ArticlePDF Available

Abstract and Figures

Transient receptor potential canonical 1 (TRPC1) protein is abundantly expressed in cardiomyocytes. While TRPC1 is supposed to be critically involved in cardiac hypertrophy, its physiological role in cardiomyocytes is poorly understood. We investigated the subcellular location of TRPC1 and its contribution to Ca2+ signaling in mammalian ventricular myocytes. Immunolabeling, three-dimensional scanning confocal microscopy and quantitative colocalization analysis revealed an abundant intracellular location of TRPC1 in neonatal rat ventricular myocytes (NRVM) and adult rabbit ventricular myocytes. TRPC1 was colocalized with intracellular proteins including sarco/endoplasmic reticulum Ca2+ ATPase 2 in the sarcoplasmic reticulum (SR). Colocalization with wheat germ agglutinin, which labels the glycocalyx and thus marks the sarcolemma including the transverse tubular system, was low. Super-resolution and immunoelectron microscopy supported the intracellular location of TRPC1. We investigated Ca2+ signaling in NRVMs after adenoviral TRPC1 overexpression or silencing. In NRVMs bathed in Na+ and Ca2+ free solution, TRPC1 overexpression and silencing was associated with a decreased and increased SR Ca2+ content, respectively. In isolated rabbit cardiomyocytes bathed in Na+ and Ca2+ free solution, we found an increased decay of the cytosolic Ca2+ concentration [Ca2+]i and increased SR Ca2+ content in the presence of the TRPC channel blocker SKF-96365. In a computational model of rabbit ventricular myocytes at physiological pacing rates, Ca2+ leak through SR TRPC channels increased the systolic and diastolic [Ca2+]i with only minor effects on the action potential and SR Ca2+ content. Our studies suggest that TRPC1 channels are localized in the SR, and not present in the sarcolemma of ventricular myocytes. The studies provide evidence for a role of TRPC1 as a contributor to SR Ca2+ leak in cardiomyocytes, which was previously explained by ryanodine receptors only. We propose that the findings guide us to an understanding of TRPC1 channels as modulators of [Ca2+]i and contractility in cardiomyocytes.
Content may be subject to copyright.
Contents lists available at ScienceDirect
Journal of Molecular and Cellular Cardiology
journal homepage: www.elsevier.com/locate/yjmcc
Location and function of transient receptor potential canonical channel 1 in
ventricular myocytes
Qinghua Hu
a,c,1
, Azmi A. Ahmad
a,b,1
, Thomas Seidel
a
, Chris Hunter
a
, Molly Streiff
a,b
,
Linda Nikolova
d
, Kenneth W. Spitzer
a
, Frank B. Sachse
a,b,
a
Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
b
Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
c
Department of Cardiovascular Surgery, Xiangya Hospital, Central-South University, Changsha, Hunan 410078, China
d
Core Research Facilities, Health Sciences Center, University of Utah, Salt Lake City, UT 84112, USA
ARTICLE INFO
Keywords:
TRPC1 channels
Ventricular myocyte
Sarcoplasmic reticulum
Calcium signaling
ABSTRACT
Transient receptor potential canonical 1 (TRPC1) protein is abundantly expressed in cardiomyocytes. While
TRPC1 is supposed to be critically involved in cardiac hypertrophy, its physiological role in cardiomyocytes is
poorly understood. We investigated the subcellular location of TRPC1 and its contribution to Ca
2+
signaling in
mammalian ventricular myocytes. Immunolabeling, three-dimensional scanning confocal microscopy and
quantitative colocalization analysis revealed an abundant intracellular location of TRPC1 in neonatal rat ven-
tricular myocytes (NRVMs) and adult rabbit ventricular myocytes. TRPC1 was colocalized with intracellular
proteins including sarco/endoplasmic reticulum Ca
2+
ATPase 2 in the sarcoplasmic reticulum (SR).
Colocalization with wheat germ agglutinin, which labels the glycocalyx and thus marks the sarcolemma in-
cluding the transverse tubular system, was low. Super-resolution and immunoelectron microscopy supported the
intracellular location of TRPC1. We investigated Ca
2+
signaling in NRVMs after adenoviral TRPC1 over-
expression or silencing. In NRVMs bathed in Na
+
and Ca
2+
free solution, TRPC1 overexpression and silencing
was associated with a decreased and increased SR Ca
2+
content, respectively. In isolated rabbit cardiomyocytes
bathed in Na
+
and Ca
2+
free solution, we found an increased decay of the cytosolic Ca
2+
concentration [Ca
2+
]
i
and increased SR Ca
2+
content in the presence of the TRPC channel blocker SKF-96365. In a computational
model of rabbit ventricular myocytes at physiological pacing rates, Ca
2+
leak through SR TRPC channels in-
creased the systolic and diastolic [Ca
2+
]
i
with only minor effects on the action potential and SR Ca
2+
content.
Our studies suggest that TRPC1 channels are localized in the SR, and not present in the sarcolemma of ven-
tricular myocytes. The studies provide evidence for a role of TRPC1 as a contributor to SR Ca
2+
leak in car-
diomyocytes, which was previously explained by ryanodine receptors only. We propose that the findings will
guide us to an understanding of TRPC1 channels as modulators of [Ca
2+
]
i
and contractility in cardiomyocytes.
1. Introduction
Transient receptor potential (TRP) channels were originally dis-
covered in Drosophila [1]. Subsequently, a variety of TRP-related
channels were identified in mammals. Based on homology of protein
sequences TRP-related channels have been classified into seven families
[2], including the family of transient receptor potential canonical
(TRPC) channels that are expressed in the mammalian heart. While
recent evidence suggests that TRPC channels are critical effectors in
cardiac hypertrophy and heart failure [3–7], their physiological role in
cardiac cells is still not understood [8]. Suggested roles for TRPC
channels in these cells include store-operated and receptor-operated
Ca
2+
entry [9]. Store-operated Ca
2+
entry is a mechanism by which the
Ca
2+
-depleted sarcoplasmic reticulum (SR) is refilled by Ca
2+
flux
through sarcolemmal ion channels into the cytosol and subsequent
uptake in the SR. Receptor-operated Ca
2+
entry is a mechanism by
https://doi.org/10.1016/j.yjmcc.2020.01.008
Received 17 August 2019; Received in revised form 16 December 2019; Accepted 21 January 2020
Abbreviations: eGFP, enhanced green fluorescent protein; NRVMs, neonatal rat ventricular myocytes; RyR, ryanodine receptor; SR, sarcoplasmic reticulum; sh, short
hairpin; TRP, transient receptor potential; TRPC1, transient receptor potential canonical 1
Corresponding author at: University of Utah, Nora Eccles Harrison Cardiovascular Research and Training Institute, 95 South 2000 East, Salt Lake City, UT 84112-
5000, USA.
E-mail address: frank.sachse@utah.edu (F.B. Sachse).
1
Contributed equally to this paper
Journal of Molecular and Cellular Cardiology 139 (2020) 113–123
Available online 23 January 2020
0022-2828/ © 2020 The Author(s). Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
T
which Ca
2+
flux through sarcolemmal ion channels is controlled by a
receptor, commonly a G-protein-coupled receptor. In addition to a role
in store-operated and receptor-operated Ca
2+
entry TRPC channels in
cardiomyocytes have been linked to mechano-electrical feedback, i.e.
modulation of cellular electrophysiology by mechanical stimuli. This
suggested role is based on studies indicating that some TRPC channels,
including TRPC1 channels, are stretch-activated [10,11].
A major obstacle to identification of a functional role of TRPC
channels in cardiomyocytes is the diversity of information concerning
subcellular location of the channels [8]. The location of TRPC channels
in ventricular cardiomyocytes was primarily investigated in rodents.
Several sarcolemmal locations were proposed: outer sarcolemma
[12–14], transverse tubular system (t-system) [12–18], and sarcolemma
at the intercalated disks [15]. Two types of sub-sarcolemmal location
were suggested: subsarcolemmal vesicles and peripheral region of the
myocytes [19,20]. While a location of TRPC channels in intracellular
membranes has been indicated in some studies [21,22], the specific
organelle was in general not specified. Interestingly, a study on skeletal
muscle in rodent reported that TRPC1 resides in the SR [23]. This study
suggested a functional role of TRPC1 channels in SR Ca
2+
leak in
skeletal myocytes.
Here, we shed light on the functional role of TRPC channels in
ventricular cardiomyocytes. We focus on TRPC1, which is abundantly
expressed in these cells. We tested the hypothesis that TRPC1 channels
localized in the SR membrane of ventricular cardiomyocytes contribute
to SR Ca
2+
leak. In these cells, SR Ca
2+
leak is commonly explained by
ryanodine receptors (RyRs) [24].
First, we investigated the spatial distribution of TRPC1 in neonatal
rat ventricular myocytes (NRVMs). NRVMs allowed us to overexpress
and silence TRPC1 expression using adenoviral TRPC1-eGFP and short
hairpin (sh)RNA TRPC1-eGFP constructs, respectively. Importantly,
NRVMs do not exhibit a t-system, which simplifies establishing a spatial
relationship of TRPC1 to the sarcolemma. Next, we studied adult rabbit
ventricular myocytes to relate findings from cultured neonatal cells to
adult native cells with a t-system similar as in human ventricular
myocytes. We performed three-dimensional (3D) confocal microscopy
to visualize and reconstruct the distribution of TRPC1, sarcolemma, SR,
and cytoskeletal proteins. We measured colocalization to investigate
the spatial relationship of TRPC1 channels with the sarcolemma and
various proteins. In addition, we applied super-resolution and im-
munoelectron microscopy to study the distribution of TRPC1 at nan-
ometer scale. Subsequently, we used a cytosolic Ca
2+
indicator and
fluorescence microscopy in NRVMs to investigate if TRPC1 over-
expression and silencing modulate SR Ca
2+
leak. These studies were
accompanied by studies on NRVMs and adult rabbit ventricular myo-
cytes using a TRPC channel blocker. Finally, we applied a computa-
tional model of rabbit ventricular myocytes to explore a potential
functional role of TRPC1 channels in electrophysiology and in-
tracellular Ca
2+
signaling.
2. Materials and methods
All studies were conducted in accordance with National Institutes of
Health Guidelines for the Care and Use of Animals and reviewed by the
Institutional Animal Care and Use Committee at the University of Utah,
where the work was performed. An additional Methods section is
available in Supplementary Material.
2.1. Preparation, adenoviral infection and culture of NRVMs
Cells were enzymatically isolated from 1-day old rats (NCIS,
Worthington Biochemical Corporation, Lakewood, NJ, USA). NRVMs
were subsequently separated from fibroblasts and plated at 175-200 k
density in 24 well tissue culture plates containing 5 mm coverslips
treated with fibronectin. In some studies, NRVMs were then infected
with an adenoviral vector containing a human TRPC1 attached to
enhanced green fluorescent protein (TRPC1-eGFP) at 200 multi-
plication of infection (MOI) or eGFP (Cat No. 1060) as control at 25
MOI. In some studies, we also applied an adenoviral vector with human
TRPC1 attached to enhanced green fluorescent protein and 6 HIS fused
to the N-terminal. Furthermore, we infected NRVMs with shRNA TRPC1
with eGFP marker (shRNA-TRPC1-eGFP, Cat No. shADV-226,536) to
silence TRPC1 expression, or a scrambled RNA with eGFP marker
(scRNA-eGFP, Cat No. 1122) as shRNA-TRPC1 control. Both infections
were performed at 80 MOI. All viral vectors had a backbone of type 5
(dE1/E3), and were produced by Vector Biolabs (Malvern, PA, USA). At
24 h after infection, cells were washed with DMEM to remove the virus.
Infected cells were then maintained and cultured for a total of 4–5 days
by regularly exchanging the culture media, and incubating them in a
humidity and CO
2
controlled incubator at 37 °C.
2.2. Immunolabeling of myocytes
NRVMs plated on coverslips were fixed for 15 min with 1% paraf-
ormaldehyde. Cells were permeabilized using 0.3% Triton X-100 (VWR
International, Radnor, PA, USA) for 18 min and then bathed in image-iT
FX Signal Enhancer (Thermo Fisher Scientific, Waltham, MA, USA) for
30 min and in 10% normal donkey serum (Millipore, Billerica, MA,
USA) for 60 min. Cells were then incubated with primary antibodies for
TRPC1 (T8666-09A, Supplementary Methods) and sarco/endoplasmic
reticulum Ca
2+
ATPase 2 (SERCA2) (MA3–910, Thermo Fisher
Scientific) overnight at 4 °C. Subsequently, cells were incubated with
secondary antibodies, a donkey anti-goat conjugated to Alexa Fluor 633
(Invitrogen, Carlsbad, California, USA) and a donkey anti-mouse con-
jugated to Alexa Fluor 555 for 60 min at room temperature. Finally,
cells were incubated with DAPI (D3571, Thermo Fisher Scientific) for
15 min to stain nuclei. Cells were triple-rinsed with phosphate-buffered
saline (PBS) for 15 min between each step.
Rabbit cardiomyocytes were isolated using enzymatic digestion
(Supplementary Methods). Cell pellets were incubated with wheat germ
agglutinin (WGA) conjugated to Alexa Fluor-633 for 20 min, followed
by 15 min of fixation with 2% paraformaldehyde. Before applying
primary antibodies, the cells were permeabilized using 0.1% Triton X-
100 and blocked using 10% normal donkey serum. Commonly, two
different primary antibodies were applied simultaneously, one raised in
goat for labeling TRPC1 channels and the other raised in mouse for
labeling other proteins. The cells were incubated with the primary
antibodies overnight at 4 °C. Two secondary antibodies, a donkey anti-
goat antibody conjugated to Alexa Fluor 488 and a donkey anti-mouse
antibody conjugated to Alexa Fluor 555, were then applied for 2 h at
room temperature.
We assessed specificity of the TRPC1 antibody using confocal mi-
croscopy of skeletal muscle tissues from WT and TRPC1 knockout mice
(Supplementary Methods and Fig. S1). Furthermore, we assessed our
TRPC1 labeling protocol in rabbit cardiomyocytes by omission of the
primary antibody and pre-incubation of the primary antibody with a
blocking peptide, which both eliminated the TRPC1 signal (Fig. S2).
2.3. Confocal imaging and image processing
For acquiring 3D stacks of NRVMs, we used a Leica SP8 TCS mi-
croscope (Leica Microsystems, Wetzlar, Germany) equipped with
GaAsP-HyD detectors and a 40× oil immersion lens (numerical aper-
ture: 1.2). Alexa Fluor 488 was excited with a laser wavelength of
488 nm and emitted light was acquired after band-pass filtering from
491 to 555 nm. Alexa Fluor 555 and 633 were excited at 561 and
633 nm wavelengths, respectively. Fluorescence for excitation at 561
and 633 nm was collected at 566–604 and 638–775 nm, respectively.
DAPI signal was excited with a 405 nm laser and collected at
410–570 nm.
For 3D imaging of segments of rabbit myocytes, we used a Zeiss LSM
5 Duo microscope (Carl Zeiss, Jena, Germany) equipped with a 63× oil
Q. Hu, et al. Journal of Molecular and Cellular Cardiology 139 (2020) 113–123
114
immersion lens (numerical aperture: 1.4). Alexa Fluor 488 was excited
with a 488 nm laser and emitted light was acquired after long pass
filtering at 505 nm. Alexa Fluor 555 and Alexa Fluor 633 were excited
at a wavelength of 543 nm and 633 nm, respectively. A 560 nm and
650 nm long pass filter was applied for light emitted from Alexa Fluor
555 and Alexa Fluor 633, respectively.
We acquired image stacks with a voxel size of 0.1 × 0.1 × 0.1 μm.
The image stacks were preprocessed including noise reduction, decon-
volution, background correction and attenuation correction as de-
scribed previously [25]. We applied Pearson's correlation coefficient R
r
to assess colocalization in the images (Supplementary Methods).
2.4. Measurement of [Ca
2+
]
i
in myocytes
Experiments were conducted on NRVMs at 4–5 days post-infection.
[Ca
2+
]
i
was measured with a Leica SP8 TCS confocal microscope
equipped with a 40× oil immersion lens (numerical aperture: 1.2) and
GaAsP-HyD detectors. NRVMs were loaded with 10 μM Rhod-3 AM
(R10145, Thermo-Fisher Scientific) in modified Tyrode solution at
37 °C for 45 min. After transfer into an imaging chamber, cells were
superfused with the modified Tyrode solution at room temperature
(22 ± 1 °C). A 488 nm laser was applied to excite eGFP, and emitted
light was collected after a 491–555 nm bandpass filter. A 561 nm laser
was then used to excite Rhod-3 Ca
2+
dye, and emitted light was col-
lected using a 566 nm long pass filter. Na
+
and Ca
2+
free Tyrode-like
solution was used to block the sodium‑calcium exchanger and other
sarcolemmal Ca
2+
currents.
Image sequences were acquired at a rate of 28 ms/image. Cells were
initially paced by field stimulation at 0.5 Hz in modified Tyrode solu-
tion until reaching steady state Ca
2+
signals. We then turned off the
stimulation and rapidly switched to Na
+
and Ca
2+
free solution for
2 min. Afterwards we rapidly applied 20 mM caffeine in the Na
+
and
Ca
2+
free solution to cause SR Ca
2+
release and to estimate SR Ca
2+
content as described previously [26]. The protocol was applied to
NRVMs infected with eGFP, TRPC1-eGFP, shRNA-TRPC1-eGFP, and
scRNA-eGFP. [Ca
2+
]
i
transients were extracted from Rhod3 signal by
cropping cytosolic regions with homogeneous intensities as described in
Fig. S3. For analyses of SR Ca
2+
content measurements, traces with
multiple transients during the Na
+
and Ca
2+
free period were ex-
cluded.
Similar studies on eGFP infected NRVMs were performed to assess
RyR Ca
2+
leak. SR Ca
2+
content and leak were assessed with or without
application of 1 mM tetracaine (Sigma-Aldrich) during the application
of Na
+
and Ca
2+
free solution. Furthermore, we carried out studies
using the TRPC1-eGFP and eGFP constructs with or without application
of 5 μM SKF-96365 (Sigma-Aldrich), a general TRPC channel blocker.
SKF-96365 was dissolved in DMSO before adding it to Na
+
and Ca
2+
free solution. Finally, we measured amplitudes of [Ca
2+
]
i
transients
from the various experiments using eGFP, TRPC1-eGFP, and shRNA-
TRPC1-eGFP infected NRVMs.
Our protocol for measurement of [Ca
2+
]
i
in rabbit ventricular
myocytes is described in supplemental material. We applied SKF-96365
at a concentration of 5 μM as a non-specific blocker of TRPC channels
[13].
2.5. Modeling of SR Ca
2+
leak and effects on [Ca
2+
]
i
Mathematical models of NRVMs and adult rabbit ventricular myo-
cytes were used to shed light on effects of SR Ca
2+
leak through TRPC1
channels on cellular electrophysiology and Ca
2+
signaling for physio-
logical pacing rates. We refer to the supplement for an introduction to
our NRVM model. Our model of adult rabbit ventricular myocytes was
based on a previously developed model [27], which comprises a de-
scription of passive SR Ca
2+
leak into the junctional space through RyR
channels. We added a description of Ca
2+
fluxes through TRPC1
channels J
SR,TRPC
from the SR into the cytosol:
=+ +
J K ([Ca ] [Ca ] )
SR,TRPC SR,TRPC 2SR 2i
with the rate of leak K
SR,TRPC
and the SR Ca
2+
concentration [Ca
2+
]
SR
.
The rate K
SR,TRPC
was determined by fitting of self-ratioed cytosolic
Ca
2+
signals measured in the presence and absence of SKF-96365 to
self-ratioed [Ca
2+
]
i
calculated with the myocyte model. Simulations
with the model were carried out using JSim (version 2.13) [28]. Si-
mulation results after 1 min of pacing at 2, 3 and 4 Hz were analyzed
for reduced, normal and increased Ca
2+
fluxes through TRPC1 chan-
nels.
2.6. Statistical analysis
Data are presented as mean ± standard error. Statistical analyses
were performed in Matlab version R2012b and higher (Mathworks Inc.,
Natick, MA, USA). Comparison of experimental data was performed
using a one-way analysis of variables (ANOVA). Comparison of data
from epifluorescence microscopy was based on the paired student t-test.
Differences were considered significant for P-values less than 0.01 or
0.05 where noted.
3. Results
3.1. Spatial distribution of TRPC1 in NRVMs
Applying confocal microscopy, we investigated the spatial dis-
tribution of wild-type (WT) TRPC1 in NRVMs that were cultured for
4–6 days, then fixed and labeled. Example sections from unprocessed
and deconvolved 3D image stacks are presented in Fig. S4 and 1, re-
spectively. Remarkably, TRPC1 exhibited a striated intracellular dis-
tribution (Fig. 1A). SERCA2 labeling presented a similar striated pat-
tern, accompanied by an irregular network pattern spanning
throughout the cell (Fig. 1B). The spatial distribution is particularly
apparent in enlarged images (Fig. 1D and E). SERCA2 and TRPC1 ap-
pear to some degree colocalized in the overlay image (Fig. 1G).
After infection with TRPC1-eGFP constructs and culture of 4–5 days,
NRVMs showed similar TRPC1 and SERCA2 patterns (Fig. 2A-D) as WT
NRVMs (Fig. 1). Unprocessed stacks are shown in Fig. S5. Fluorescence
of eGFP confirmed adenoviral TRPC1 expression (Fig. 2E and F).
Overlay images present a similar colocalization of TRPC1 and SERCA2
antibody associated fluorescence (Fig. 2G) as in WT NRVMs, but only
weak colocalization of TRPC1 and TRPC1-eGFP fluorescence (Fig. 2H)
as well as partial colocalization of TRPC1-eGFP with SERCA2 fluores-
cence (Fig. 2I).
WT NRVMs exhibited high colocalization of SERCA2 and TRPC1
(R
r
= 0.4 ± 0.03, n
cells
= 5). Similarly, R
r
values for TRPC1 coloca-
lization with SERCA2 across eGFP, TRPC1-eGFP, and shRNA-TRPC1-
eGFP infected NRVMs were 0.46 ± 0.03 (n
cells
= 5), 0.41 ± 0.03
(n
cells
= 5), and 0.47 ± 0.03 (n
cells
= 5), respectively (Fig. 2J). Dif-
ferences were not significant between the groups. Colocalization of
eGFP signal with SERCA2 was higher for TRPC1-eGFP versus eGFP and
shRNA-TRPC1-eGFP infected cells (0.41 ± 0.04 versus 0.26 ± 0.02
and 0.22 ± 0.04, respectively P< .05, n
cells
= 5) (Fig. 2K).
Example images from confocal microscopy of TRPC1 labeled
NRVMs infected with eGFP (7 images, ~30 cells), TRPC1-eGFP (14
images, ~40 cells), shRNA-TRPC1-eGFP (10 images, ~25 cells) and
scrambled RNA constructs (3 images, ~30 cells) are shown in Fig. S6. In
shRNA-TRPC1-eGFP infected cells, TRPC1 signals were small and
striations rare (Fig. S6C) suggesting silencing of TRPC1 protein ex-
pression.
We acquired super-resolution images of TRPC1 and SERCA2 in WT
NRVMs using single molecule localization (Supplementary Methods).
The NRVMs exhibited a striated arrangement of TRPC1 clusters of
variable size (Fig. 3A). SERCA2 exhibited a similar pattern and was in
part in close proximity to TRPC1 (Fig. 3B and C).
Q. Hu, et al. Journal of Molecular and Cellular Cardiology 139 (2020) 113–123
115
3.2. Spatial relationship of TRPC1, SERCA2 and cytoskeletal proteins in
rabbit ventricular myocytes
We imaged rabbit ventricular myocytes labeled with WGA and
TRPC1 antibodies. WGA served as a marker of the outer sarcolemma
and t-system. Exemplary images from co-labeling of WGA and TRPC1
are presented in Fig. 4. TRPC1 exhibited a striated transversal dis-
tribution along Z-lines as indicated by the t-system. The striated dis-
tribution is particularly evident in 3D visualization of cell segments
(Fig. 4E). The majority of signal for TRPC1 did not overlap with WGA
signal, suggesting that TRPC1 is localized in an intracellular membrane.
We confirmed presence of TRPC1 in rabbit ventricular myocardium
using RNA sequencing (Supplementary Material, Fig. S7 and [29]).
TRPC1 mRNA expression was higher than expression of other TRPCs.
Similar as for NRVMs, we investigated whether TRPC1 is located in
the SR of adult rabbit cardiomyocytes, using SERCA2 as a marker
(Fig. 5). SERCA2 labeling presented a lattice-like pattern including
transverse components adjacent to the Z-lines and longitudinal com-
ponents spanning throughout the whole cell (Fig. 5B), which is in
agreement with previous studies [30]. Visual inspection of the overlay
images indicates that a large portion of TRPC1 was colocalized with
transverse components of SERCA2 (Fig. 5C and D).
It has been suggested that the cytoskeleton is a major modulator of
localization and function of the TRPC channels [31,32]. Thus, we
characterized the spatial relationship between TRPC1 and cytoskeletal
proteins using confocal microscopy (Fig. 5E-H). Major components of
the cytoskeleton are actin filaments, microtubules, and intermediate
filaments. We therefore investigated α-actinin, which is an established
marker for the Z-lines of sarcomeres, and vinculin, which is abundantly
expressed in the costamere and intercalated disks and plays a role in
anchoring actin filaments to integrins. We also investigated β-tubulin,
one of the subunits forming the heterodimer tubulin of microtubules,
Fig. 1. Confocal microscopic images of fixed WT NRVMs labeled for TRPC1, SERCA2 and nuclei. The images were extracted from deconvolved 3D image stacks. (A)
The spatial distribution of TRPC1 exhibits a striated pattern. (B) SERCA2, a marker for SR, presents a striated pattern accompanied with mesh-network pattern. (C)
DAPI was used a marker for the nucleus. (D-F) Enlarged regions marked with box in (A-C), respectively. (G) Enlarged overlay of (D-F). Orange indicates overlap of
TRPC1 and SERCA2 associated fluorescence. The scale bar in (A) applies to (B) and (C). Scale bar in (D) applies to (E) and (F).
Fig. 2. Confocal microscopic images of fixed TRPC1-eGFP infected NRVMs and colocalization analyses. (A) Similar as in WT NRVMS, antibody labeling reveals a
striated pattern of TRPC1. (B) Enlarged region marked with box in (A). (C) SERCA2 antibody indicating SR. (D) Enlarged region marked with box in (C). (E) Image of
expressed TRPC1-eGFP construct. (F) Enlarged region marked with box in (E). (G) Overlay of (B) and (D). (H) Overlay of (B) and (F). (I) Overlay of (D) and (F). (J)
Pearson correlation coefficient (R
r
), calculated from image stacks of TRPC1 and SERCA antibody in different infection groups. (K) R
r
calculated from image stacks of
eGFP signal and SERCA antibody associated fluorescence in three infection groups. The scale bar in (A) applies to (C) and (E). Scale bar in (B) applies to (D), (F) and
(G-I). Brackets mark significant differences (P< .05).
Q. Hu, et al. Journal of Molecular and Cellular Cardiology 139 (2020) 113–123
116
and desmin, a type III intermediate filament located adjacent to Z-lines.
TRPC1 was highly colocalized with α-actinin (Fig. 5E) and desmin
(Fig. 5F). Beta-tubulin indicated a predominantly longitudinal or-
ientation of microtubules, and its overlap with TRPC1 signals was small
(Fig. 5G). TRPC1 was colocalized with vinculin (Fig. 5H) to a similar
degree as with SERCA2.
In addition to visual inspection, we assessed the colocalization of
TRPC1, WGA, SERCA2A and proteins of the cytoskeleton by calculating
Pearson's correlation coefficient R
r
from 3D image stacks. Our results,
using image stacks from cell segments (n
cells
= 48), showed high values
of R
r
for TRPC1 with sarcomeric α-actinin (0.647 ± 0.033), indicating
spatial adjacency (Fig. 5I). Values of R
r
were moderate for TRPC1 with
desmin, vinculin and SERCA2. Small values of R
r
for TRPC1 with β-
tubulin and WGA indicate remoteness of TRPC1 from microtubules and
sarcolemma, respectively. The colocalization of WGA with TRPC1 was
as small as with α-actinin, β-tubulin and desmin (Fig. 5J).
3.3. Immunoelectron microscopy of TRPC1 in rabbit ventricular myocytes
Using immunoelectron microscopy we imaged the distribution of
TRPC1 at nanometer resolution (Supplementary Material, Fig. S8).
TRPC1 was extensively marked adjacent to the Z-lines and along the
sarcomeres, but only few markers were found elsewhere. The abun-
dance of the TRPC1 labeling appeared higher in regions close (within
200 nm) to the Z-lines than elsewhere, which agreed with our findings
from confocal microscopy. However, instead of the regular striated
pattern observed in confocal microscopy, a clustered distribution of
TRPC1 was found in electron microscopy. In our images from
Fig. 3. Example super-resolution images of fixed WT NRVMs labeled with (A) TRPC1 and (B) SERCA2 antibody. (C) Overlay of images (A) and (B). (D-F) Enlarged
regions marked by box in (A-C). TRPC1 and SERCA2 present a striated clustered arrangement. The scale bar in (A) applies to (B) and (C). Scale bar in (D) applies to
(E) and (F).
Fig. 4. Confocal microscopic images of TRPC1 and sarcolemma in adult rabbit ventricular myocyte. (A) TRPC1 exhibits a transversely striated distribution with the
majority of TRPC1 within the cell. (B) WGA was used a marker for sarcolemma including t-system. (C) Overlay of (A) and (B). (D) Enlarged region marked with box in
(C). (E) Three-dimensional visualization of cell segment.
Q. Hu, et al. Journal of Molecular and Cellular Cardiology 139 (2020) 113–123
117
immunoelectron microscopy TRPC1 was not visible in the t-system (Fig.
S8A) and outer sarcolemma (Fig. S8B), supporting the hypothesis that
TRPC1 is located within the myocyte. Omission of primary antibodies
served as negative control (Fig. S9).
3.4. Measurement of SR Ca
2+
content in NRVMs
Our studies on TRPC1 location in NRVMs suggested that the chan-
nels are not in the sarcolemma, but in an organelle. Using confocal
imaging and a Ca
2+
sensitive dye we investigated SR Ca
2+
content in
NRVMs infected with eGFP, TRPC1-eGFP and shRNA-TRPC1-eGFP
constructs (Fig. 6A, B and C, respectively). The protocol involved pa-
cing of NRVMs, followed by bathing of quiescent cells in Na
+
and Ca
2+
free solution for 2 min, followed by rapid application of caffeine (Fig.
S3). Rapid application of caffeine led to a sudden increase of the Ca
2+
signal, which is explained by RyR channel opening and Ca
2+
release
from the SR into the cytosol. We evaluated the SR content by measuring
the amplitude of caffeine-induced peaks (Fig. 7). The SR Ca
2+
content
was measured in NRVMs infected with either eGFP (Fig. 7A), TRPC1-
eGFP (Fig. 7B) or shRNA-TRPC1-eGFP (Fig. 7C). SR Ca
2+
content was
Fig. 5. Spatial distribution of TRPC1 in rabbit ventricular myocytes. Confocal microscopic images of (A) TRPC1 and (B) SERCA2. (C) Overlay of (A) and (B). (D)
Enlarged region marked with box in (C). Yellow regions indicate colocalization of SERCA2 and TRPC1. The scale bar in (A) applies to (B) and (C). Confocal
microscopic images of TRPC1 with (E) α-actinin, (F) desmin, (G) β-tubulin and (H) vinculin. (I) Pearson correlation coefficients (R
r
) calculated from image stacks of
TRPC1, intracellular proteins and WGA. (J) R
r
determined from image stacks of WGA with TRPC1 and intracellular proteins. Brackets mark significant differences
(P< .01). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Fig. 6. Confocal microscopic images of living NRVMs infected with (A) eGFP at 25 MOI (B) TRPC1-eGFP at 200 MOI, and (C) shRNA-TRPC1-eGFP at 80 MOI.
Q. Hu, et al. Journal of Molecular and Cellular Cardiology 139 (2020) 113–123
118
smaller in NRVMs overexpressing TRPC1 (1.64 ± 0.21, n
cells
= 21)
than in NRVMs infected with eGFP (3.18 ± 0.23, n
cells
= 15,
P< .01). In contrast, SR Ca
2+
content was higher in NRVMs expres-
sing shRNA-TRPC1 (4.30 ± 0.49, n
cells
= 12, P< .05) than in
NRVMs expressing TRPC1-eGFP or eGFP. Studies were done on at least
8 litters per group.
Next, we studied effects of the TRPC channel inhibitor SKF-96365
on infected NRVMs (Fig. S10). Application of SKF-96365 increased SR
Ca
2+
content in both TRPC1-eGFP (2.56 ± 0.45, n
cells
= 11, P < .05)
and eGFP (2.92 ± 0.43, n
cells
= 13, P < .01) versus TRPC1-eGFP
infected cells in the absence of SKF-96365. As in Fig. 7D, NRVMs
overexpressing TRPC1-eGFP had reduced SR Ca
2+
content compared to
eGFP NRVMs (1.56 ± 0.22, n
cells
= 17 vs. 2.5 ± 0.24, n
cells
= 14,
P< .01). Studies were done on at least 5 litters per group.
To study possible effects of short-hairpin infection on NRVMs, we
conducted similar measurements of SR Ca
2+
content with a scrambled
RNA vector to serve as a separate control for the shRNA-TRPC1-eGFP
group (Fig. S11). The difference was not significant between eGFP and
scRNA-eGFP cells, suggesting that effects of shRNA-TRPC1-eGFP are
due to silencing of TRPC1 expression.
To assess RyR Ca
2+
leak, we applied 1 mM tetracaine. Example
traces with and without tetracaine application are shown in Fig. S12A
and B, respectively. We found differences in SR Ca
2+
content (Fig.
S12C) and self-ratioed [Ca
2+
]
i
at the end of 2-min rest period (Fig.
S12D). Application of tetracaine increased SR Ca
2+
content
(3.96 ± 0.29, n
cells
= 21 vs 2.82 ± 0.28, n
cells
= 17, p≤ .01). Self-
ratioed [Ca
2+
]
i
at the end of 2-min rest period decreased with tetra-
caine (0.92 ± 0.04 vs 1.09 ± 0.06, p≤ .05).
Amplitudes of [Ca
2+
]
i
transients in response to the 0.5 Hz pacing de-
creased in NRVMs infected with TRPC1-eGFP (1.72 ± 0.09, n
cells
= 49)
compared to eGFP (2.19 ± 0.17, n
cells
= 34, P≤ .05) and shRNA-TRPC1-
eGFP (2.42 ± 0.24, n
cells
= 14, P ≤ .01). Differences between eGFP and
shRNA-TRPC1-eGFP infected cells were not significant.
3.5. Measurement of [Ca
2+
]
i
and SR Ca
2+
content in rabbit ventricular
myocytes
Using epifluorescence microscopy and a Ca
2+
sensitive dye, we
evaluated the decay of [Ca
2+
]
i
and the SR Ca
2+
content in rabbit
ventricular myocytes bathed in Na
+
and Ca
2+
free solution in absence
and presence of SKF-96365. We observed a slow decay of the Ca
2+
signal in quiescent cells in the absence of SKF-96365 (Fig. 8A), which is
commonly explained by SR uptake and sarcolemmal leak of cytosolic
Ca
2+
. Application of SKF-96365 intensified the Ca
2+
signal decay
versus control (−14.8% vs. -9.4%, n
cells
= 9, P < .05) (Fig. 8B and C).
The SR Ca
2+
content in the presence of SKF-96365 was slightly ele-
vated (1.50 ± 0.26 versus 1.76 ± 0.26, +17.3%, n
cells
= 6,
P < .05) (Fig. 8D). Peak systolic Ca
2+
signals during action potentials
just before switching to Na
+
and Ca
2+
free solution were similar in the
control and SKF-96365 group (Fig. S13), indicating that the SR Ca
2+
content was also similar at that time.
3.6. Modeling of SR Ca
2+
leak and effects on [Ca
2+
]
i
in NRVMs
A mathematical model of NRVM electrophysiology [33] was mod-
ified to qualitatively reproduce our measurements of SR Ca
2+
content.
Applying the protocol from our studies on NRVMs (Fig. 7A-C) and
varying SR Ca
2+
leak, the model revealed a negative relationship be-
tween SR Ca
2+
leak and release. The model reproduced experimental
differences (Fig. 7D) of caffeine-induced ∆[Ca
2+
]
i
due to TRPC1 si-
lencing with a 98% decrease in leak and due to TRPC1 overexpression
with 331% leak (Fig. S14B-E). The model also qualitatively predicted
the decrease of [Ca
2+
]
i.
Amplitudes in TRPC1-eGFP infected versus
eGFP and shRNA-TRPC1-eGFP NRVMs that we found in our experi-
mental studies.
3.7. Modeling of SR Ca
2+
leak and physiological effects on [Ca
2+
]
i
in
rabbit ventricular myocytes
We used computational modeling to shed light on the role of TRPC1
channels in electrophysiology and Ca
2+
signaling of rabbit myocytes at
physiological pacing rates. Measured cytosolic Ca
2+
decay in quiescent
rabbit ventricular myocytes in the presence of SKF-96365 (Fig. 8C) was
reconstructed by setting K
SR,TRPC
to −1.1 × 10
−5
/ms (Fig. S15A and
B). The faster [Ca
2+
]
i
decay in the presence of SKF-96365 was ac-
companied by a slowed decay of [Ca
2+
]
SR
(Fig. S15C and D). After two
minutes decay, [Ca
2+
]
SR
was elevated with SKF-96365 vs. control (at
end of pacing: 0.52 mM, after 2 min: 0.50 mM vs. 0.48 mM, −4.0% vs%
vs. -8.8%). The model predicts increased [Ca
2+
]
SR
after SKF-96365
application (+5.2%), which is consistent with increased measured SR
Ca
2+
content (Fig. 8D).
We compared [Ca
2+
]
i
transients in control cells, cells in presence of
SKF-96365 and cells with increased expression of TRPC1 at pacing rates of
2, 3 and 4 Hz (Fig. 9A). We assumed that increased expression (or activa-
tion) of TRPC1 channels leads to an increase of SR Ca
2+
leak to 400%
versus control levels. Thus, effects of upregulated TRPC1 channels were
reconstructed by increasing SR Ca
2+
leak (K
SR,TRPC
= 4.4 × 10
−5
/ms).
For all cell models, minimal (diastolic) and maximal (systolic)
[Ca
2+
]
i
increased with increasing pacing rate (Fig. 9B and C). For all
pacing rates, SR Ca
2+
leak exhibited a positive relationship with ex-
trema and the amplitude of the [Ca
2+
]
i
transient. Modulation of the
diastolic [Ca
2+
]
i
was strongest (+23.1%) for high SR Ca
2+
leak and
low pacing rate. Modulation of the systolic [Ca
2+
]
i
and amplitude was
strongest (+7.4% and 3.3%, respectively) for high SR Ca
2+
leak and
high pacing rate.
Similarly, [Ca
2+
]
SR
increased with pacing rates for all cell models
(Fig. 9D). While SR Ca
2+
leak exhibited a positive relationship with
[Ca
2+
]
SR
minima (Fig. 9E), the relationship with [Ca
2+
]
SR
maxima was
negative (Fig. 9F).
The effect of SR Ca
2+
leak on action potentials was small (Fig. 9G and
H). The action potential duration exhibited a negative relationship with
pacing rate. Increased SR Ca
2+
leak was associated with marginally in-
creased action potential duration at 90% repolarization (APD
90
) (Fig. 9I).
Fig. 7. Measurement and analysis of Ca
2+
transients in NRVMs using confocal microscopy. Self-ratioed Ca
2+
signals (F/F
0
) from a cell infected with (A) eGFP, (B)
TRPC1-eGFP and (C) shRNA-TRPC1-eGFP constructs. SR Ca
2+
content was assessed using rapid application of caffeine (20 mM). (D) Statistical analysis of SR Ca
2+
release. Cells overexpressing TRPC1 and after silencing exhibited a decreased and increased amplitude of the self-ratioed Ca
2+
signal ΔF
Caff
/F
0
, respectively, versus
control. Brackets mark significant differences (P < .05).
Q. Hu, et al. Journal of Molecular and Cellular Cardiology 139 (2020) 113–123
119
Fig. 8. Epifluorescence microscopy of rabbit myocyte using a cytosolic Ca
2+
indicator. Self-ratioed Ca
2+
signals (F/F
0
) are presented for a cell undergoing pacing,
followed by bathing with Na
+
and Ca
2+
free solution (A) without and (B) with the TRPC channel blocker SKF-96365. In this cell, F/F
0
decreased in Na
+
and Ca
2+
free solution. Subsequently, caffeine (20 mM) was rapidly applied. (C) Decay of F/F
0
after 2 min in Na
+
and Ca
2+
free solution was increased in the presence of SKF-
96365 (SKF) versus control (CTR). (D) Application of caffeine in cells bathed in SKF-96365 caused an increased amplitude of the self-ratioed Ca
2+
signal ΔF
Caff
/F
0
versus control reflecting an increased SR Ca
2+
content. Brackets mark significant differences (P < .05).
Fig. 9. Effects of SR Ca
2+
leak current investigated in a computational model of rabbit ventricular myocytes. Simulations were performed at a pacing rate of 2, 3 and
4 Hz. Block (SKF-96365) and increased expression (400%) of TRPC1 channels were modeled by decreased and increased SR Ca
2+
leak, respectively. (A) SR Ca
2+
leak
affected [Ca
2+
]
i
at physiological pacing rates. In particular, SR Ca
2+
leak exhibited a positive relationship with (B) minimal and (C) maximal [Ca
2+
]
i
. (D) [Ca
2+
]
SR
transients with associated (E) minima and (F) maxima. While SR Ca
2+
leak exhibited a positive relationship with the minimal [Ca
2+
]
SR
, the relationship with
maximal [Ca
2+
]
SR
was negative. (G,H) SR Ca
2+
leak had only a marginal effect on V
m
. (I) In particular, variation of SR Ca
2+
leak led to small differences of APD
90
versus control.
Q. Hu, et al. Journal of Molecular and Cellular Cardiology 139 (2020) 113–123
120
4. Discussion
Our studies provide insights into the subcellular location and
functional role of TRPC1 in ventricular cardiomyocytes. Previous stu-
dies on cardiomyocytes suggested that TRPC1 are localized in the sar-
colemma and involved in membrane electrophysiology and mechano-
electrical coupling. Our studies using immunolabeling and confocal
microscopy revealed an intracellular distribution of TRPC1 in ven-
tricular myocytes isolated from neonatal rat and adult rabbit hearts.
TRPC1 was colocalized with SERCA2, an established marker of SR
(Figs. 1, 2 and 5). Quantitative colocalization analysis based on Pear-
son's correlation coefficient calculated from 3D microscopic image
stacks further supported intracellular expression of TRPC1 adjacent to
Z-lines in adult rabbit myocytes. TRPC1 exhibited high colocalization
with sarcomeric α-actinin, which is a marker of the Z-line (Figs. 5E and
I). The degree of colocalization with WGA, an established marker of
sarcolemma, was significantly lower (Figs. 4 and 5I) and comparable to
colocalization of WGA with α-actinin, desmin and tubulin (Fig. 5J),
which are cytoskeletal proteins not located in the sarcolemma. Due to
limited spatial resolution of confocal microscopy, our approach cannot
completely exclude sarcolemmal expression of TRPC1. To address this
issue, we employed immunoelectron and super-resolution microscopy
with a spatial resolution in the nanometer range. For NRVMs, super-
resolution microscopy reproduced the striated intracellular pattern of
TRPC1 in proximity to SERCA2 (Fig. 3) as observed with confocal mi-
croscopy (Fig. 1). Electron microscopy of rabbit ventricular myocytes
supported the absence of TRPC1 expression in the outer sarcolemma
and t-system (Fig. S8). The images indicate a discrete intracellular
distribution with partial clustering of TRPC1 adjacent to Z-lines. While
our approach for immunoelectron microscopy did not identify the in-
tracellular compartment where the TRPC1 reside, the majority of
TRPC1 was found in or close to z-lines as well as within sarcomeres.
The SR membrane has a high density in the region of Z-lines in adult
rat and sheep myocytes [34]. Accordingly, in our studies using confocal
microscopy on adult rabbit ventricular myocytes we found an increased
SERCA2 fluorescence associated with the Z-line. The colocalization of
TRPC1 with SERCA2 and the striated pattern of TRPC1 support our
hypothesis that TRPC1 is located in the SR membrane. We note that the
cultured NRVMs used in our study do not exhibit a t-system, yet the
striated organization of native TRPC1 (Fig. 1A) was similar to the
TRPC1 organization in adult rabbit myocytes. Furthermore, regions of
rabbit myocytes devoid of t-tubules still showed a prominent TRPC1
signal (Fig. 4). This suggests that TRPC1 localization is independent of
sarcolemmal organization.
To provide further evidence for a SR location of TRPC1 channels in
ventricular myocytes, we investigated the relationship between TRPC1
expression and SR Ca
2+
content in quiescent NRVMs held in Na
+
and
Ca
2+
free solution for 2 min. SR Ca
2+
leak was assessed by measuring
SR Ca
2+
content. We found that TRPC1 expression exhibited a negative
relationship with SR Ca
2+
content: expression of TRPC1-eGFP construct
decreased the SR Ca
2+
content in NRVMs, while silencing of TRPC1
with shRNA-TRPC1-eGFP increased SR Ca
2+
content (Fig. 7). These
results were qualitatively reproduced in a mathematical model with
increased and decreased SR Ca
2+
leak corresponding to TRPC1 upre-
gulation and silencing, respectively (Fig. S14). Furthermore, applica-
tion of 5 μM SKF-96365, an established blocker of TRPC channels, in-
creased the SR Ca
2+
content in TRPC1-eGFP infected NRVMs (Fig.
S10). Since the experiments were performed in Ca
2+
free bathing so-
lution and Ca
2+
cannot enter the cells from the extracellular space, the
source of increased [Ca
2+
]
i
after caffeine application must be in in-
tracellular pools. The action of SKF-96365 to increase SR Ca
2+
content
points at the SR as the intracellular Ca
2+
pool, which supports our
imaging results. Blocking RyR leak with tetracaine (Fig. S12) led to
similar increase in SR Ca
2+
content as measured after silencing of
TRPC1, using our shRNA-TRPC1-eGFP construct (Fig. S12C vs. 7D).
Increased SR Ca
2+
content was associated with a decrease in [Ca
2+
]
i
at
the end of the 2 min rest period (Fig. S12D). The similarity of effects of
TRPC1 silencing and RyR block indicates that both localize to the same
organelle. Collectively, these results suggest that TRPC1 channels
contribute to Ca
2+
leak from the SR into the cytosol.
Cultured NRVMS are in many aspects different than native ven-
tricular myocytes, so we furthered our studies using adult rabbit ven-
tricular myocytes. RNA sequencing suggested that TRPC1 is the most
expressed member of the TRPC family in these cells (Fig. S7).
Application of SKF-96365 increased the decay of [Ca
2+
]
i
and the SR
Ca
2+
content in cells bathed in Na
+
and Ca
2+
free solution (Fig. 8). As
for NRVMs (Fig. S10), we explain this finding by block of TRPC1
channels in the SR and a subsequent reduction in SR Ca
2+
leak.
Our studies provide evidence for the contribution of TRPC1 chan-
nels to Ca
2+
leak from the SR into the cytosol in cardiomyocytes. SR
Ca
2+
leak in these cells has been explained by spontaneous sparks from
RyRs, non-spark-mediated RyR leak and non-RyR leak [24,26]. The
contribution of spark-mediated to total leak is thought to be small at
low SR Ca
2+
concentration, but large for high SR Ca
2+
concentrations.
Non-RyR leak was suggested to amount to around 50% of the non-
spark-mediated RyR leak. Knowledge on the structural basis of non-RyR
leak is sparse. A 1,4,5-inositol-trisphosphate (InsP
3
) dependent SR Ca
2+
leak through InsP
3
receptors has been identified, but was found to be
very low in the absence of InsP
3
. Also, non-RyR leak was insensitive to
block of the InsP
3
receptor. Based on our studies, we suggest that the
enigmatic non-RyR leak is caused, at least in part, by fluxes through SR
TRPC1 channels.
Our computational simulations using a mathematical model of
rabbit ventricular myocytes with the addition of TRPC1 channels in the
SR suggest that TRPC1 channels modulate Ca
2+
transients at physio-
logical pacing rates (Fig. 9). We assumed that TRPC1 channels allow
Ca
2+
to leak from the SR into the cytosol, which was described in a
similar manner as Ca
2+
leak through RyR channels already in-
corporated in this model. In our model, the leak was proportional to the
difference between [Ca
2+
]
SR
and [Ca
2+
]
i
, and thus dominated by
[Ca
2+
]
SR
, which is approximately three orders of magnitude larger than
[Ca
2+
]
i
. In the simulations we focused on sub-acute effects of TRPC1
channel activation at a similar time scale as in our experimental as-
sessment of [Ca
2+
]
i
. Increased SR Ca
2+
leak augmented [Ca
2+
]
i
during
all phases of action potentials at physiological pacing rates. In parti-
cular, diastolic and systolic [Ca
2+
]
i
were increased by TRPC1 leak.
Interestingly, effects of TRPC1 channels on action potentials were small
with only marginal prolongation of APD
90
. The prolongation is ex-
plained by increased [Ca
2+
]
i
causing increased forward so-
dium‑calcium exchanger activity, which is electrogenic. The prolonga-
tion also causes increased Ca
2+
influx through L-type Ca
2+
channels,
which contributes to the increase of [Ca
2+
]
i
. Based on these simula-
tions, we propose a physiological role for TRPC1 channels in the SR of
cardiac myocytes in modulation of [Ca
2+
]
i
and thus contractility. In
particular, the simulations suggest that leak Ca
2+
through TRPC1
channels increases contractility in paced myocytes.
Our localization study on TRPC1 in NRVMs and rabbit ventricular
myocytes is in conflict with previous studies on rodent cardiomyocytes,
many of which used confocal microscopy [8]. It is, however, difficult to
describe spatial relationships of the t-system and proteins in rodent
using confocal microscopy. Compared to rabbit, the t-system in rodents
is denser and tubules are of smaller diameter, which hinders in-
vestigations using confocal microscopy due to limitations of spatial
resolution [35,36]. To illustrate the issue, we imaged ventricular
myocytes from adult rat using a similar experimental approach as for
ventricular myocytes of rabbits (Supplementary Methods, Fig. S16).
TRPC1 did not outline the outer sarcolemma. However, the dense t-
system obscured localization of TRPC1.
Similar discrepancies regarding TRPC1 location emerged in studies
of skeletal myocytes. In a study on mouse skeletal muscle, Gervasio
et al. reported sarcolemmal expression of TRPC1 [37], whereas Stiber
et al. described a sarcolemmal pattern of TRPC1 corresponding to
Q. Hu, et al. Journal of Molecular and Cellular Cardiology 139 (2020) 113–123
121
costameres at the level of Z discs [38]. In contrast, Berbey et al. pre-
sented a striated pattern of TRPC1 that matched SERCA1 im-
munolabeling in mouse skeletal myocytes [23]. The study applied im-
munolabeling for endogenous TRPC1 and overexpressing TRPC1
conjugated with yellow fluorescent protein, which revealed an in-
tracellular distribution of TRPC1 similar to our findings in cardio-
myocytes.
Previous studies on oocytes and CHO cells suggested that TRPC1
constitute stretch-activated ion channels [10], which is however still
controversially discussed [39,40]. Based on those studies, a role in
membrane electrophysiology and mechano-electrical coupling of myo-
cytes was proposed. While our studies did not provide evidence for a
direct role of TRPC1 in membrane electrophysiology, the studies shed
new light on mechanisms by which TRPC1 may contribute to mechano-
electrical coupling. The mechano-sensitivity of TRPC1 channels may
contribute to stretch-modulated SR Ca
2+
leak.
Another topic that we attempted to address in this study is the
spatial relationship between TRPC1 and cytoskeleton in cardiomyo-
cytes. The cytoskeleton plays a critical role in anchoring, trafficking and
functionally regulating TRPC channels [31]. Also, Ca
2+
influx through
TRPC channels was suggested to be involved in rearrangement and
remodeling of the cytoskeleton in many cell types [32]. Our confocal
microscopic images revealed that TRPC1 is highly colocalized with
sarcomeric α-actinin, and to some degree with desmin and vinculin
along the Z-lines, whereas the distribution of TRPC1 appeared to be
perpendicular to microtubules. Although the degree of functional co-
localization with cytoskeletal proteins is unknown, we suggest that the
intimate relationship with the sarcomeres allows TRPC1 channels to
sense mechanical strain in the cell interior.
Our studies provide insights into the functional role of TRPC1, thus
it is tempting to extrapolate our findings towards understanding the
pathophysiological role of TRPC1 channels. It was suggested that
TRPC1 channels are critical players in cardiac hypertrophy and heart
failure [41]. Several studies indicated pathological up-regulation of
TRPC1 [4,42]. For example, hypertrophic agents such as endothelin-1
were found to increase TRPC1 expression to 410% in NRVMs. A simple
prediction from our findings and reported pathological up-regulation of
TRPCs is that increased Ca
2+
leak from the SR into the cytosol causes
sustainedly increased [Ca
2+
]
i
. Sustained increase of [Ca
2+
]
i
has been
implicated previously in hypertrophic signaling in cardiac myocytes
[43]. Recent studies indicate that TRPC channels are potential targets
for treating heart failure [7] and cardiac dystrophy [44]. Based on our
findings the mechanism of TRPC blockade as a way to treat cardio-
vascular disease would be explained by reduction of SR Ca
2+
leak
leading to normalization of [Ca
2+
]
i
, and inhibition of hypertrophic
signaling.
Our interpretation of the presented studies assumes that TRPC1
forms channels that conduct Ca
2+
. Extensive prior work revealed that
TRPC1 forms heteromeric channels with other members of the TRPC,
TRPP and TRPV families [39]. This suggests that modulation of TRPC1
expression as performed in our studies will, beyond modulation of the
density of homomeric TRPC1 channels, modulate the density of het-
eromeric channels and contribution of TRPC1 to those channels. Re-
cently, it was hypothesized that TRPC1 acts as regulatory subunit in
heteromeric channel complexes [39]. Further studies will be required to
understand effects of modulation of TRPC1 expression and interpret our
findings in the context of this hypothesis.
Limitations of our approach include the spatial resolution of con-
focal microscopy [36]. Limited resolution exacerbates assessment of
colocalization of proteins. Another limitation is related to our approach
for immunoelectron microscopy, which did not provide direct in-
formation on SR membrane.
We note the high variability of results using NRVMs, which is, in
part, explained by the variable degree of expression of adenoviral
constructs. Even beyond variable expression of adenoviral constructs,
we noticed large variability in our studies on NRVMs. For instance,
eGFP expressing NRVMs exhibited a large variability of SR Ca
2+
con-
tent. This explains why application of SKF-96365 in these cells did not
yield statistically significant differences (Fig. S10). The high variability
complicates testing of statistical hypotheses. For instance, a-priori
power analysis based on preliminary studies on cells infected with eGFP
with and without SKF-96365 application suggested that a large sample
size (n~1000) is required for statements on statistically significant
differences between the two groups.
High variability in NRVM phenotypes also limits accuracy of the
mathematical model. The original model was constructed from ex-
periments in various laboratories where culture conditions were pre-
sumably heterogeneous and different to the conditions in our labora-
tory. Notably, temperature differences between the model and our
experiments restrict direct comparison. The original model was devel-
oped from measurements with NRVMs at 32 °C, while our experiments
were performed at room temperature (~22 °C). We applied room
temperature in order to control pacing rate of the cells, but temperature
affects many cellular functions. Thus, the simulations served primarily
for a qualitative comparison with our experimental findings. A general
limitation of our models is related to modeling of caffeine effects. We
limited our modeling of NRVMs to the initial phase of Ca
2+
release (Fig.
S14D) relevant for comparison with our measurements. Further work,
e.g. accounting for wash-in and out of caffeine, will be necessary to
accurately model the decay phase.
We also note differences of the spatial distribution on native TRPC1
and TRPC1-eGFP (Fig. 2H). We speculate that different time scales for
expression and localization of the native TRPC1 and TRPC1-eGFP ex-
plain the differences. A significant amount of the TRPC1-eGFP construct
may be still trafficking to the sites, where the native TRPC1 resides.
Nevertheless, similar R
r
values for SERCA2 with native TRPC1 (Fig. 2J)
and TRPC1-eGFP (Fig. 2K) construct suggest trafficking of the construct
to SR regions. The TRPC1-eGFP vector comprises fused eGFP molecule
yielding fluorescence upon expression of the TRPC1 protein (Fig. 6B).
In contrast, the eGFP and shRNA-TRPC1-eGFP vectors comprise a non-
fused eGFP yielding fluorescence upon successful infection of the cell.
Images from living NRVMs infected with eGFP and shRNA-TRPC1-eGFP
constructs indicate cytosolic and nuclear localization of the constructs
(Fig. 6A and C), likely due to intracellular diffusion of the small non-
fused eGFP molecule.
We acknowledge limitations of our functional studies related to the
absence of specific blockers or activators for TRPC1 channels. These
limitations triggered our development and application of adenoviral
TRPC1 overexpression and silencing in the presented studies on
NRVMs. Nevertheless, in some studies we applied SKF-96365, which is
an unspecific TRPC channel blocker, which also affects TRPV2, TRPM8
and voltage-gated calcium (Ca
v
) 1–3 channels [45]. We accounted, in
part, for the weak specificity of SKF-96365 with our experimental
protocol. SKF-96365 was applied to myocytes after pacing and during
rest for 2 min. The membrane voltage of myocytes at rest is close to the
Nernst voltage for potassium, and thus voltage gated channels, such as
Ca
V
1–3, are closed and not involved in our measurements. Further in-
sights into potential effects of unspecific action of SKF-96365 arise from
our RNASeq data from rabbit ventricular myocardium. These data
suggest that expression of TRPC1 is much higher than expression of
TRPC 3, 4, 6 and 7 (Fig. S7) as well as TRPV2 and TRPM8 [29]. While
we cannot fully exclude that SKF-96365 effects on these channels affect
our measurements, we note that the results from application of SKF-
96365 (Figs. 8D and S12) are consistent with results applying TRPC1
silencing (Fig. 7).
Supplementary data to this article can be found online at https://
doi.org/10.1016/j.yjmcc.2020.01.008.
Author contributions
Designed research: QH, AAA, KWS, FBS.
Performed research: QH, AAA, MS, KWS, CH, LN.
Q. Hu, et al. Journal of Molecular and Cellular Cardiology 139 (2020) 113–123
122
Analyzed data: QH, AAA, TS, MS, FBS.
Wrote the manuscript: QH, AAA, MS, FBS.
Sources of funding
We acknowledge funding by the Nora Eccles Treadwell Foundation
and the National Heart, Lung and Blood Institute (R01HL094464 and
R01HL132067).
Disclosures
None declared.
Acknowledgements
We acknowledge molecular biology support and expert advice on
adenoviral expression of the TRPC1 constructs from Dr. Michael
Sanguinetti. We thank Dr. Boris Martinac for providing us with the
TRPC1 construct. We also thank Mr. Brett Milash for his help with the
RNASeq data analysis. We appreciate the support of Dr. Kate Larson,
who provided us with tissue samples from WT and TRPC1 KO mice.
References
[1] C. Montell, G.M. Rubin, Molecular characterization of the Drosophila trp locus: a
putative integral membrane protein required for phototransduction, Neuron 2 (4)
(1989) 1313–1323.
[2] B. Nilius, G. Owsianik, T. Voets, J.A. Peters, Transient receptor potential cation
channels in disease, Physiol. Rev. 87 (1) (2007) 165–217.
[3] H. Watanabe, M. Murakami, T. Ohba, K. Ono, H. Ito, The pathological role of
transient receptor potential channels in heart disease, Circ. J. 73 (3) (2009)
419–427.
[4] M. Seth, Z.S. Zhang, L. Mao, V. Graham, J. Burch, J. Stiber, L. Tsiokas, M. Winn,
J. Abramowitz, H.A. Rockman, L. Birnbaumer, P. Rosenberg, TRPC1 channels are
critical for hypertrophic signaling in the heart, Circ. Res. 105 (10) (2009)
1023–1030.
[5] P. Eder, J.D. Molkentin, TRPC channels as effectors of cardiac hypertrophy, Circ.
Res. 108 (2) (2011) 265–272.
[6] R. Vennekens, Emerging concepts for the role of TRP channels in the cardiovascular
system, J. Physiol. 589 (Pt 7) (2011) 1527–1534.
[7] K. Seo, P.P. Rainer, V. Shalkey Hahn, D.I. Lee, S.H. Jo, A. Andersen, T. Liu, X. Xu,
R.N. Willette, J.J. Lepore, J.P. Marino Jr., L. Birnbaumer, C.G. Schnackenberg,
D.A. Kass, Combined TRPC3 and TRPC6 blockade by selective small-molecule or
genetic deletion inhibits pathological cardiac hypertrophy, Proc. Natl. Acad. Sci. U.
S. A. 111 (4) (2014) 1551–1556.
[8] A.A. Ahmad, M. Streiff, C. Hunter, Q. Hu, F.B. Sachse, Physiological and patho-
physiological role of transient receptor potential canonical channels in cardiac
myocytes, Prog. Biophys. Mol. Biol. 130 (Pt B) (2017) 254–263.
[9] A.B. Parekh, J.W. Putney Jr., Store-operated calcium channels, Physiol. Rev. 85 (2)
(2005) 757–810.
[10] R. Maroto, A. Raso, T.G. Wood, A. Kurosky, B. Martinac, O.P. Hamill, TRPC1 forms
the stretch-activated cation channel in vertebrate cells, Nat. Cell Biol. 7 (2) (2005)
179–185.
[11] M.A. Spassova, T. Hewavitharana, W. Xu, J. Soboloff, D.L. Gill, A common me-
chanism underlies stretch activation and receptor activation of TRPC6 channels,
Proc. Natl. Acad. Sci. U. S. A. 103 (44) (2006) 16586–16591.
[12] H. Huang, W. Wang, P. Liu, Y. Jiang, Y. Zhao, H. Wei, W. Niu, TRPC1 expression
and distribution in rat hearts, Eur. J. Histochem. 53 (4) (2009) e26.
[13] X. Wu, P. Eder, B. Chang, J.D. Molkentin, TRPC channels are necessary mediators of
pathologic cardiac hypertrophy, Proc. Natl. Acad. Sci. U. S. A. 107 (15) (2010)
7000–7005.
[14] Y. Jiang, H. Huang, P. Liu, H. Wei, H. Zhao, Y. Feng, W. Wang, W. Niu, Expression
and localization of TRPC proteins in rat ventricular myocytes at various develop-
mental stages, Cell Tissue Res. 355 (1) (2014) 201–212.
[15] M. Goel, C.D. Zuo, W.G. Sinkins, W.P. Schilling, TRPC3 channels colocalize with Na
+/Ca2+ exchanger and Na+ pump in axial component of transverse-axial tubular
system of rat ventricle, Am. J. Physiol. Heart Circ. Physiol. 292 (2) (2007)
H874–H883.
[16] M.L. Ward, I.A. Williams, Y. Chu, P.J. Cooper, Y.K. Ju, D.G. Allen, Stretch-activated
channels in the heart: contributions to length-dependence and to cardiomyopathy,
Prog. Biophys. Mol. Biol. 97 (2–3) (2008) 232–249.
[17] V. Dyachenko, B. Husse, U. Rueckschloss, G. Isenberg, Mechanical deformation of
ventricular myocytes modulates both TRPC6 and Kir2.3 channels, Cell Calcium 45
(1) (2009) 38–54.
[18] M.C. Mohl, S.E. Iismaa, X.H. Xiao, O. Friedrich, S. Wagner, V. Nikolova-Krstevski,
J. Wu, Z.Y. Yu, M. Feneley, D. Fatkin, D.G. Allen, R.M. Graham, Regulation of
murine cardiac contractility by activation of alpha(1A)-adrenergic receptor-oper-
ated ca(2+) entry, Cardiovasc. Res. 91 (2) (2011) 310–319.
[19] J. Fauconnier, J.T. Lanner, A. Sultan, S.J. Zhang, A. Katz, J.D. Bruton,
H. Westerblad, Insulin potentiates TRPC3-mediated cation currents in normal but
not in insulin-resistant mouse cardiomyocytes, Cardiovasc. Res. 73 (2) (2007)
376–385.
[20] A. Kojima, H. Kitagawa, M. Omatsu-Kanbe, H. Matsuura, S. Nosaka, Ca2+ paradox
injury mediated through TRPC channels in mouse ventricular myocytes, Br. J.
Pharmacol. 161 (8) (2010) 1734–1750.
[21] P. Eder, M. Poteser, K. Groschner, TRPC3: a multifunctional, pore-forming signal-
ling molecule, Handb. Exp. Pharmacol. 179 (2007) 77–92.
[22] K. Kuwahara, Y. Wang, J. McAnally, J.A. Richardson, R. Bassel-Duby, J.A. Hill,
E.N. Olson, TRPC6 fulfills a calcineurin signaling circuit during pathologic cardiac
remodeling, J. Clin. Invest. 116 (12) (2006) 3114–3126.
[23] C. Berbey, N. Weiss, C. Legrand, B. Allard, Transient receptor potential canonical
type 1 (TRPC1) operates as a sarcoplasmic reticulum calcium leak channel in ske-
letal muscle, J. Biol. Chem. 284 (52) (2009) 36387–36394.
[24] D.M. Bers, Cardiac sarcoplasmic reticulum calcium leak: basis and roles in cardiac
dysfunction, Annu. Rev. Physiol. 76 (2014) 107–127.
[25] F.B. Sachse, N.S. Torres, E. Savio-Galimberti, T. Aiba, D.A. Kass, G.F. Tomaselli,
J.H. Bridge, Subcellular structures and function of myocytes impaired during heart
failure are restored by cardiac resynchronization therapy, Circ. Res. 110 (4) (2012)
588–597.
[26] T.R. Shannon, K.S. Ginsburg, D.M. Bers, Quantitative assessment of the SR Ca2+
leak-load relationship, Circ. Res. 91 (7) (2002) 594–600.
[27] T.R. Shannon, F. Wang, J. Puglisi, C. Weber, D.M. Bers, A mathematical treatment
of integrated Ca dynamics within the ventricular myocyte, Biophys. J. 87 (5) (2004)
3351–3371.
[28] E. Butterworth, B.E. Jardine, G.M. Raymond, M.L. Neal, J.B. Bassingthwaighte,
JSim, an open-source modeling system for data analysis, F1000Res 2 (2013) 288.
[29] M. Streiff, A.A. Ahmad, C. Hunter, F.B. Sachse, Adult rabbit left ventricle RNAseq,
Gene Expression Omnibus. Series Accession: GSE115605. ID: 200115605, 2018.
[30] G. Isenberg, B. Borschke, U. Rueckschloss, Ca2+ transients of cardiomyocytes from
senescent mice peak late and decay slowly, Cell Calcium 34 (3) (2003) 271–280.
[31] T. Smani, N. Dionisio, J.J. Lopez, A. Berna-Erro, J.A. Rosado, Cytoskeletal and
scaffolding proteins as structural and functional determinants of TRP channels,
Biochim. Biophys. Acta 1838 (2) (2014) 658–664.
[32] J.A. Stiber, Y. Tang, T. Li, P.B. Rosenberg, Cytoskeletal regulation of TRPC channels
in the cardiorenal system, Curr. Hypertens. Rep. 14 (6) (2012) 492–497.
[33] T. Korhonen, S.L. Hanninen, P. Tavi, Model of excitation-contraction coupling of rat
neonatal ventricular myocytes, Biophys. J. 96 (3) (2009) 1189–1209.
[34] C. Pinali, H. Bennett, J.B. Davenport, A.W. Trafford, A. Kitmitto, Three-dimensional
reconstruction of cardiac sarcoplasmic reticulum reveals a continuous network
linking transverse-tubules: this organization is perturbed in heart failure, Circ. Res.
113 (11) (2013) 1219–1230.
[35] C. Soeller, M.B. Cannell, Examination of the transverse tubular system in living
cardiac rat myocytes by 2-photon microscopy and digital image-processing tech-
niques, Circ. Res. 84 (3) (1999) 266–275.
[36] E. Savio-Galimberti, J. Frank, M. Inoue, J.I. Goldhaber, M.B. Cannell, J.H. Bridge,
F.B. Sachse, Novel features of the rabbit transverse tubular system revealed by
quantitative analysis of three-dimensional reconstructions from confocal images,
Biophys. J. 95 (4) (2008) 2053–2062.
[37] O.L. Gervasio, N.P. Whitehead, E.W. Yeung, W.D. Phillips, D.G. Allen, TRPC1 binds
to caveolin-3 and is regulated by Src kinase - role in Duchenne muscular dystrophy,
J. Cell Sci. 121 (Pt 13) (2008) 2246–2255.
[38] J.A. Stiber, Z.S. Zhang, J. Burch, J.P. Eu, S. Zhang, G.A. Truskey, M. Seth,
N. Yamaguchi, G. Meissner, R. Shah, P.F. Worley, R.S. Williams, P.B. Rosenberg,
Mice lacking Homer 1 exhibit a skeletal myopathy characterized by abnormal
transient receptor potential channel activity, Mol. Cell. Biol. 28 (8) (2008)
2637–2647.
[39] A. Dietrich, M. Fahlbusch, T. Gudermann, Classical Transient Receptor Potential 1
(TRPC1): channel or channel regulator? Cells 3 (4) (2014) 939–962.
[40] P. Gottlieb, J. Folgering, R. Maroto, A. Raso, T.G. Wood, A. Kurosky, C. Bowman,
D. Bichet, A. Patel, F. Sachs, B. Martinac, O.P. Hamill, E. Honore, Revisiting TRPC1
and TRPC6 mechanosensitivity, Pflugers Arch. 455 (6) (2008) 1097–1103.
[41] J. Rowell, N. Koitabashi, D.A. Kass, TRP-ing up heart and vessels: canonical tran-
sient receptor potential channels and cardiovascular disease, J. Cardiovasc. Transl.
Res. 3 (5) (2010) 516–524.
[42] T. Ohba, H. Watanabe, M. Murakami, Y. Takahashi, K. Iino, S. Kuromitsu, Y. Mori,
K. Ono, T. Iijima, H. Ito, Upregulation of TRPC1 in the development of cardiac
hypertrophy, J. Mol. Cell. Cardiol. 42 (3) (2007) 498–507.
[43] S.R. Houser, J.D. Molkentin, Does contractile Ca2+ control calcineurin-NFAT sig-
naling and pathological hypertrophy in cardiac myocytes? Sci. Signal. 1 (25) (2008)
pe31.
[44] K. Seo, P.P. Rainer, D.I. Lee, S. Hao, D. Bedja, L. Birnbaumer, O.H. Cingolani,
D.A. Kass, Hyperactive adverse mechanical stress responses in dystrophic heart are
coupled to transient receptor potential canonical 6 and blocked by cGMP-protein
kinase G modulation, Circ. Res. 114 (5) (2014) 823–832.
[45] M. Schaefer, TRPs: modulation by drug-like compounds, Handb. Exp. Pharmacol.
223 (2014) 1077–1106.
Q. Hu, et al. Journal of Molecular and Cellular Cardiology 139 (2020) 113–123
123
... The mechanism of toxic action of unsaturated aldehydes on CM is associated with their high electrophilicity and the ability to covalently bind to cysteine residues in the TRPA1 molecule, leads to the opening of these channels and an increase sarcoplasmic reticulum Ca2+ release flux into the cytosol [116]. Overexpression of TRPС1 channels also contributes to Ca2+ leakage from SR [117]. As a result, Ca2+ overload of СM, leading to impaired contractility, heart failure and myocardial infarction occurs [117]. ...
... Overexpression of TRPС1 channels also contributes to Ca2+ leakage from SR [117]. As a result, Ca2+ overload of СM, leading to impaired contractility, heart failure and myocardial infarction occurs [117]. ...
Preprint
Full-text available
Cardiovascular diseases (CVD) account for about 17.3 million annual deaths worldwide. 85% of these deaths occurred as a result of myocardial infarction (MI) and stroke. Chronic heart diseases, such as arterial hypertension (AH), coronary heart disease, various cerebrovascular diseases, dilated and hypertrophic cardiomyopathies are widespread, with a fairly high incidence of mortality and disability. Most of these diseases are characterized by cardiac arrhythmias, conduction and contractility disorders. Additionally, interruption of the electrical activity of the heart, the appearance of extensive ectopic foci and heart failure are all symptoms of a number of severe hereditary diseases. The molecular mechanisms leading to the development of CVD are associated with impaired permeability and excitability of cell membranes and mainly caused by dysfunction of cardiac Ca2+ channels. Acquired channelopathies are caused by metabolic disorders, increased tone of the sympathetic nervous system, age-related changes that lead to deterioration of coronary blood flow and hemodynamics. Currently known channelopathies, such as the long or short QT syndromes, Brugada and Lenegre syndromes, catecholaminergic polymorphic ventricular tachycardia, etc., are congenital and genetic disorders caused by mutations of genes "responsible" for the conductive properties of certain channel-forming proteins, including pore-forming subunits of Ca2+ channels. Over the past 50 years, more than 100 varieties of ion channels have been found in the cardiovascular system cells. The relationship between the activity of these channels and cardiac pathology, as well as the general cellular biological function, has been intensively studied on several cell types and experimental animal models in vivo и in situ. In this review, we discuss the origin of Ca2+ channelopathies and the role of Ca2+ channels of various types: L- R-, T-types voltage-gated calcium channels, RyR2, non-selective hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, transient receptor potential (TRPC, TRPM7, TRPA1) channels in the development of cardio-vascular pathology.
... Members of the TRP family have also been suggested to contribute to SR Ca 2+ leak. TRPC1 was found to operate as a SR Ca 2+ leak channel in skeletal muscle (Berbey et al., 2009) and more recently in cardiomyocytes (Hu et al., 2020). Additional evidence suggests contribution of TRPC6, TRPM8, TRPP2, and TRPV1 to endoplasmic reticulum Ca 2+ leak in various cell types, although characterization is still incomplete for cardiomyocytes (Lemos et al., 2021). ...
... Non-RyR leak that can be modulated by stretch may provide a more moderate and steady regulation on a beat-by-beat basis in conjunction with background Ca 2+ entry modulated by stretch. Recently we demonstrated that TRPC1 constitutes an SR Ca 2+ leak channel, and its overexpression resulted in decreased SR Ca 2+ load (Hu et al., 2020). TRPC1 channels are suggested to be modulated by stretch, indicating that the reduction in SR Ca 2+ load could be a regulatory mechanism to match increased background Ca 2+ entry through, e.g., TRPC6 channels. ...
Article
Full-text available
The intricate regulation of the compartmental Ca2+ concentrations in cardiomyocytes is critical for electrophysiology, excitation-contraction coupling, and other signaling pathways. Research into the complex signaling pathways is motivated by cardiac pathologies including arrhythmia and maladaptive myocyte remodeling, which result from Ca2+ dysregulation. Of interest to this investigation are two types of Ca2+ currents in cardiomyocytes: 1) background Ca2+ entry, i.e., Ca2+ transport across the sarcolemma from the extracellular space into the cytosol, and 2) Ca2+ leak from the sarcoplasmic reticulum (SR) across the SR membrane into the cytosol. Candidates for the ion channels underlying background Ca2+ entry and SR Ca2+ leak channels include members of the mechano-modulated transient receptor potential (TRP) family. We used a mathematical model of a human ventricular myocyte to analyze the individual contributions of background Ca2+ entry and SR Ca2+ leak to the modulation of Ca2+ transients and SR Ca2+ load at rest and during action potentials. Background Ca2+ entry exhibited a positive relationship with both [Ca2+]i and [Ca2+]SR. Modulating SR Ca2+ leak had opposite effects of background Ca2+ entry. Effects of SR Ca2+ leak on Ca2+ were particularly pronounced at lower pacing frequency. In contrast to the pronounced effects of background and leak Ca2+ currents on Ca2+ concentrations, the effects on cellular electrophysiology were marginal. Our studies provide quantitative insights into the differential modulation of compartmental Ca2+ concentrations by the background and leak Ca2+ currents. Furthermore, our studies support the hypothesis that TRP channels play a role in strain-modulation of cardiac contractility. In summary, our investigations shed light on the physiological effects of the background and leak Ca2+ currents and their contribution to the development of disease caused by Ca2+ dysregulation.
... It is known the TRPA channel painless is required for mechanotransduction in the Drosophila heart 64 , and strong genetic interaction between painless and Piezo in the context of detection of mechanical nociception in Drosophila sensory neurons 29 hint that they might also be part of the same pathway in the Drosophila heart. A recent paper showing that TRPC1 localises to the SR in rat cardiomyocytes lends additional support to this hypothesis 65 . A further candidate is the recently described Piezo-like channel 66 , however it is not known whether Piezo-like is expressed in Drosophila cardiomyocytes. ...
... There are indications that TRP channels might be involved, e.g. TRPC1, TRPC3 and TRPC6 are both expressed at high levels in the heart, with evidence that TRPC3 and TRPC6 contribute to stretch-induced SFR 25,65,[72][73][74] . Piezo has also been considered a candidate as Piezo1 is found in the vertebrate cardiovascular system and has been shown to be upregulated upon myocardial infarction 28,[75][76][77] . ...
Preprint
Full-text available
Throughout its lifetime the heart is buffeted continuously by dynamic mechanical forces resulting from contraction of the heart muscle itself and fluctuations in haemodynamic load and pressure. These forces are in flux on a beat-by-beat basis, resulting from changes in posture, physical activity or emotional state, and over longer timescales due to altered physiology (e.g. pregnancy) or as a consequence of ageing or disease (e.g. hypertension). It has been known for over a century of the heart′s ability to sense differences in haemodynamic load and adjust contractile force accordingly. These adaptive behaviours are important for cardiovascular homeostasis, but the mechanism(s) underpinning them are incompletely understood. Here we present evidence that the mechanically-activated ion channel, Piezo, is an important component of the Drosophila heart′s ability to adapt to mechanical force. We find Piezo is a sarcoplasmic reticulum (SR)-resident channel and is part of a mechanism that regulates Ca2+ handling in cardiomyocytes in response to mechanical stress. Our data support a simple model in which Drosophila Piezo transduces mechanical force such as stretch into a Ca2+ signal, originating from the SR, that modulates cardiomyocyte contraction. We show that Piezo mutant hearts fail to buffer mechanical stress, have altered Ca2+ handling, become prone to arrhythmias and undergo pathological remodelling.
... Thus, Ca 2+ -permeable TRP channels may serve as potential novel targets for developing anti-fibrotic drugs [7]. Hu et al. [8] also found that the TRPC1 protein is abundantly expressed in cardiomyocytes and is closely related to cardiac hypertrophy, which can act as a regulator of Ca 2+ concentration and contractility of mammalian cardiomyocytes. Kuwahara et al. [9] showed that TRPC6 fulfils a calcineurin signalling circuit during pathologic cardiac remodelling. ...
Article
Full-text available
Background: Diabetes mellitus type 2 is a risk factor for developing heart failure and myocardial fibrosis, but there is no specific therapy for diabetic heart disease. 1-[2-(4-methoxyphenyl)]-2-[3-(4-methoxyphenyl) propoxy]ethyl-1H-imidazole (SKF96365) is regarded as an inhibitor of receptor-mediated calcium ion (Ca2+) entry. This study aimed to explore the effects of SKF96365 on diabetic myocardial fibrosis. Methods: A type 2 diabetic rat model induced by a high-sugar and high-fat diet combined with streptozotocin was established. Thirty specific pathogen-free male Wistar rats were divided randomly into three groups: group A (the blank control group), group B (the diabetes group) and group C (the diabetes + transient receptor potential canonical channel [TRPC] blocker intervention group). Group C was given 0.74-µmol/kg SKF96365 by intraperitoneal injection, and groups A and B were given the same amount of normal saline by intraperitoneal injection. The weight and blood sugar of the rats were monitored. After 12 weeks, the weight of the whole heart and the left ventricle was measured, and the heart and the left ventricular weight ratios were calculated. Haematoxylin-eosin (HE) staining was used to observe pathological changes in the myocardial tissue and the distribution of nuclei. Masson staining was used to identify collagen and muscle fibres, and the myocardial collagen volume fraction (CVF) was calculated. Semi-quantitative reverse transcription-polymerase chain reaction was used to detect the messenger ribonucleic acid (mRNA) expression of SKF96365 target genes. A value of p < 0.05 indicated that the difference between the groups was statistically significant. Results: Compared with the weight of the rats in group A, the weight of those in groups B and C decreased, while blood sugar, whole heart weight and left ventricular weight increased (p < 0.05). There was no significant difference in body weight between the rats in groups B and C (p > 0.05). The HE staining results showed that the arrangement of cardiomyocytes in groups B and C was irregular, and focal necrosis was seen in severe cases. The degree of diabetic cardiomyopathy (DCM) in group C was less severe than that in group B. Masson staining showed that the CVF increased in groups B and C, with group B > group C (p < 0.05); the mRNA expressions of TRPC3 and TRPC6 were upregulated in groups A, B and C, and the mRNA expressions of TRPC3 and TRPC6 in group C were downregulated compared with those in group B (p < 0.05). Compared with the expression levels of SKF96365 target genes (STIM1, Orai1 and Homer1) in group A, those in group B were lower, while the administration of SKF96365 in group C did not affect the expression levels of those genes. Conclusions: SKF96365 can effectively improve myocardial fibrosis in type-II diabetic rats.
... Upregulation of TRPC1 contributed to the development of cardiac hypertrophy [34,35]. TRPC1 might regulate Ca 2+ leakage from the ER in neonatal rat ventricular myocytes [36]. However, it remains unclear whether TRPC1 plays a role in modulating Ca 2+ signaling in cardiac fibroblasts. ...
Article
Full-text available
Fibroblast growth factor (FGF)-23 induces hypertrophy and calcium (Ca2+) dysregulation in cardiomyocytes, leading to cardiac arrhythmia and heart failure. However, knowledge regarding the effects of FGF-23 on cardiac fibrogenesis remains limited. This study investigated whether FGF-23 modulates cardiac fibroblast activity and explored its underlying mechanisms. We performed MTS analysis, 5-ethynyl-2’-deoxyuridine assay, and wound-healing assay in cultured human atrial fibroblasts without and with FGF-23 (1, 5 and 25 ng/mL for 48 h) to analyze cell proliferation and migration. We found that FGF-23 (25 ng/mL, but not 1 or 5 ng/mL) increased proliferative and migratory abilities of human atrial fibroblasts. Compared to control cells, FGF-23 (25 ng/mL)-treated fibroblasts had a significantly higher Ca2+ entry and intracellular inositol 1,4,5-trisphosphate (IP3) level (assessed by fura-2 ratiometric Ca2+ imaging and enzyme-linked immunosorbent assay). Western blot analysis showed that FGF-23 (25 ng/mL)-treated cardiac fibroblasts had higher expression levels of calcium release-activated calcium channel protein 1 (Orai1) and transient receptor potential canonical (TRPC) 1 channel, but similar expression levels of α-smooth muscle actin, collagen type IA1, collagen type Ⅲ, stromal interaction molecule 1, TRPC 3, TRPC6 and phosphorylated-calcium/calmodulin-dependent protein kinase II when compared with control fibroblasts. In the presence of ethylene glycol tetra-acetic acid (a free Ca2+ chelator, 1 mM) or U73122 (an inhibitor of phospholipase C, 1 μM), control and FGF-23-treated fibroblasts exhibited similar proliferative and migratory abilities. Moreover, polymerase chain reaction analysis revealed that atrial fibroblasts abundantly expressed FGF receptor 1 but lacked expressions of FGF receptors 2-4. FGF-23 significantly increased the phosphorylation of FGF receptor 1. Treatment with PD166866 (an antagonist of FGF receptor 1, 1 μM) attenuated the effects of FGF-23 on cardiac fibroblast activity. In conclusion, FGF-23 may activate FGF receptor 1 and subsequently phospholipase C/IP3 signaling pathway, leading to an upregulation of Orai1 and/or TRPC1-mediated Ca2+ entry and thus enhancing human atrial fibroblast activity.
Article
Full-text available
Chronic heart diseases, such as coronary heart disease, heart failure, secondary arterial hypertension, and dilated and hypertrophic cardiomyopathies, are widespread and have a fairly high incidence of mortality and disability. Most of these diseases are characterized by cardiac arrhythmias, conduction, and contractility disorders. Additionally, interruption of the electrical activity of the heart, the appearance of extensive ectopic foci, and heart failure are all symptoms of a number of severe hereditary diseases. The molecular mechanisms leading to the development of heart diseases are associated with impaired permeability and excitability of cell membranes and are mainly caused by the dysfunction of cardiac Ca2+ channels. Over the past 50 years, more than 100 varieties of ion channels have been found in the cardiovascular cells. The relationship between the activity of these channels and cardiac pathology, as well as the general cellular biological function, has been intensively studied on several cell types and experimental animal models in vivo and in situ. In this review, I discuss the origin of genetic Ca2+ channelopathies of L- and T-type voltage-gated calcium channels in humans and the role of the non-genetic dysfunctions of Ca2+ channels of various types: L-, R-, and T-type voltage-gated calcium channels, RyR2, including Ca2+ permeable nonselective cation hyperpolarization-activated cyclic nucleotide-gated (HCN), and transient receptor potential (TRP) channels, in the development of cardiac pathology in humans, as well as various aspects of promising experimental studies of the dysfunctions of these channels performed on animal models or in vitro.
Article
Full-text available
Transient receptor potential canonical 1 (TRPC1) channels are Ca2+-permeable ion channels expressed in cardiomyocytes. An involvement of TRPC1 channels in cardiac diseases is widely established. However, the physiological role of TRPC1 channels and the mechanisms through which they contribute to disease development are still under investigation. Our prior work suggested that TRPC1 forms Ca2+ leak channels located in the sarcoplasmic reticulum (SR) membrane. Prior studies suggested that TRPC1 channels in the cell membrane are mechanosensitive, but this was not yet investigated in cardiomyocytes or for SR localized TRPC1 channels. We applied adenoviral transfection to overexpress or suppress TRPC1 expression in neonatal rat ventricular myocytes (NRVMs). Transfections were evaluated with RT-qPCR, western blot, and fluorescent imaging. Single-molecule localization microscopy revealed high colocalization of exogenously expressed TRPC1 and the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA2). To test our hypothesis that TRPC1 channels contribute to mechanosensitive Ca2+ SR leak, we directly measured SR Ca2+ concentration ([Ca2+]SR) using adenoviral transfection with a novel ratiometric genetically encoded SR-targeting Ca2+ sensor. We performed fluorescence imaging to quantitatively assess [Ca2+]SR and leak through TRPC1 channels of NRVMs cultured on stretchable silicone membranes. [Ca2+]SR was increased in cells with suppressed TRPC1 expression vs. control and Transient receptor potential canonical 1-overexpressing cells. We also detected a significant reduction in [Ca2+]SR in cells with Transient receptor potential canonical 1 overexpression when 10% uniaxial stretch was applied. These findings indicate that TRPC1 channels underlie the mechanosensitive modulation of [Ca2+]SR. Our findings are critical for understanding the physiological role of TRPC1 channels and support the development of pharmacological therapies for cardiac diseases.
Article
Full-text available
Throughout its lifetime the heart is buffeted continuously by dynamic mechanical forces resulting from contraction of the heart muscle itself and fluctuations in haemodynamic load and pressure. These forces are in flux on a beat-by-beat basis, resulting from changes in posture, physical activity or emotional state, and over longer timescales due to altered physiology (e.g. pregnancy) or as a consequence of ageing or disease (e.g. hypertension). It has been known for over a century of the heart’s ability to sense differences in haemodynamic load and adjust contractile force accordingly (Frank, Z. biology, 1895, 32, 370–447; Anrep, J. Physiol., 1912, 45 (5), 307–317; Patterson and Starling, J. Physiol., 1914, 48 (5), 357–79; Starling, The law of the heart (Linacre Lecture, given at Cambridge, 1915), 1918). These adaptive behaviours are important for cardiovascular homeostasis, but the mechanism(s) underpinning them are incompletely understood. Here we present evidence that the mechanically-activated ion channel, Piezo, is an important component of the Drosophila heart’s ability to adapt to mechanical force. We find Piezo is a sarcoplasmic reticulum (SR)-resident channel and is part of a mechanism that regulates Ca2+ handling in cardiomyocytes in response to mechanical stress. Our data support a simple model in which Drosophila Piezo transduces mechanical force such as stretch into a Ca2+ signal, originating from the SR, that modulates cardiomyocyte contraction. We show that Piezo mutant hearts fail to buffer mechanical stress, have altered Ca2+ handling, become prone to arrhythmias and undergo pathological remodelling.
Article
Cardiac dysfunction is a vital complication of endotoxemia (ETM) with limited therapeutic options. Transient receptor potential canonical channel (TRPC)1 was involved in various heart diseases. While, the role of TRPC1 in ETM-induced cardiac dysfunction remains to be defined. In this study, we found that TRPC1 protein expression was significantly upregulated in hearts of lipopolysaccharide (LPS)-challenged mice. What’s more, TRPC1 knockdown significantly alleviated LPS-induced cardiac dysfunction and injury. Further myocardial mRNA-sequencing analysis revealed that TRPC1 might participate in pathogenesis of ETM-induced cardiac dysfunction via mediating myocardial apoptosis and autophagy. Data showed that knockdown of TRPC1 significantly ameliorated LPS-induced myocardial apoptotic injury, cardiomyocytes autophagosome accumulation, and myocardial autophagic flux. Simultaneously, deletion of TRPC1 reversed LPS-induced molecular changes of apoptosis/autophagy signaling pathway in cardiomyocytes. Moreover, TRPC1 could promote LPS-triggered intracellular Ca²⁺ release, subsequent calpain activation and caveolin-1 degradation. Either blocking calpain by PD150606 or enhancing the amount of caveolin-1 scaffolding domain that interacts with TRPC1 by cell-permeable peptide cavtratin significantly alleviated the LPS-induced cardiac dysfunction and cardiomyocytes apoptosis/autophagy. Furthermore, cavtratin could inhibit LPS-induced calpain activation in cardiomyocytes. caveolin-1 could directly interact with calpain 2 both in vivo and in vitro. Importantly, cecal ligation and puncture-stimulated cardiac dysfunction and mortality were significantly alleviated in Trpc1-/- and cavtratin-treated mice, which further validated the contribution of TRPC1-caveolin-1 signaling axis in sepsis-induced pathological process. Overall, this study indicated that TRPC1 could promote LPS-triggered intracellular Ca²⁺ release, mediate caveolin-1 reduction, and in turn activates calpain to regulate myocardial apoptosis and autophagy, contributing to ETM-induced cardiac dysfunction of mice.
Article
Full-text available
Located at the level of the endoplasmic reticulum (ER) membrane, stromal interacting molecule 1 (STIM1) undergoes a complex conformational rearrangement after depletion of ER luminal Ca2+. Then, STIM1 translocates into discrete ER-plasma membrane (PM) junctions where it directly interacts with and activates plasma membrane Orai1 channels to refill ER with Ca2+. Furthermore, Ca2+ entry due to Orai1/STIM1 interaction may induce canonical transient receptor potential channel 1 (TRPC1) translocation to the plasma membrane, where it is activated by STIM1. All these events give rise to store-operated calcium entry (SOCE). Besides the main pathway underlying SOCE, which mainly involves Orai1 and TRPC1 activation, STIM1 modulates many other plasma membrane proteins in order to potentiate the influxof Ca2+. Furthermore, it is now clear that STIM1 may inhibit Ca2+ currents mediated by L-type Ca2+ channels. Interestingly, STIM1 also interacts with some intracellular channels and transporters, including nuclear and lysosomal ionic proteins, thus orchestrating organellar Ca2+ homeostasis. STIM1 and its partners/effectors are significantly modulated in diverse acute and chronic neurodegenerative conditions. This highlights the importance of further disclosing their cellular functions as they might represent promising molecular targets for neuroprotection.
Article
Full-text available
Transient receptor potential canonical (TRPC) proteins have been identified as a family of plasma membrane calcium-permeable channels. TRPC proteins can be activated by various stimuli and act as cellular sensors in mammals. Stretch-activated ion channels (SACs) have been proposed to underlie cardiac mechano-electric feedback (MEF), although the molecular entity of SAC remains unknown. There is evidence suggesting that transient receptor potential canonical 1 (TRPC1) is a stretch-activated ion channel. As a non-selective cation channel, TRPC1 may cause stretch-induced depolarization and arrhythmia and thus may contribute to the MEF of the heart. In this study, we examined the expression patterns of TRPC1 in detail at both the mRNA and protein levels in rat hearts.We isolated total RNA from the left and right atria, and the left and right ventricles, and detected TRPC1 mRNA in these tissues using reverse-transcriptase polymerase chain reaction (RT-PCR). To study the protein localization and targeting, we performed immunohistochemistry and immunofluorescence labeling with the antibody against TRPC1. TRPC1 was detected in the cardiomyocytes of the ventricle and atrium at both the mRNA and protein levels. The cell membrane and Ttubule showed strong fluorescence labeling in the ventricular myocytes. Purkinje cells, the endothelial cells and smooth muscle cells of the coronary arterioles also displayed TRPC1 labeling. No TRPC1 was detected in fibroblasts. In conclusion, TRPC1 is widely expressed in the rat heart, including in working cells, Purkinje cells and vascular cells, suggesting that it plays an important role in the heart. The specific distribution pattern offered a useful insight into its function in adult rat ventricular cells. Further investigations are needed to clarify the role of TRPC1 in regulating cardiac activity, including cardiac MEF.
Article
Full-text available
This article addresses whether TRPC1 or TRPC6 is an essential component of a mammalian stretch-activated mechano-sensitive Ca(2+) permeable cation channel (MscCa). We have transiently expressed TRPC1 and TRPC6 in African green monkey kidney (COS) or Chinese hamster ovary (CHO) cells and monitored the activity of the stretch-activated channels using a fast pressure clamp system. Although both TRPC1 and TRPC6 are highly expressed at the protein level, the amplitude of the mechano-sensitive current is not significantly altered by overexpression of these subunits. In conclusion, although several TRPC channel members, including TRPC1 and TRPC6, have been recently proposed to form MscCa in vertebrate cells, the functional expression of these TRPC subunits in heterologous systems remains problematic.
Article
Full-text available
In contrast to other Classical Transient Receptor Potential TRPC channels the function of TRPC1 as an ion channel is a matter of debate, because it is often difficult to obtain substantial functional signals over background in response to over-expression of TRPC1 alone. Along these lines, heterologously expressed TRPC1 is poorly translocated to the plasma membrane as a homotetramer and may not function on its own physiologically, but may rather be an important linker and regulator protein in heteromeric TRPC channel tetramers. However, due to the lack of specific TRPC1 antibodies able to detect native TRPC1 channels in primary cells, identification of functional TRPC1 containing heteromeric TRPC channel complexes in the plasma membrane is still challenging. Moreover, an extended TRPC1 cDNA, which was recently discovered, may seriously question results obtained in heterologous expression systems transfected with shortened cDNA versions. Therefore, this review will focus on the current status of research on TRPC1 function obtained in primary cells and a TRPC1-deficient mouse model.
Article
Full-text available
JSim is a simulation system for developing models, designing experiments, and evaluating hypotheses on physiological and pharmacological systems through the testing of model solutions against data. It is designed for interactive, iterative manipulation of the model code, handling of multiple data sets and parameter sets, and for making comparisons among different models running simultaneously or separately. Interactive use is supported by a large collection of graphical user interfaces for model writing and compilation diagnostics, defining input functions, model runs, selection of algorithms solving ordinary and partial differential equations, run-time multidimensional graphics, parameter optimization (8 methods), sensitivity analysis, and Monte Carlo simulation for defining confidence ranges. JSim uses Mathematical Modeling Language (MML) a declarative syntax specifying algebraic and differential equations. Imperative constructs written in other languages (MATLAB, FORTRAN, C++, etc.) are accessed through procedure calls. MML syntax is simple, basically defining the parameters and variables, then writing the equations in a straightforward, easily read and understood mathematical form. This makes JSim good for teaching modeling as well as for model analysis for research. For high throughput applications, JSim can be run as a batch job. JSim can automatically translate models from the repositories for Systems Biology Markup Language (SBML) and CellML models. Stochastic modeling is supported. MML supports assigning physical units to constants and variables and automates checking dimensional balance as the first step in verification testing. Automatic unit scaling follows, e.g. seconds to minutes, if needed. The JSim Project File sets a standard for reproducible modeling analysis: it includes in one file everything for analyzing a set of experiments: the data, the models, the data fitting, and evaluation of parameter confidence ranges. JSim is open source; it and about 400 human readable open source physiological/biophysical models are available at http://www.physiome.org/jsim/.
Article
Full-text available
Significance Cardiac hypertrophy and dysfunction in response to sustained hormonal and mechanical stress are sentinel features of most forms of heart disease. Activation of non–voltage-gated transient receptor potential canonical channels TRPC3 and TRPC6 may contribute to this pathophysiology and provide a therapeutic target. Effects from combined selective inhibition have not been tested previously. Here we report the capability of highly selective TRPC3/6 inhibitors to block pathological hypertrophic signaling in several cell types, including adult cardiac myocytes. We show in vivo redundancy of each channel; individual gene deletion was not protective against sustained pressure overload, whereas combined deletion ameliorated the response. These data strongly support a role for both channels in cardiac disease and the utility of selective combined inhibition.
Article
Transient receptor potential canonical (TRPC) channels constitute a family of seven Ca²⁺ permeable ion channels, named TRPC1 to 7. These channels are abundantly expressed in the mammalian heart, yet mechanisms underlying activation of TRPC channels and their precise role in cardiac physiology remain poorly understood. In this review, we perused original literature regarding TRPC channels in cardiomyocytes. We first reviewed studies on TRPC channel assembly and sub-cellular localization across multiple species and cell types. Our review indicates that TRPC localization in cardiac cells is still a topic of controversy. We then examined common molecular biology tools used to infer on location and physiological roles of TRPC channels in the heart. We subsequently reviewed pharmacological tools used to modulate TRPC activity in both cardiac and non-cardiac cells. Suggested physiological roles in the heart include modulation of heart rate and sensing of mechanical strain. We examined studies on the contribution of TRPC to cardiac pathophysiology, mainly hypertrophic signaling. Several TRPC channels, particularly TRPC1, 3 and 6 were proposed to play a crucial role in hypertrophic signaling. Finally, we discussed gaps in our understanding of the location and physiological role of TRPC channels in cardiomyocytes. Closing these gaps will be crucial to gain a full understanding of the role of TRPC channels in cardiac pathophysiology and to further explore these channels as targets for treatments for cardiac diseases, in particular, hypertrophy.
Article
Drug-like compounds that exert biological activity towards TRP channels are either being used as cell biological tools or further developed into pharmacological lead structures aiming at therapeutic use in diseased states. Although drug-likeliness is not easy to predict, common rules include a relatively low molecular weight, physicochemical constraints, and the absence of known reactive or otherwise toxic groups. Small molecules that exert a biological activity to block, activate, or modulate TRP channels are intensely sought. Such tool compounds may be useful to assign native currents to a certain TRP channel and to validate the channel as a candidate target for future pharmacological intervention. Depending on the TRP channel isotype, these activities have reached different levels, with only few TRP channels modulators already being clinically tested in humans, whereas other compounds only underwent a preliminary validation. For some TRP channels, reliable low molecular weight inhibitors are not yet available. Hence, further efforts need to be undertaken in order to explore the physiological impact and possible therapeutic potential of TRP channel targeting with drug-like compounds.
Article
The heart is exquisitely sensitive to mechanical stimuli in order to rapidly adapt to physiological demands. In muscle lacking dystrophin, such as Duchenne muscular dystrophy (DMD), increased load during contraction triggers pathological responses thought to worsen the disease. The relevant mechano-transducers and therapies to target them remain unclear. We tested the role of transient receptor potential canonical channels TRPC3 and TRPC6 and their modulation by protein kinase G in controlling cardiac systolic mechano-sensing, and determined their pathophysiological relevance in an experimental model of DMD. Contracting isolated papillary muscles and/or cardiomyocytes from controls and mice genetically lacking either TRPC3 or TRPC6 were subjected to auxotonic load to induce stress-stimulated contractility (SSC, gradual rise in force and intracellular Ca(2+)). Incubation with cGMP (PKG activator) markedly blunted SSC in controls and Trpc3(-/-); whereas in Trpc6(-/-), the resting SSC response was diminished and cGMP had no impact. In DMD myocytes (mdx/utrophin deficient), the SSC was excessive and arrhythmogenic. Gene deletion or selective drug blockade of TRPC6, or cGMP/PKG activation, all reversed this phenotype. Chronic PDE5A inhibition also normalized abnormal mechano-sensing while blunting progressive chamber hypertrophy in DMD mice. PKG is a potent negative-modulator of cardiac systolic mechano-signaling that requires TRPC6 as the target effector. In dystrophic hearts, excess SSC and arrhythmia are coupled to TRPC6 and are ameliorated by its targeted suppression or PKG activation. These results highlight novel therapeutic targets for this disease.
Article
Synchronized SR calcium (Ca) release is critical to normal cardiac myocyte excitation-contraction coupling, and ideally this release shuts off completely between heartbeats. However, other SR Ca release events are referred to collectively as SR Ca leak (which includes Ca sparks and waves as well as smaller events not detectable as Ca sparks). Much, but not all, of the SR Ca leak occurs via ryanodine receptors and can be exacerbated in pathological states such as heart failure. The extent of SR Ca leak is important because it can (a) reduce SR Ca available for release, causing systolic dysfunction; (b) elevate diastolic [Ca]i, contributing to diastolic dysfunction; (c) cause triggered arrhythmias; and (d) be energetically costly because of extra ATP used to repump Ca. This review addresses quantitative aspects and manifestations of SR Ca leak and its measurement, and how leak is modulated by Ca, associated proteins, and posttranslational modifications in health and disease. Expected final online publication date for the Annual Review of Physiology Volume 76 is February 10, 2014. Please see http://www.annualreviews.org/catalog/pubdates.aspx for revised estimates.
Article
Growing evidence indicates that transient receptor potential canonical (TRPC) channels play important roles in various Ca(2+)-mediated physiological and pathophysiological processes, including development. Many types of TRPC proteins are expressed in the heart. However, limited data are available comparing the expression and localization among TRPC proteins in the ventricular myocyte at various developmental stages. Our purpose is to investigate the expression and localization profile of TRPC proteins in ventricular myocytes of fetal (18.5 days), neonatal (< 24 h after birth) and adult (8 week old) rats. Western blotting, immunofluorescence and confocal laser scanning microscopy were employed. TRPC1/3-6 proteins were expressed in the rat ventricle throughout the three developmental stages. The expression profile of TRPC1/3/4 in the ventricle followed an upward trend from the fetus to the adult. By contrast, TRPC6 in the ventricle was expressed at the highest level in the fetal group and was sharply down-regulated immediately after birth. TRPC5 expression in the ventricle did not change significantly during the three stages. TRPC1/3/5/6 proteins were localized to the T-tubule and TRPC1/3/4/6 to intercalated disks in adult myocytes. The wide spatiotemporal overlap and dynamic regulation of TRPC expression in ventricular myocytes indicates potential complex combinations and redundancy of native TRPC proteins in the heart and gives important clues for further investigations into the exact subunit compositions and functional properties of native TRPC channels in the heart.