC3 as a pivotal signaling gateway in Ca(2+)-dependent control of cardiac gene expression.
Content may be subject to copyright.
PKC-dependent coupling of calcium permeation
through transient receptor potential canonical 3
(TRPC3) to calcineurin signaling in HL-1 myocytes
Michael Poteser
a
, Hannes Schleifer
a
, Michaela Lichtenegger
a
, Michaela Schernthaner
a
, Thomas Stockner
b
,
C. Oliver Kappe
c
, Toma N. Glasnov
c
, Christoph Romanin
d
, and Klaus Groschner
a,1
a
Institute of Pharmaceutical Sciences, University of Graz, 8010 Graz, Austria;
b
Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria;
c
Institute of Chemistry, University of Graz, 8010 Graz, Austria; and
d
Institute of Biophysics, University of Linz, 4040 Linz, Austria
Edited* by Lutz Birnbaumer, National Institute of Environmental Health Sciences, Research Triangle Park, NC, and approved May 17, 2011 (received for review
April 21, 2011)
Cardiac transient receptor potential canonical (TRPC) channels are
crucial upstream components of Ca
2+
/calcineurin/nuclear factor of
activated T cells (NFAT) signaling, thereby controlling cardiac tran-
scriptional programs. The linkage between TRPC-mediated Ca
2+
sig-
nals and NFAT activity is still incompletely understood. TRPC
conductances may govern calcineurin activity and NFAT transloca-
tion by supplying Ca
2+
either directly through the TRPC pore into
a regulatory microdomain or indirectly via promotion of voltage-
dependent Ca
2+
entry. Here, we show that a point mutation in the
TRPC3 selectivity lter (E630Q), which disrupts Ca
2+
permeability
but preserves monovalent permeation, abrogates agonist-induced
NFAT signaling in HEK293 cells as well as in murine HL-1 atrial myo-
cytes. The E630Q mutation fully retains the ability to convert phos-
pholipase C-linked stimuli into L-type (Ca
V
1.2) channel-mediated
Ca
2+
entry in HL-1 cells, thereby generating a dihydropyridine-
sensitive Ca
2+
signal that is isolated from the NFAT pathway. Pre-
vention of PKC-dependent modulation of TRPC3 by either inhibition
of cellular kinase activity or mutation of a critical phosphorylation
site in TRPC3 (T573A), which disrupts targeting of calcineurin into
the channel complex, converts cardiac TRPC3-mediated Ca
2+
signal-
ing into a transcriptionally silent mode. Thus, we demonstrate a di-
chotomy of TRPC-mediated Ca
2+
signaling in the heart constituting
two distinct pathways that are differentially linked to gene tran-
scription. Coupling of TRPC3 activity to NFAT translocation requires
microdomain Ca
2+
signaling by PKC-modied TRPC3 complexes.
Our results identify TRPC3 as a pivotal signaling gateway in Ca
2+
-
dependent control of cardiac gene expression.
Ca
2+
homeostasis
|
NFATc1 transactivation
|
transient receptor potential
canonical
|
divalent permeation
As a universal and versatile second messenger, calcium (Ca
2+
)
governs a multitude of cellular effector functions in the
heart including transcriptional programs and cellular remodeling
processes (1). Coordinated control of cardiac functions by Ca
2+
re-
quires efcient segregation of Ca
2+
signals into regulatory micro-
domains, resulting in specicity of coupling between Ca
2+
sources
and Ca
2+
-dependent effector systems. So far, the molecular com-
position and architecture of Ca
2+
signaling microdomains for
control of cardiac transcriptional programs is incompletely un-
derstood. Cation channels of the transient receptor potential ca-
nonical (TRPC) family constitute a ubiquitous signal transduction
machinery for Ca
2+
entry and have recently been identied as ion
channels that trigger pathophysiological activation of nuclear factor
of activated T-cell (NFAT)-mediated gene transcription and hy-
pertrophic remodeling in the heart (24). TRPC proteins form
Ca
2+
permeable plasma membrane channels that are typically ac-
tivated in response to hormonal stimuli linked to phospholipase
C signaling (5). These channels lack, or display only modest, se-
lectivity for Ca
2+
over monovalent cation (6) and are able to gen-
erate increases in cytosolic Ca
2+
via multiple mechanisms including
indirect initiation of Ca
2+
entry via voltage-gated Ca
2+
channels (7)
or the sodium calcium exchanger (NCX) because of modulation of
membrane potential and/or local Na
+
gradients (8). TRPC chan-
nels are expected to contribute divergently to Ca
2+
signaling in
nonexcitable and in excitable cells, which provide a certain reper-
toire of voltage-dependent Ca
2+
transport systems.
TRPC3 is a lipid-regulated member of the TRPC subfamily and
a potential player in cardiac pathophysiology (9). For homomeric
TRPC3 channels, a Ca
2+
/Na
+
permeability ratio of 1.6 was
determined (10) and functional crosstalk of TRPC3 channels with
cardiac voltage-gated Ca
2+
channels and NCX1 has been sug-
gested (7, 11). Representing a typical nonselective cation channel,
TRPC3 controls cellular processes by either Ca
2+
permeation
through its pore and generation of a local Ca
2+
signal at the TRP
channel signalplex or by remote effects on voltage-gated Ca
2+
channels or electrogenic transporters. According to a paradigm of
cardiac (patho)physiology, beat-to-beat Ca
2+
cycling (E-C cou-
pling) in the heart is separated from Ca
2+
signaling events that
control gene expression (12). Interestingly, TRPC3 has been
suggested to govern gene transcription by mechanisms involving
a linkage to voltage-dependent Ca
2+
entry (7), which is also es-
sential for E-C coupling. However, the Ca
2+
entry mechanism,
which links cardiac TRPC3 activity to gene expression is elusive
and it is unclear whether nonselective TRPC channels serve as
dual Ca
2+
signaling units that control segregated Ca
2+
pools. To
address these questions, we set out to engineer the TRPC3 cation
permeation pathway and generated a single point mutation
(E630Q) that lacks divalent- but retains monovalent permeability
and, thus, the potential to control voltage-dependent Ca
2+
entry.
This mutant was found capable of functional coupling to cardiac
Ca
V
1.2 signaling but not to NFAT activation. We present evi-
dence for a tight link between TRPC3 channel activity and NFAT
nuclear translocation based on Ca
2+
permeation through the
TRPC3 pore, generating a local Ca
2+
signaling event that is
sensed by the downstream effector calcineurin (CaN), which is
targeted to cardiac TRPC3 channels. Moreover, we demonstrate
that protein kinase C-dependent modulation of the channel
enables switching between transcriptionally active and inactive
TRPC3-signaling modes.
Results and Discussion
Identication of E630 as a Critical Residue in the TRPC3 Selectivity
Filter. In an attempt to obtain TRPC mutants with altered ion
selectivity, we initially generated a structural model of the TRPC3
pore region by using a recently developed alignment strategy (13)
Author contributions: M.P. and K.G. designed research; M.P., H.S., M.L., M.S., and T.S.
performed research; C.O .K., T.N.G., and C.R. co ntributed new reagents/a nalytic tools;
M.P., H.S., M.L., M.S., and T.S. analyzed data; and M.P., H.S., and K.G. wrote the paper.
The authors declare no conict of interest.
*This Direct Submission article had a prearranged editor.
Freely available online through the PNAS open access option.
1
To whom correspondence should be addressed. E-mail: klaus.groschner@uni-graz.at.
This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.
1073/pnas.1106183108/-/DCSupplemental.
1055610561
|
PNAS
|
June 28, 2011
|
vol. 108
|
no. 26 www.pnas.org/cgi/doi/10.1073/pnas.1106183108
and structure information available for KcsA as well as a Kv1.2-
Kv1.3 chimeric pore. Our hypothetical pore model is illustrated in
Fig. S1Aalong with the sequence comparison of TRPC3 and
template pore structures (Fig. S1B). Three of the ve negative
residues were predicted to be accessible and exposed to the per-
meation pathway. According to our molecular model, only one
glutamate (E630) was localized within the central part of the
permeation pathway, whereas the other two residues (E616 and
D639) were predicted as part of the extracellular vestibule. Mu-
tagenesis and functional analysis conrmed a critical role of the
central glutamate in position 630. Charge inversion (E630K)
yielded a nonfunctional channel (Fig. S2), whereas neutralization
(E630Q) produced a channel that displayed moderately altered
current to voltage (I-V) relation in normal extracellular (Na
+
plus
Ca
2+
containing; Fig. 1 Aand C) solution with a relative increase
in the conductance at neutral potential. The I-V relation of the
TRPC3-E630Q mutant was virtually insensitive to changes in
extracellular Ca
2+
(Fig. S3). Inspection of I-V relations with Ca
2+
as the sole extracellular cationic charge carrier and BAPTA in the
pipette solution to eliminate indirect, Ca
2+
-mediated currents
revealed that the E630Q mutation profoundly reduced Ca
2+
permeation although the channel complex (Fig. 2 Band D). In-
ward currents were essentially small or lacking with Ca
2+
as
a charge carrier even at large hyperpolarizing potentials, and re-
versal potentials were difcult to determine in most experiments.
Nonetheless, from six experiments a mean of 79.6 ±6.3 mV was
calculated, demonstrating a substantial shift in reversal potential
compared with wild-type channels (1.9 ±1.4; n= 7). The Ca
2+
/
Cs
+
permeability ratio was reduced from 4.2 to less than 0.02 in
the mutant. Thus, our experiments identied a key amino acid
within the cation permeation structure of TRPC3. The negative
charge in position 630 is apparently essential for divalent per-
meation but not for transition of monovalent cations through the
TRPC3 pore. Our nding is in line with previous reports on the
role of negatively charged residues in Ca
2+
transition through
TRP pores (1417) and, specically, with a prediction of critical
residues in the TRPC selectivity lter obtained by Liu et al. in a
study with the prototypical Drosophila TRP channel (14). It is of
note that the identied negatively charged residue is conserved
in the TRPC3/6/7 subfamily but absent in more distant relatives of
TRPC3. Therefore, Ca
2+
permeation in within the TRPC family
of cation channels may involve distinctly different molecular
mechanisms. Our results support the notion that monovalent and
divalent permeation through nonselective TRP cation channels
may involve separate, specic interaction sites within the pore,
which combine to a nonselectivepathway that conducts both
types of charge carriers. Based on our observation that a single
point mutation within the TRPC3 pore causes specic elimination
of Ca
2+
permeation, we set out to use this genetically engineered
cation channel to explore the cellular impact of the TRPC3-
mediated monovalent conductance and to identify downstream
signaling pathways that are specically linked to either the
monovalent transport or to Ca
2+
entry through the channel pore.
Because the TRPC monovalent conductance is considered of
particular importance in excitable cells, and because recentstudies
have demonstrated the relevance of TRPC channels in cardiac
pathophysiology (2, 4, 7, 18) we focused on the cardiac system,
using the HL-1 murine atrial cell line. Initially, we compared basic
properties of TRPC3 signaling in HL-1 cells with those in the
well characterized electrically nonexcitable HEK293 system.
TRPC3 Mediates Agonist-Induced Ca
2+
Signals in HEK293 and HL-1
Cells by Divergent Mechanisms. So far, the relative contribution
of direct Ca
2+
permeation through the TRPC3 pore and indirect
mechanisms involving TRPC-mediated changes in membrane
potential and voltage-dependent signaling partners such as NCX1
has not been evaluated in HEK293 cells. Expression levels of
endogenous voltage-gated Ca
2+
channels are below the detection
threshold, and NCX1 expression is typically moderate to low.
Hence, only a minor fraction of the TRPC3-mediated Ca
2+
signal
is expected to involve indirect mechanisms in HEK293. Pharma-
cological characterization of the TRPC3-mediated Ca
2+
entry
pathway in HEK293 and HL-1 cells, determined by using a clas-
sical Ca
2+
readdition protocol, revealed that global Ca
2+
signals
were based on distinctly different mechanisms (Fig. S4). Elec-
trophysiological experiments conrmed that TRPC3 channels
were active when Ca
2+
was elevated after activation by agonist
administration in Ca
2+
-free solution (Fig. S5). TRPC3 over-
expressing HEK293 as well as HL-1 displayed Ca
2+
entry that
was highly sensitive to inhibition by the TRPC3 blocker Pyr3 (19).
KB-R7943, an inhibitor of NCX reverse mode operation, sup-
pressed Ca
2+
entry only moderately in HEK293 cells and lacked
inhibitory effects in HL-1 cells. Block of voltage-gated, L-type
Ca
2+
channels by nifedipine strongly suppressed Ca
2+
entry
into endothelin-stimulated HL-1 cells but had no effect on the
Ca
2+
signal in HEK293 cells (Fig. S4). These results indicate that
TRPC3 is effectively linked to voltage-gated Ca
2+
signaling in
cardiac cells and are able to produce large global cytosolic Ca
2+
rises via promotion of Ca
2+
entry through Ca
V
1.2 channels. The
observed lack of NCX-mediated Ca
2+
entry into HL-1 cells may
be explained by predominant forward mode operation in these
cardiac cells, based on a tight functional coupling to voltage-gated
Ca
2+
entry channels as well as more negative membrane poten-
tials compared with HEK293 cells. Thus, HL-1 myocytes repre-
sent an electrically excitable cell type that displays functional
cross-talk and signaling partnership between nonselective TRPC
conductances and voltage-gated Ca
2+
channels. Thereby, TRPC3
signaling in HL-1 cells is distinctly different from that in the
nonelectrically excitable HEK293 cell system.
As a next step, we aimed to delineate the cellular role of direct
Ca
2+
permeation through TRPC3 channels in these cells by char-
acterizing coupling between Ca
2+
entry and the NFAT down-
stream effector system for wild type and pore mutants (E630Q and
E630K) of TRPC3.
Ca
2+
Permeation Through the Pore of TRPC3 Channels Is Essential for
Activation of the NFAT Pathway. Carbachol-induced Ca
2+
entry as
well as NFAT translocation was strongly reduced by expression of
either the Ca
2+
permeation-decient mutant (E630Q) or a pore-
dead mutant(E630K) (Fig. 2). Basal Ca
2+
entry into nonstimulated
cells was reduced when expressing either of the TRPC3 pore
mutants to the level of vector-transfected controls (Fig. 2B). As
expected from the proposed NCX1-mediated Ca
2+
entry contri-
Fig. 1. Neutralization of E630 in TRPC3 (E630Q) eliminates Ca
2+
but not
monovalent permeability. Representative ramp protocol recordings from
HEK293 cells transfected by either TRPC3-WT (Aand B) or the mutant
channel E630Q (Cand D) in the presence of 140 mM extracellular Na
+
and
2mMCa
2+
(Aand C), or in absence of extracellular Na
+
and presence of
10 mM Ca
2+
using 10 mM BAPTA in the pipette solution (Band D), before
(black) and after stimulation with 100 μM carbachol (+CCh, red).
Poteser et al. PNAS
|
June 28, 2011
|
vol. 108
|
no. 26
|
10557
CELL BIOLOGY
bution, TRPC3-E630Q didnot fully eliminate the Ca
2+
entry signal.
Ca
2+
entry into cells expressing TRPC3-E630Q remained signi-
cantly higher than in cells transfected to express TRPC3-E630K.
Nonetheless, NFAT translocation in HEK293 cells was completely
suppressed with either pore mutation of TRPC3 (Fig. 2 Cand D).
This nding indicated that Ca
2+
permeation through the pore is
essential to initiate NFAT translocation, whereas indirect NCX-
mediated signaling was barely involved because the NCX inhibitor
KB-R7943 (5 μM) failed to prevent NFAT translocation (Fig. S6).
A possible dual Ca
2+
signaling function of TRPC3 was further
investigated in the electrically excitable cardiac HL-1 cell line. As
illustrated in Fig. 3, Ca
2+
entry into endothelin-stimulated cells
was slightly enhanced compared with vector-transfected controls
(Fig. 3A) by expression of either wild-type TRPC3 (Fig. 3B)or
TRPC3-E630Q (Fig. 3C) but reduced down to basal (non-
stimulated) level with TRPC3-E630K (Fig. 3D). Expression of
wild-type TRPC3 (Fig. 3B) or TRPC3-E630Q (Fig. 3C) generated a
Ca
2+
signal that was mainly based on voltage-gated Ca
V
1.2
channels as evident by its sensitivity to nifedipine (3 μM). In clear
contrast to the observed Ca
2+
signals, cells expressing TRPC3-
E630Q lacked endothelin-stimulated NFAT translocation (Fig.
3C). Thus, the TRPC3 mutant with impaired divalent conductance
(E630Q) is able to initiate a large global Ca
2+
signal via promotion
of voltage-gated Ca
2+
entry, but this signal is not translated into
NFAT activation. Importantly, even endogenous TRPC3 channels
appear sufcient to exert a signicant impact on NFAT trans-
location as evident from vector-transfected controls displaying
higher translocation than cells expression the pore-dead domi-
nant-negative E630K mutant. It is of note that, in contrast to
HEK293 cells, NFAT translocation in HL-1 cells was not signi-
cantly promoted in response to depletion of intracellular stores
with thapsigargin (Fig. S7), indicating that the channels involved in
NFAT signaling of HL-1 are not classical store-operated Ca
2+
entry channels. Opening of TRPC3 channels resulted in NFAT
activation, but also generation of an additional intracellular Ca
2+
signal mediated by voltage-gated Ca
2+
channels that was fairly
well segregated from the NFAT pathway. Notably, TRPC3-
mediated NFAT activation can occur at barely detectable global
Ca
2+
changes such as in the presence of nifedipine (3 μM) in
control cells or cells overexpressing TRPC3 (Fig. 3 Aand B),
therefore we hypothesized that the triggering Ca
2+
elevation is
likely to take place in a restricted signaling microdomain at the
TRPC3 channel complex, containing essential downstream sig-
naling components such as calmodulin and calcineurin (CaN) to
allow specic transduction of this local Ca
2+
signal. Indeed, direct
association of TRPC3 with CaN have been demonstrated (20, 21)
and the existence of a dynamic TRPC/CaN signaling complexes
have been proposed.
Coupling of Cardiac TRPC3 Signaling to Activation of the NFAT
Pathway Involves PKC-Dependent Phosphorylation. Previous inves-
tigations demonstrated assembly of CaN along with immuno-
phyllins (FKBP12) into TRPC6 signalplexes and dependency of
this process on protein kinase C-mediated phosphorylation (21).
PKC-mediated phosphorylation of TRPC3 appears essential for
both recruitment of CaN into TRPC complexes and inhibitory
regulation (22, 23). This result prompted us to hypothesize that
suppression of PKC phosphorylation may disrupt the functional
TRPC3/CaN signaling unit without preventing channel function.
Consequently, we set out to test whether TRPC3 linkage to NFAT
nuclear translocation depends on regulation of the channel
complex by PKC. To suppress TRPC3 phosphorylation by PKC
isoenzymes, we performed experiments with GF109203X, a com-
pound that inhibits conventional PKC isoforms including PKC-γ,
as one essential player in the control of TRPC3 channels (23).
Because CaN has been shown to associate with TRPC3/6 channels
in a manner dependent on phosphorylation by PKC (21), we
speculated that prevention of PKC phosphorylation may disrupt
TRPC/CaN complexes. Indeed, immunoprecipitation experi-
ments in HEK293 cells conrmed that the PKC inhibitor prevents
association of CaN into TRPC3 complexes along with reduction
of threonine phosphorylation of the channel protein (Fig. 4).
Alternatively, a mutant that is defective in PKC-γmediated in-
hibitory modulation, i.e., T573A corresponding to the murine
TRPC3-moonwalker (Mwk) mutation, was expressed in HEK293
and HL-1 cells. It is of note that overexpression of the phos-
phorylation-decient TRPC3-Mwk by itself was barely tolerated
by the cells, presumably due to a gain in function leading to Ca
2+
overload. Therefore, we transfected cells to overexpress the
T573A mutant along with wild-type TRPC3 (DNA ratio 3:1). This
transfection resulted in a heteromeric TRPC3 conductance larger
than that generated by wild-type TRPC3 alone (Fig. S8) but es-
sentially tolerated by the host cells. The derived heteromers
appeared correctly targeted into the membrane as indicated by
uorescence microscopy. Interestingly, currents through TRPC3-
Mwk/TRPC3-WT channels were rather stable during continuous
agonist stimulation, most likely due to lack of inhibitory regula-
tion of the channels by PKC and recordings in Na
+
-free extra-
cellular solution conrmed Ca
2+
permeability of the channels.
Either treatment of cells expressing TRPC3-WT with
GF109203X (2 μM) or transfection of cells with TRPC3-Mwk/
TRPC3-WT produced a similar cellular phenotype, displaying
BCDA
Fig. 2. Receptor-stimulated Ca
2+
inux as well as NFAT translocation are impaired in HEK293 cells expressing the Ca
2+
impermeable TRPC3-E630Q or the
impermeant TRPC3-E630K, compared with cells expressing wild-type TRPC3. (A) Representative traces of fura-2 Ca
2+
-imaging experiments. Cells were stim-
ulated by 100 μM carbachol (arrow). (B) Mean Δratio values (±SEM, n>40) derived from fura-2 Ca
2+
-imaging. Black bars indicate the basal (unstimulated)
Ca
2+
entry at indicated transfections. (C) Mean nuclear/cytosol uorescence intensity ratio (±SEM, n>11) of HEK293 cells expressing GFP-NFAT and the
respective channel protein after stimulation and application of the same protocol as used in fura-2 experiments. Black bar (basal) represents mean nuclear/
cytosol uorescence intensity ratio in HEK293 cells transfected with GFP-NFAT only. (Band C) Asterisks indicate statistically signicant of difference to TRPC3-
WTexpressing cells. (D) Representative uorescence images recorded in GFP-NFAT translocation experiments shown at Left. Positions of nuclei are indicated
by arrowheads. Nucleus/cytosol uorescence ratios of example images are indicated.
10558
|
www.pnas.org/cgi/doi/10.1073/pnas.1106183108 Poteser et al.
large-agonistinduced Ca
2+
entry signals that were not accom-
panied by signicant NFAT nuclear accumulation (Fig. 5). This
transcriptionally silentCa
2+
entry into HL-1 cells was for a large
part mediated by voltage-gated L-type Ca
2+
channels as indicated
by sensitivity to nifedipine (3 μM). Our results demonstrate dis-
ruption of transcriptional TRPC3 signaling in response to reduced
A
B
C
D
Fig. 3. Ca
2+
entry through TRPC3 is critical for activation of the calcineurin/NFAT pathway in HL-1 atrial myocytes. HL-1 cells were transfected with vector
control (A), TRPC3-WT (B), or the indicated pore mutant (Cand D). (Left) Representative traces of fura-2 imaging in cells at basal conditions (unstimulated +
Ca
2+
readdition) and stimulated with 100 nM endothelin (+ ET-1, arrow) in the absence or presence of 3 μM nifedipine (+ Nif). (Center Left) Mean fura-2 Δ
ratio values (±SEM, n>20). (Center Right) Mean nuclear/cytosolic NFAT-GFP uorescence ratio (±SEM, n>8) in unstimulated HL-1 cells (basal), HL-1 cells
stimulated with 100 nM endothelin (+ ET-1) in the absence and presence of 3 μM nifedipine (+ Nif) and application of the same protocol as used in fura-2
experiments. Asterisks indicate statistically signicant inhibition by nifedipine. (Right) Representative images of NFAT-localization before stimulation and
Ca
2+
readdition (control), after Ca
2+
readdition (basal), and after stimulation by endothelin and subsequent Ca
2+
readdition (+ ET-1) in the absence and
presence of 3 μM nifedipine (+ Nif). Positions of nuclei are indicated by arrows. Nucleus/cytosol uorescence ratios of example images are indicated.
Poteser et al. PNAS
|
June 28, 2011
|
vol. 108
|
no. 26
|
10559
CELL BIOLOGY
PKC-dependent phosphorylation of the channel or as a conse-
quence of the TRPC3-Mwk mutation. This fact may be consid-
ered as part of the molecular mechanism underlying the moon-
walker pathophysiology (23).
Impaired PKC regulation of cardiac TRPC3 is shown to result in
uncoupling from the NFAT pathway without disrupting the link-
age between TRPC3 and global myocyte Ca
2+
via voltage-gated
Ca
2+
entry. We provide evidence that TRPC3-CaN/NFAT
sign aling takes place in a restricted microdomain and requiresboth
direct Ca
2+
permeation through the TRPC3 pore as well as CaN
targeting into the signal complex. Cardiac TRPC3 complexes are
shown to produce Ca
2+
signals both via direct Ca
2+
transport and
by control of voltage-dependent Ca
2+
entry. Our results demon-
strate the ability of cardiac TRPC3 channels to switch in a phos-
phorylation-dependent manner between a transcriptionally active
and a transcriptionally silent signaling mode (Fig. 6). Because
TRPC3 is likely to change in expression along with other TRPC
species during pathophysiologic stress, the formation of divergent
heteromeric TRPC3 channel complexes may be anticipated. The
observed dominantnegative effect of the phosphorylation-decient
moonwalker mutation on CaN/NFAT activation suggests a prom-
inent role of phosphorylation in maintenance of transcriptionally
active TRPC complexes in the heart. Situations of hampered
PKC phopshorylation or promoted dephosphorylation may con-
vert TRPC complexes into a cardiac Ca
2+
signaling unit that is
functionally isolated from the CaN/NFAT activation pathway.
Our ndings highlight the key role of nonselective TRPC
channels in the control of transcriptional programs and extend
this concept by demonstrating a pivotal role of Ca
2+
transport
trough the TRP pore structure along with a unique phosphory-
lation-dependent molecular switch that allows efcient control of
cardiac gene transcription by neurotransmitters and hormones.
Materials and Methods
Homology Modeling. For details on sequence alignment, homology modeling
and model evaluation, see SI Materials and Methods.
A
BC
Fig. 4. GF109203X inhibits phosphorylation of TRPC3
and its association with calcineurin. HEK-293 cells
expressing HA-tagged TRPC3 were incubated with or
without 2 μM GF109203X (GFX) and lysed and sub-
jected to SDS/PAGE and immunoprecipitation. (ALeft)
Total HEK-cell lysates were immunoprecipitated by
using an anti-HA antibody and immunoblotted with
an anti-phospho-threonine antibody. (A Right) Bars
representing the densitometric analysis of phospho-
threonine-immunoreactivity. Mean values are given
for carbachol-stimulated cells in the absence and pres-
ence of GFX (±SEM, n= 4). Asterisk indicates statisti-
cally signicant differences. (B) Stripped membranes
were immunoblotted again by using an anti-HA anti-
body (Lower). Proteins detected in total cell lysates
(lane 1, Input), immunocomplexes (lane 2, IP-HA-C3)
and lysates precipitated only with beads (lane 3, Ctrl.).
(C) Coimmunoprecipitations of cell homogenates using
antibodies against the HA-tag and calcineurin, and
immunoblotted against calcineurin. Proteins detected
in total cell lysates (lane 1, Input), immunocomplexes
(lane 2, IP-HA-C3; lane 3, IP-CN), and lysates pre-
cipitated only with beads (lane 4, Ctrl.).
ABCD
Fig. 5. Phosphorylation of threonine 573 of the TRPC3 channel protein is essential for the activation of the calcineurin/NFAT pathway in HL-1 atrial myocytes. (A)
Representative traces of fura-2 Ca
2+
-imaging experiments in cells transfected with either TRPC3-WT or TRPC3-T573A and TRPC3-WT (DNA ratio 3:1, MWK/TRPC3-
WT), stimulated by 100 nM endothelin (arrow) and in the absence or presence of 3 μM nifedipine (+ Nif) or 2 μM GF109203X (+ GFX), as indicated. (B) Mean Δratio
values (±SEM, n>30) of fura-2 Ca
2+
-imaging experiments. Asterisks indicate statistically signicant nifedipine-induced inhibition. (C) Mean nucleus/cytosol
uorescence intensity ratio (±SEM, n>9) in HL-1 cells expressing GFP-NFAT and the respective (mutant) channel protein after stimulation and application of the
same protocol as used in fura-2 experiments. (Band C) Asterisks indicate statistically signicant difference to TRPC3-WTexpressing cells in the absence of
inhibitors (white bar). (D) Representative uorescence images recorded in GFP-NFAT translocation experiments. Individual nucleus/cytosol uorescence ratios
are given, and positions of nuclei are indicated by arrows.
10560
|
www.pnas.org/cgi/doi/10.1073/pnas.1106183108 Poteser et al.
DNA and Mutagenesis. Site-directed mutagenesis was performed with stan-
dard protocols. Details on cDNA constructs and cloning procedures are
provided in SI Materials and Methods.
Cell Culture and Transfection. Cell lines were cultured at 37 °C and 5% CO
2
.For
HEK293 cells DMEM (Invitrogen) supplied with 10% FCS and for HL-1 atrial
myocytes Claycomb medium (Sigma) supplied with 100 μM norepinephrin,
4mML-glutamin and 10% FCS were used. Lipofection was used for genetrans-
fer; for details on DNA amounts and reagents, see SI Materials and Methods.
Electrophysiology. Standard patch clamp protocols were used (SI Materials
and Methods). Standard bath solutions contained 140 or 0 mM NaCl, 0 or
140 mM NMDG, 2 mM MgCl
2
, 10 mM glucose, 10 mM Hepes, 2 or 0 mM
CaCl
2
, and 0 or 2 mM BaCl
2
at pH adjusted to 7.4 with NaOH or NMDG. Pi-
pette solution contained 120 mM cesium methanesulfonate, 20 mM CsCl,
15 mM Hepes, 5 mM MgCl
2
, and 3 mM EGTA, at pH adjusted to pH 7.3 with
CsOH. For delineation of Ca
2+
permeability of TRPC3 mutants, a bath solu-
tion containing 132 mM NMDG, 2 mM MgCl
2
, 10 mM Glucose, 10 mM Hepes,
3 mM CaCl
2
, 7 mM Ca-Gluconate, at pH adjusted to 7.4 with methanesulfonic
acid and a pipette solution composed of 140 mM cesium methanesulfonate,
15 mM Hepes, 5 mM MgCl
2
, and 10 mM BAPTA at pH 7.3 was used.
Measurement of NFAT-Translocation. Cells were transfected to express an N-
terminally GFP-tagged NFATc1 fusion (15) and plated on coverslips. For
buffers and solutions see SI Materials and Methods. Agonists as well as
inhibitors (Pyr3, GFX109203, nifedipine, or KB-R7943) remained present
continuously after administration. GFP-NFAT translocation was monitored
(488 nm excitation) with standard uorescence microscopy (Zeiss Axiovert
equipped with Coolsnap HQ). GFP-NFAT and YFP-TRPC3-WT/-mutant uo-
rescence were discriminated by specic cellular localization. Nuclear/cytosol
uorescence intensity ratios of cells were calculated with ImageJ software.
Measurement of Intracellular Ca
2+
Signaling. For details on fura-2 calcium
imaging experiments see SI Materials and Methods.
Immunoprecipation. In short, protein-A- or protein-G-bead-precleared
supernatants of lysates from stimulated HEK293 cells were incubated with
precipitating antibody overnight. After the addition of respective protein-A-
or protein-G-beads, washing, and denaturation in Lämmli buffer, the im-
munocomplexes were separated by SDS/PAGE and subjected to Western
blotting. See SI Materials and Methods for details.
Reagents. Chemicals, reagents, and antibodies were purchased from Sigma
Aldrich. KB-R7943 and GF109203X were from Tocris Biosciences. The TRPC3
pore blocker Pyr3 was synthesized as published (16).
Statistics. Data are presented as mean values ±SEM and was tested for
statistical signicance by using the Student ttest (*P<0.05).
ACKNOWLEDGMENTS. We thank Dr. R. Kehlenbach for providing the GFP-
NFAT construct and Mrs. R. Schmidt for excellent technical assistance. This
work was supported by FWF (Austrian Science Fund) Grant P21925-B19
(to K.G.), P22565 (to C.R.), and DK+Metabolic and Cardovascular Disease
Grant W2126-B18.
1. KraniasEG, Bers DM (2007)Calcium and cardiomyopathies. Subcell Biochem45:523537.
2. Bush EW, et al. (2006) Canonical transient receptor potential channels promote
cardiomyocyte hypertrophy through activation of calcineurin signaling. J Biol Chem
281:3348733496.
3. Kuwahara K, et al. (2006) TRPC6 ful lls a calcineurin signaling circuit during
pathologic cardiac remodeling. J Clin Invest 116:31143126.
4. Nakayama H, Wilkin BJ, Bodi I, Molkentin JD (2006) Calcineurin-dependent cardio-
myopathy is activated by TRPC in the adult mouse heart. FASEB J 20:16601670.
5. Montell C, Birnbaumer L, Flockerzi V (2002) The TRP channels, a remarkably
functional family. Cell 108:595598.
6. Owsianik G, Talavera K, Voets T, Nilius B (2006) Permeation and selectivity of TRP
channels. Annu Rev Physiol 68:685717.
7. Onohara N, et al. (2006) TRPC3 and TRPC6 are essential for angiotensin II-induced
cardiac hypertrophy. EMBO J 25:53055316.
8. Rosker C, et al. (2004) Ca(2+) signaling by TRPC3 involves Na(+) entry and local
coupling to the Na(+)/Ca(2+) exchanger. J Biol Chem 279:1369613704.
9. Eder P, Groschner K (2008) TRPC3/6/7: Topical aspects of biophysics and
pathophysiology. Channels (Austin) 2:9499.
10. Kamouchi M, et al. (1999) Properties of heterologously expressed hTRP3 channels in
bovine pulmonary artery endothelial cells. J Physiol 518:345358.
11. Eder P, et al. (2007) Phospholipase C-dependent control of cardiac calcium
homeostasis involves a TRPC3-NCX1 signaling complex. Cardiovasc Res 73:111119.
12. Molkentin JD (2006) Dichotomy of Ca2+ in the heart: Contraction versus intracellular
signaling. J Clin Invest 116:623626.
13. Notredame C, Higgins DG, Heringa J (2000) T-Coffee: A novel method for fast and
accurate multiple sequence alignment. J Mol Biol 302:205217.
14. Liu CH, et al. (2007) In vivo identication and manipulation of the Ca2+ selectivity
lter in the Drosophila transient receptor potential channel. J Neurosci 27:604615.
15. Nilius B, et al. (2005) The selectivity lter of the cation channel TRPM4. J Biol Chem
280:2289922906.
16. Voets T, Janssens A, Droogmans G, Nilius B (2004) Outer pore architecture of a
Ca2+-selective TRP channel. J Biol Chem 279:1522315230.
17. Garcia-Martinez C, Morenilla-Palao C, Planells-Cases R, Merino JM, Ferrer-Montiel A
(2000) Identication of an aspartic residue in the P-loop of the vanilloid receptor that
modulates pore properties. J Biol Chem 275:3255232558.
18. Seth M, et al. (2009) TRPC1 channels are critical for hypertrophic signaling in the
heart. Circ Res 105:10231030.
19. Kiyonaka S, et al. (2009) Selective and direct inhibition of TRPC3 channels underlies
biological activities of a pyrazole compound. Proc Natl Acad Sci USA 106:54005405.
20. Sinkins WG, Goel M, Estacion M, Schilling WP (2004) Association of immunophilins
with mammalian TRPC channels. J Biol Chem 279:3452134529.
21. Kim JY, Saffen D (2005) Activation of M1 muscarinic acetylcholine receptors
stimulates the formation of a multiprotein complex centered on TRPC6 channels.
J Biol Chem 280:3203532047.
22. Trebak M, et al. (2005) Negative regulation of TRPC3 channels by protein kinase C-
mediated phosphorylation of serine 712. Mol Pharmacol 67:558563.
23. Becker EB, et al. (2009) A point mutation in TRPC3 causes abnormal Purkinje cell
development and cerebellar ataxia in moonwalker mice. Proc Natl Acad Sci USA 106:
67066711.
Fig. 6. Phosporylation of TRPC3 at position 573 (via PLC) enables Ca
2+
/
calmodulin/calcineurin (Ca
2+
, Cm, CaN)-dependent activation upon receptor-
activated Ca
2+
entry through TRPC3. Dephosporylation of position 573 turns
TRPC transcriptionally silentwhile enhancing itsgeneral activity.L-type channels
are controlled by TRPC3-induced changes in membrane potential, providing
only negligible effects on Ca
2+
-induced NFAT activation in HL-1 atrial myocytes.
Poteser et al. PNAS
|
June 28, 2011
|
vol. 108
|
no. 26
|
10561
CELL BIOLOGY
Correction
CELL BIOLOGY
Correction for PKC-dependent coupling of calcium permeation
through transient receptor potential canonical 3 (TRPC3) to
calcineurin signaling in HL-1 myocytes,by Michael Poteser,
Hannes Schleifer, Michaela Lichtenegger, Michaela Schern-
thaner, Thomas Stockner, C. Oliver Kappe, Toma N. Glasnov,
Christoph Romanin, and Klaus Groschner, which appeared in
issue 26, June 28, 2011, of Proc Natl Acad Sci USA (108:10556
10561; rst published June 8, 2011; 10.1073/pnas.1106183108).
The authors note that Figs. 2, 3, and 5 appeared incorrectly.
The corrected gures and their corresponding legends appear
below.
ABCD
Fig. 2. Receptor-stimulated Ca
2+
inux as well as NFAT translocation are impaired in HEK293 cells expressing the Ca
2+
impermeable TRPC3-E630Q or the
impermeant TRPC3-E630K, compared with cells expressing wild-type TRPC3. (A) Representative traces of fura-2 Ca
2+
-imaging experiments. Cells were stimu-
lated by 100 μM carbachol (arrow). (B) Mean Δratio values (±SEM, n>40) derived from fura-2 Ca
2+
-imaging. Black bars indicate the basal (unstimulated) Ca
2+
entry at indicated transfections. (C) Mean nuclear/cytosol uorescence intensity ratio (±SEM, n>11) of HEK293 cells expressing GFP-NFAT and the respective
channel protein after stimulation and application of the same protocol as used in fura-2 experiments. Black bar (basal) represents mean nuclear/cytosol
uorescence intensity ratio in HEK293 cells transfected with GFP-NFAT only. (Band C) Asterisks indicate statistically signicance of difference to TRPC3-WT
expressing cells. (D) Representative uorescence images recorded in GFP-NFAT translocation experiments shown at Left. Positions of nuclei are indicated by
arrowheads. Nucleus/cytosol uorescence ratios of example images are indicated.
1387613878
|
PNAS
|
August 16, 2011
|
vol. 108
|
no. 33 www.pnas.org
A
B
C
D
Fig. 3. Ca
2+
entry through TRPC3 is critical for activation of the calcineurin/NFAT pathway in HL-1 atrial myocytes. HL-1 cells were transfected with vector
control (A), TRPC3-WT (B), or the indicated pore mutant (Cand D). (Left) Representative traces of fura-2 imaging in cells at basal conditions (unstimulated +
Ca
2+
readdition) and stimulated with 100 nM endothelin (+ ET-1, arrow) in the absence or presence of 3 μM nifedipine (+ Nif). (Center Left) Mean fura-2 Δ
ratio values (±SEM, n>20). (Center Right) Mean nuclear/cytosolic NFAT-GFP uorescence ratio (±SEM, n>8) in unstimulated HL-1 cells (basal), HL-1 cells
stimulated with 100 nM endothelin (+ ET-1) in the absence and presence of 3 μM nifedipine (+ Nif) and application of the same protocol as used in fura-2
experiments. Asterisks indicate statistically signicant inhibition by nifedipine. (Right) Representative images of NFAT-localization before stimulation and Ca
2+
readdition (control), after Ca
2+
readdition (basal), and after stimulation by endothelin and subsequent Ca
2+
readdition (+ ET-1) in the absence and presence of
3μM nifedipine (+ Nif). Positions of nuclei are indicated by arrowheads. Nucleus/cytosol uorescence ratios of example images are indicated.
PNAS
|
August 16, 2011
|
vol. 108
|
no. 33
|
13877
CORRECTION
www.pnas.org/cgi/doi/10.1073/pnas.1111388108
ABCD
Fig. 5. Phosphorylation of threonine 573 of the TRPC3 channel protein is essential for the activation of the calcineurin/NFAT pathway in HL-1 atrial myocytes.
(A) Representative traces of fura-2 Ca
2+
-imaging experiments in cells transfected with either TRPC3-WT or TRPC3-T573A and TRPC3-WT (DNA ratio 3:1, MWK/
TRPC3-WT), stimulated by 100 nM endothelin (arrow) and in the absence or presence of 3 μM nifedipine (+ Nif) or 2 μM GF109203X (+ GFX), as indicated. (B)
Mean Δratio values (±SEM, n>30) of fura-2 Ca
2+
-imaging experiments. Asterisks indicate statistically signicant nifedipine-induced inhibition. (C) Mean
nucleus/cytosol uorescence intensity ratio (±SEM, n>9) in HL-1 cells expressing GFP-NFAT and the respective (mutant) channel protein after stimulation and
application of the same protocol as used in fura-2 experiments. (Band C) Asterisks indicate statistically signicant difference to TRPC3-WTexpressing cells in
the absence of inhibitors (white bar). (D) Representative uorescence images recorded in GFP-NFAT translocation experiments. Individual nucleus/cytosol
uorescence ratios are given, and positions of nuclei are indicated by arrowheads.
13878
|
www.pnas.org
... The homotetrameric TRPC3 channel displays only modest selectivity for Ca 2+ ions over Na + (PCa/PNa = 1.6) [17] and exhibits a higher degree of basal activity than the closely related TRPC6/7 channels. The slight divalent cation selectivity of TRPC3 arises due to a negatively charged glutamate residue in its selectivity filter, E630 [18,19] (UniProt isoform 3/Q13507-3). TRPC3/6/7 are activated downstream of G-protein coupled receptor (GPCR) subunit with helices coloured as in (A), PDB code 6CUD. ...
... The homotetrameric TRPC3 channel displays only modest selectivity for Ca 2+ ions over Na + (P Ca /P Na = 1.6) [17] and exhibits a higher degree of basal activity than the closely related TRPC6/7 channels. The slight divalent cation selectivity of TRPC3 arises due to a negatively charged glutamate residue in its selectivity filter, E630 [18,19] (UniProt isoform 3/Q13507-3). TRPC3/6/7 are activated downstream of G-protein coupled receptor (GPCR) activation by diacylglycerol (DAG), a product of phospholipase C (PLC) activation, in a membrane-delimited manner [20]. ...
... As such, the channel has been identified as a potential player in, and therapeutic target for, cardiac pathologies including hypertrophy, atrial fibrillation and arrhythmias [32,33]. Ca 2+ entry through TRPC3 has been linked to multiple downstream signalling pathways, such as activation of angiotensin II-and noradrenaline-induced nuclear factor of activated T cells (NFAT) [18]. Activation of the NFAT/calcineurin pathway via TRPC3 in cardiac myocytes is associated with pathologies such as arrhythmia and hypertrophy [32]. ...
Article
Full-text available
Canonical transient receptor potential 3 (TRPC3) channel is a non-selective cation permeable channel that plays an essential role in calcium signalling. TRPC3 is highly expressed in the brain and also found in endocrine tissues and smooth muscle cells. The channel is activated directly by binding of diacylglycerol downstream of G-protein coupled receptor activation. In addition, TRPC3 is regulated by endogenous factors including Ca2+ ions, other endogenous lipids, and interacting proteins. The molecular and structural mechanisms underlying activation and regulation of TRPC3 are incompletely understood. Recently, several high-resolution cryogenic electron microscopy structures of TRPC3 and the closely related channel TRPC6 have been resolved in different functional states and in the presence of modulators, coupled with mutagenesis studies and electrophysiological characterisation. Here, we review the recent literature which has advanced our understanding of the complex mechanisms underlying modulation of TRPC3 by both endogenous and exogenous factors. TRPC3 plays an important role in Ca2+ homeostasis and entry into cells throughout the body, and both pathological variants and downstream dysregulation of TRPC3 channels have been associated with a number of diseases. As such, TRPC3 may be a valuable therapeutic target, and understanding its regulatory mechanisms will aid future development of pharmacological modulators of the channel.
... Here we set out to further shed light on the nanojunctional signaling function of TRPC3 by utilizing, on the one hand, previously established knowledge on mutational modification of the TRPC permeation pathway [29,30] and, on the other hand, molecular targeting of a genetically encoded Ca 2+ biosensor to the channel structure to monitor Ca 2+ signals at the TRPC channels' local signaling domain [31]. We provide evidence that TRPC3, as a primary lipid (DAG)-gated cation channel, governs the temporal structure of junctional Ca 2+ signals by local and dynamic communications with IP 3 Rs. ...
... This approach was expected to provide further insight into the role of Ca 2+ permeation through TRPC3 channels in nanojunctional signaling. For defined manipulation of TRPC3 Ca 2+ entry, we introduced several previously characterized point mutations, which generate non-functional tetrameric complexes (E630K), restrict channel permeability to monovalent ions (E630Q) [29], or impair activation of the channel by endogenous DAGs (G652A) [30]. ...
Article
Full-text available
Communication between TRPC channels and IP3 receptors (IP3R) is considered pivotal in the generation of spatiotemporal Ca²⁺ signalling patterns. Here we revisited the role of TRPC3-IP3R coupling for local Ca²⁺ signaling within TRPC3-harbouring micro/nanodomains using R-GECO as a reporter, fused to the channel´s C-terminus. Cytoplasmic Ca²⁺ changes at TRPC3 originated from both the entry of Ca²⁺ through the TRPC channel and Ca²⁺ mobilization via IP3R. Local Ca²⁺ changes at TRPC3 channels expressed in HEK293 cells were predominantly biphasic with IP3R-dependent initial Ca²⁺ transients, while exclusively monophasic signals were recorded when all three IP3R isoforms were lacking. Abrogation of Ca²⁺ entry through TRPC3 by point mutations, which impair Ca²⁺ permeability (E630Q), cation permeation (E630K), or DAG sensitivity (G652A), promoted microdomain Ca²⁺ oscillations. Ca²⁺ signals at E630Q, E630K, and G652A channels featured initial Ca²⁺ transients along with oscillatory activity. Similarly, when extracellular Ca²⁺ was omitted, IP3R-mediated Ca²⁺ transients and Ca²⁺ oscillations were promoted at the cytoplasmic face of wild-type TRPC3 channels. By contrast, oscillations, as well as initial Ca²⁺ transients, were virtually lacking, when the TRPC3 channels were sensitized by preexposure to low-level PLC activity. TIRF imaging provided evidence for dynamic colocalization of TRPC3 and IP3R. We suggest that TRPC3-mediated Ca²⁺ entry controls IP3R activity at ER-PM junctions to determine Ca²⁺ signaling signatures and enable specificity of downstream signaling.
... Thus, to establish our high-content quantitative stretch-Ca 2+ signalling platform in 2D adherent cardiac cells as a step before turning to more elaborate 'cell-in-a-gel' adult cardiomyocyte settings, we chose the use of HL-1 cells here. Apart from Piezo1 [12], HL-1 cells have also been shown to express mechanosensitive TRPM4 [24] and TRPC3 [36] channels. However, TRP channels have recently been reported to be insensitive to direct membrane stretch activation [32], thus, our IsoStretcher setting is mainly tracking Piezo1-mediated Ca 2+ fluctuations. ...
Article
Full-text available
The importance of mechanosensory transduction pathways in cellular signalling has prominently come to focus in the last decade with the discovery of the Piezo ion channel family. Mechanosignaling involving Piezo1 ion channels in the function of the heart and cardiovascular system has only recently been identified to have implications for cardiovascular physiology and pathophysiology, in particular for heart failure (i.e., hypertrophy or dilative cardiomyopathy). These results have emphasized the need for higher throughput methods to study single-cell cardiovascular mechanobiology with the aim of identifying new targets for therapeutic interventions and stimulating the development of new pharmacological agents. Here, we present a novel method to assess mechanosignaling in adherent cardiac cells (murine HL-1 cell line) using a combination of isotropic cell stretch application and simultaneous Ca²⁺ fluorescence readout with quantitative analysis. The procedure implements our IsoStretcher technology in conjunction with a single-cell- and population-based analysis of Ca²⁺ signalling by means of automated image registration, cell segmentation and analysis, followed by automated classification of single-cell responses. The method is particularly valuable for assessing the heterogeneity of populations with distinct cellular responses to mechanical stimulation and provides more user-independent unbiased drug response classifications.
... It is important to note that the readout in the experiments was global cytoplasmic Ca 2+ , while for many TRPC signaling cascades local Ca 2+ changes and specifically the temporal pattern of local Ca 2+ is of critical importance. It appears therefore plausible to speculate that the observed modulation of TRPC3 kinetics, generated by DAGinduced sensitization, is likely to affect all cellular functions of TRPC3 that rely on localized Ca 2+ signal patterns such as Ca 2+ transcription coupling (Poteser et al, 2011) or neuronal frequency modulation (Neuner et al, 2015;Tiapko & Groschner, 2021). ...
Article
Full-text available
Coordination of lipids within transient receptor potential canonical channels (TRPCs) is essential for their Ca2+ signaling function. Single particle cryo-EM studies identified two lipid interaction sites, designated L1 and L2, which are proposed to accommodate diacylglycerols (DAGs). To explore the role of L1 and L2 in TRPC3 function, we combined structure-guided mutagenesis and electrophysiological recording with molecular dynamics (MD) simulations. MD simulations indicate rapid DAG accumulation within both L1 and L2 upon its availability within the plasma membrane. Electrophysiological experiments using a photoswitchable DAG-probe reveal potentiation of TRPC3 currents during repetitive activation by DAG. Importantly, initial DAG exposure generates a subsequently sensitized channel state that is associated with significantly faster activation kinetics. TRPC3 sensitization is specifically promoted by mutations within L2, with G652A exhibiting sensitization at very low levels of active DAG. We demonstrate the ability of TRPC3 to adopt a closed state conformation that features partial lipidation of L2 sites by DAG and enables fast activation of the channel by the phospholipase C-DAG pathway.
... This was confirmed by a study in which overexpression of TRPC3 in HeLa cells led to a substantial reduction in cytosolic Ca 2+ . In contrast, cytosolic Ca 2+ increased in cells expressing the mutant TRPC3 (E630Q) [75]. It has been described that TRPC3 favors Ca 2+ entry into the matrix when the concentration of this divalent cation is high (≥50 µM) [63]. ...
Article
Full-text available
Calcium is used in many cellular processes and is maintained within the cell as free calcium at low concentrations (approximately 100 nM), compared with extracellular (millimolar) concentrations, to avoid adverse effects such as phosphate precipitation. For this reason, cells have adapted buffering strategies by compartmentalizing calcium into mitochondria and the endoplasmic reticulum (ER). In mitochondria, the calcium concentration is in the millimolar range, as it is in the ER. Mitochondria actively contribute to buffering cellular calcium, but if matrix calcium increases beyond physiological demands, it can promote the opening of the mitochondrial permeability transition pore (mPTP) and, consequently, trigger apoptotic or necrotic cell death. The pathophysiological implications of mPTP opening in ischemia-reperfusion, liver, muscle, and lysosomal storage diseases, as well as those affecting the central nervous system, for example, Parkinson’s disease (PD), Alzheimer’s disease (AD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS) have been reported. In this review, we present an updated overview of the main cellular mechanisms of mitochondrial calcium regulation. We specially focus on neurodegenerative diseases related to imbalances in calcium homeostasis and summarize some proposed therapies studied to attenuate these diseases.
... The expression of this mutant, in contrast to the expression of the wild-type TRPC3, had virtually no effect on the IP 3 -induced Ca 2+ -release ( Fig. 4g and Supplementary Fig. 4d). In addition, the inhibitory effect of TRPC3 overexpression on the IP 3 R-mediated Ca 2+ release was only slightly reduced when the pore channel mutants, E630Q ("Ca 2+ permeation-deficient") or E630K ("pore-dead") 45 were overexpressed instead of wild-type TRPC3 ( Fig. 4h and Supplementary Fig. 4e), indicating that TRPC3 exerts its inhibitory effects via direct interaction with IP 3 R. ...
Article
Full-text available
Cellular senescence is implicated in a great number of diseases including cancer. Although alterations in mitochondrial metabolism were reported as senescence drivers, the underlying mechanisms remain elusive. We report the mechanism altering mitochondrial function and OXPHOS in stress-induced senescent fibroblasts. We demonstrate that TRPC3 protein, acting as a controller of mitochondrial Ca2+ load via negative regulation of IP3 receptor-mediated Ca2+ release, is down regulated in senescence regardless of the type of senescence inducer. This remodelling promotes cytosolic/mitochondrial Ca2+ oscillations and elevates mitochondrial Ca2+ load, mitochondrial oxygen consumption rate and oxidative phosphorylation. Re-expression of TRPC3 in senescent cells diminishes mitochondrial Ca2+ load and promotes escape from OIS-induced senescence. Cellular senescence evoked by TRPC3 downregulation in stromal cells displays a proinflammatory and tumour-promoting secretome that encourages cancer epithelial cell proliferation and tumour growth in vivo. Altogether, our results unravel the mechanism contributing to pro-tumour behaviour of senescent cells. Mitochondrial Ca2+ homeostasis is reported to influence cellular senescence. Here the authors show that TRPC3 limits senescence by inhibiting IP3R-mediated Ca2+ release and ER mitochondria Ca2+ transfer and that the downregulation of TRPC3 in stromal cells affects SASP production and tumour progression.
... NFAT is a transcription factor activated by Ca 2+ , which is mediated through the Ca 2+ -binding proteins calmodulin and calcineurin [11], where TRPC channels could mediate Ca 2+ entry [20]. Previous research has documented that Ca 2+ entry through other TRPC channels, including TRPC1, TRPC3, and TRPC6, could accelerate NFAT-dependent gene transcription [20,21]. A putative NFAT binding site can be found in the 5'-flanking sequence of MDR1. ...
Article
Full-text available
P-glycoprotein (P-gp) acts as a pump to transport cytotoxic drugs out of cells and is upregulated in cancer cells. Suppressing the expression of P-gp is an effective strategy to overcome multidrug resistance in cancer chemotherapy. Temozolomide (TMZ) is the recommended drug for the standard treatment of patients with glioblastoma, but its clinical application is restricted due to drug resistance. Transient receptor potential channel-5 (TRPC5), a Ca²⁺-permeable channel, has been attributed to a different drug resistance mechanism except DNA repair system; therefore, we aimed to elucidate the mechanism regarding the role of TRPC5 in TMZ resistance. TRPC5 and P-glycoprotein (P-gp) are upregulated in TMZ-resistant glioblastoma cell lines. The downregulation of TRPC5 inhibited P-gp expression and led to a significant reversal of TMZ resistance in TMZ-resistant cell lines. TRPC5-siRNA restricted the growth of tumour xenografts in an athymic nude mouse model of TMZ-resistant cells. In specimens from patients with recurrent glioblastoma, TRPC5 was found to be highly expressed, accompanied by the upregulation of P-gp expression. The nuclear factor of activated T cell isoform c3 (NFATc3), which acts as a transcriptional factor, bridges TRPC5 activity to P-gp induction. In conclusion, these results demonstrate the functional role of the TRPC5-NFATc3-P-gp signalling pathway in TMZ resistance in glioblastoma cells.
... The participation of TRPC1, C3, C4, C5 and C6 in SOCE has been examined in adult rodent cardiac myocytes, in cardiac cell line and in neonatal rat ventricle myocytes, using RNA silencing, neutralizing antibodies, or dominant-negative transgenic micetransgenic mice expressing that are dominant-negative of for these proteins. [69][70][71][72][73] ...
Article
Ca2+ mishandling is a common feature in several cardiovascular diseases such as heart failure (HF). In many cases, impairment of key players in intracellular Ca2+ homeostasis has been identified as the underlying mechanism of cardiac dysfunction and cardiac arrhythmias associated with HF. In this review, we summarize primary novel findings related to Ca2+ mishandling in HF progression. HF research has increasingly focused on the identification of new targets and the contribution of their role in Ca2+ handling to the progression of the disease. Recent research studies have identified potential targets in three major emerging areas implicated in regulation of Ca2+ handling: the innate immune system, bone metabolism factors, and post-translational modification of key proteins involved in regulation of Ca2+ handling. Here, we describe their possible contributions to the progression of HF.
... Of note, in certain cellular settings TRPC3-generated Ca 2+ signals failed to serve as an upstream trigger signal for NFAT activation (32). Nonetheless, the coupling of TRPC activity to downstream effectors may be strictly dependent on their mode of activation (33). In a recent study, we were able to demonstrate the linkage of recombinant TRPC channels to the calcineurin (CaN)/NFAT pathway in HEK293 cells (26). ...
Article
Full-text available
Canonical transient receptor potential (TRPC) channels are considered as elements of the immune cell Ca²⁺ handling machinery. We therefore hypothesized that TRPC photopharmacology may enable uniquely specific modulation of immune responses. Utilizing a recently established TRPC3/6/7 selective, photochromic benzimidazole agonist OptoBI-1, we set out to test this concept for mast cell NFAT signaling. RBL-2H3 mast cells were found to express TRPC3 and TRPC7 mRNA but lacked appreciable Ca²⁺/NFAT signaling in response to OptoBI-1 photocycling. Genetic modification of the cells by introduction of single recombinant TRPC isoforms revealed that exclusively TRPC6 expression generated OptoBI-1 sensitivity suitable for opto-chemical control of NFAT1 activity. Expression of any of three benzimidazole-sensitive TRPC isoforms (TRPC3/6/7) reconstituted plasma membrane TRPC conductances in RBL cells, and expression of TRPC6 or TRPC7 enabled light-mediated generation of temporally defined Ca²⁺ signaling patterns. Nonetheless, only cells overexpressing TRPC6 retained essentially low basal levels of NFAT activity and displayed rapid and efficient NFAT nuclear translocation upon OptoBI-1 photocycling. Hence, genetic modification of the mast cells’ TRPC expression pattern by the introduction of TRPC6 enables highly specific opto-chemical control over Ca²⁺ transcription coupling in these immune cells.
Article
Full-text available
The hereditary ataxias are a complex group of neurological disorders characterized by the degeneration of the cerebellum and its associated connections. The molecular mechanisms that trigger the loss of Purkinje cells in this group of diseases remain incompletely understood. Here, we report a previously undescribed dominant mouse model of cerebellar ataxia, moonwalker (Mwk), that displays motor and coordination defects and loss of cerebellar Purkinje cells. Mwk mice harbor a gain-of-function mutation (T635A) in the Trpc3 gene encoding the nonselective transient receptor potential cation channel, type C3 (TRPC3), resulting in altered TRPC3 channel gating. TRPC3 is highly expressed in Purkinje cells during the phase of dendritogenesis. Interestingly, growth and differentiation of Purkinje cell dendritic arbors are profoundly impaired in Mwk mice. Our findings define a previously unknown role for TRPC3 in both dendritic development and survival of Purkinje cells, and provide a unique mechanism underlying cerebellar ataxia.
Article
Full-text available
Canonical transient receptor potential (TRPC) channels control influxes of Ca(2+) and other cations that induce diverse cellular processes upon stimulation of plasma membrane receptors coupled to phospholipase C (PLC). Invention of subtype-specific inhibitors for TRPCs is crucial for distinction of respective TRPC channels that play particular physiological roles in native systems. Here, we identify a pyrazole compound (Pyr3), which selectively inhibits TRPC3 channels. Structure-function relationship studies of pyrazole compounds showed that the trichloroacrylic amide group is important for the TRPC3 selectivity of Pyr3. Electrophysiological and photoaffinity labeling experiments reveal a direct action of Pyr3 on the TRPC3 protein. In DT40 B lymphocytes, Pyr3 potently eliminated the Ca(2+) influx-dependent PLC translocation to the plasma membrane and late oscillatory phase of B cell receptor-induced Ca(2+) response. Moreover, Pyr3 attenuated activation of nuclear factor of activated T cells, a Ca(2+)-dependent transcription factor, and hypertrophic growth in rat neonatal cardiomyocytes, and in vivo pressure overload-induced cardiac hypertrophy in mice. These findings on important roles of native TRPC3 channels are strikingly consistent with previous genetic studies. Thus, the TRPC3-selective inhibitor Pyr3 is a powerful tool to study in vivo function of TRPC3, suggesting a pharmaceutical potential of Pyr3 in treatments of TRPC3-related diseases such as cardiac hypertrophy.
Article
Full-text available
Nonselective and lipid-regulated cation channels formed by TRPC3, TRPC6 and TRPC7 have recently obtained attention in view their potential pathophysiological impact. It appears as a particular challenge to understand the molecular basis of TRPC3/6/7-related diseases in order to further delineate their value as therapeutic targets. The multifunctional nature of these channel proteins, based on a complex, versatile heteromerization potential along with highly promiscuous gating and mixed cation permeation properties, make these channels pivotal players in cellular signaling networks. Here we summarize newsworthy aspects of TRPC3/6/7 physiology and pathophysiology that open the view on novel therapeutic strategies based on these TRPCs as target structures.
Article
Full-text available
Vanilloid receptor subunit 1 (VR1) is a nonselective cation channel that integrates multiple pain-producing stimuli. VR1 channels are blocked with high efficacy by the well established noncompetitive antagonist ruthenium red and exhibit high permeability to divalent cations. The molecular determinants that define these functional properties remain elusive. We have addressed this question and evaluated by site-specific neutralization the contribution on pore properties of acidic residues located in the putative VR1 pore region. Mutant receptors expressed in Xenopus oocytes exhibited capsaicin-operated ionic currents akin to those of wild type channels. Incorporation of glutamine residues at Glu(648) and Glu(651) rendered minor effects on VR1 pore attributes, while Glu(636) slightly modulated pore blockade. In contrast, replacement of Asp(646) by asparagine decreased 10-fold ruthenium red blockade efficacy and reduced 4-fold the relative permeability of the divalent cation Mg(2+) with respect to Na(+) without changing the selectivity of monovalent cations. At variance with wild type channels and E636Q, E648Q, and E651Q mutant receptors, ruthenium red blockade of D646N mutants was weakly sensitive to extracellular pH acidification. Collectively, our results suggest that Asp(646) is a molecular determinant of VR1 pore properties and imply that this residue may form a ring of negative charges that structures a high affinity binding site for cationic molecules at the extracellular entryway.
Article
Full-text available
The TRP superfamily forms a functionally important class of cation channels related to the product of the Drosophila trp gene. TRP channels display an unusual diversity in activation mechanisms and permeation properties, but the basis of this diversity is unknown, as the structure of these channels has not been studied in detail. To obtain insight in the pore architecture of TRPV6, a Ca(2+)-selective member of the TRPV subfamily, we probed the dimensions of its pore and determined pore-lining segments using cysteine-scanning mutagenesis. Based on the permeability of the channel to organic cations, we estimated a pore diameter of 5.4 A. Mutating Asp(541), a residue involved in high affinity Ca(2+) binding, altered the apparent pore diameter, indicating that this residue lines the narrowest part of the pore. Cysteines introduced in a region preceding Asp(541) displayed a cyclic pattern of reactivity to Ag(+) and cationic methylthio-sulfanate reagents, indicative of a pore helix. The anionic methanethiosulfonate ethylsulfonate showed only limited reactivity in this region, consistent with the presence of a cation-selective filter at the outer part of the pore helix. Based on these data and on homology with the bacterial KcsA channel, we present the first structural model of a TRP channel pore. We conclude that main structural features of the outer pore, namely a selectivity filter preceded by a pore helix, are conserved between K(+) channels and TRPV6. However, the selectivity filter of TRPV6 is wider than that of K(+) channels and lined by amino acid side chains rather than main chain carbonyls.
Article
We describe a new method (T-Coffee) for multiple sequence alignment that provides a dramatic improvement in accuracy with a modest sacrifice in speed as compared to the most commonly used alternatives. The method is broadly based on the popular progressive approach to multiple alignment but avoids the most serious pitfalls caused by the greedy nature of this algorithm. With T-Coffee we pre-process a data set of all pair-wise alignments between the sequences. This provides us with a library of alignment information that can be used to guide the progressive alignment. Intermediate alignments are then based not only on the sequences to be aligned next but also on how all of the sequences align with each other. This alignment information can be derived from heterogeneous sources such as a mixture of alignment programs and/or structure superposition. Here, we illustrate the power of the approach by using a combination of local and global pair-wise alignments to generate the library. The resulting alignments are significantly more reliable, as determined by comparison with a set of 141 test cases, than any of the popular alternatives that we tried. The improvement, especially clear with the more difficult test cases, is always visible, regardless of the phylogenetic spread of the sequences in the tests.
Article
Cardiac muscle adapts to increase workload by altering cardiomyocyte size and function resulting in cardiac hypertrophy. G protein-coupled receptor signaling is known to govern the hypertrophic response through the regulation of ion channel activity and downstream signaling in failing cardiomyocytes. Transient receptor potential canonical (TRPC) channels are G protein-coupled receptor operated channels previously implicated in cardiac hypertrophy. Our objective of this study is to better understand how TRPC channels influence cardiomyocyte calcium signaling. Here, we used whole cell patch clamp of adult cardiomyocytes to show upregulation of a nonselective cation current reminiscent of TRPC channels subjected to pressure overload. This TRPC current corresponds to the increased TRPC channel expression noted in hearts of mice subjected to pressure overload. Importantly, we show that mice lacking TRPC1 channels are missing this putative TRPC current. Moreover, Trpc1(-)(/)(-) mice fail to manifest evidence of maladaptive cardiac hypertrophy and maintain preserved cardiac function when subjected to hemodynamic stress and neurohormonal excess. In addition, we provide a mechanistic basis for the protection conferred to Trpc1(-)(/)(-) mice as mechanosensitive signaling through calcineurin/NFAT, mTOR and Akt is altered in Trpc1(-)(/)(-) mice. From these studies, we suggest that TRPC1 channels are critical for the adaptation to biomechanical stress and TRPC dysregulation leads to maladaptive cardiac hypertrophy and failure.
Article
1. We combined patch clamp and fura-2 fluorescence methods to characterize human TRP3 (hTRP3) channels heterologously expressed in cultured bovine pulmonary artery endothelial (CPAE) cells, which do not express the bovine trp3 isoform (btrp3) but express btrp1 and btrp4. 2. ATP, bradykinin and intracellular InsP3 activated a non-selective cation current (IhTRP3) in htrp3-transfected CPAE cells but not in non-transfected wild-type cells. During agonist stimulation, the sustained rise in [Ca2+]i was significantly higher in htrp3-transfected cells than in control CPAE cells. 3. The permeability for monovalent cations was PNa > PCs approximately PK > PNMDG and the ratio PCa/PNa was 1.62 +/- 0.27 (n = 11). Removal of extracellular Ca2+ enhanced the amplitude of the agonist-activated IhTRP3 as well as that of the basal current The trivalent cations La3+ and Gd3+ were potent blockers of IhTRP3 (the IC50 for La3+ was 24.4 +/- 0.7 microM). 4. The single-channel conductance of the channels activated by ATP, assessed by noise analysis, was 23 pS. 5. Thapsigargin and 2,5-di-tert-butyl-1, 4-benzohydroquinone (BHQ), inhibitors of the organellar Ca2+-ATPase, failed to activate IhTRP3. U-73122, a phospholipase C blocker, inhibited IhTRP3 that had been activated by ATP and bradykinin. Thimerosal, an InsP3 receptor-sensitizing compound, enhanced IhTRP3, but calmidazolium, a calmodulin antagonist, did not affect IhTRP3. 6. It is concluded that hTRP3 forms non-selective plasmalemmal cation channels that function as a pathway for agonist-induced Ca2+ influx.
Article
TRP cation channels display an extraordinary assortment of selectivities and activation mechanisms, some of which represent previously unrecognized modes for regulating ion channels. Moreover, the biological roles of TRP channels appear to be equally diverse and range from roles in pain perception to male aggression.