ArticlePDF Available

Canonical Transient Receptor Potential Channels Promote Cardiomyocyte Hypertrophy through Activation of Calcineurin Signaling

Authors:

Abstract

The calcium/calmodulin-dependent phosphatase calcineurin plays a central role in the control of cardiomyocyte hypertrophy in response to pathological stimuli. Although calcineurin is present at high levels in normal heart, its activity appears to be unaffected by calcium during the course of a cardiac cycle. The mechanism(s) whereby calcineurin is selectively activated by calcium under pathological conditions has remained unclear. Here, we demonstrate that diverse signals for cardiac hypertrophy stimulate expression of canonical transient receptor potential (TRPC) channels. TRPC consists of a family of seven membrane-spanning nonselective cation channels that have been implicated in the nonvoltage-gated influx of calcium in response to G protein-coupled receptor signaling, receptor tyrosine kinase signaling, and depletion of internal calcium stores. TRPC3 expression is up-regulated in multiple rodent models of pathological cardiac hypertrophy, whereas TRPC5 expression is induced in failing human heart. We demonstrate that TRPC promotes cardiomyocyte hypertrophy through activation of calcineurin and its downstream effector, the nuclear factor of activated T cells transcription factor. These results define a novel role for TRPC channels in the control of cardiac growth, and suggest that a TRPC-derived pool of calcium contributes to selective activation of calcineurin in diseased heart.
Canonical Transient Receptor Potential Channels Promote
Cardiomyocyte Hypertrophy through Activation
of Calcineurin Signaling
*
Received for publication, June 9, 2006, and in revised form, August 31, 2006 Published, JBC Papers in Press, September 1, 2006, DOI 10.1074/jbc.M605536200
Erik W. Bush
‡1
, David B. Hood
, Philip J. Papst
, Joseph A. Chapo
, Wayne Minobe
§
, Michael R. Bristow
§
,
Eric N. Olson
¶2
, and Timothy A. McKinsey
‡3
From
Myogen, Inc., Westminster, Colorado 80021, the
§
Division of Cardiology, University of Colorado Health Sciences Center,
Denver, Colorado 80262, and the
University of Texas Southwestern Medical Center, Dallas, Texas 75390
The calcium/calmodulin-dependent phosphatase calcineurin
plays a central role in the control of cardiomyocyte hypertrophy
in response to pathological stimuli. Although calcineurin is
present at high levels in normal heart, its activity appears to be
unaffected by calcium during the course of a cardiac cycle. The
mechanism(s) whereby calcineurin is selectively activated by
calcium under pathological conditions has remained unclear.
Here, we demonstrate that diverse signals for cardiac hypertro-
phy stimulate expression of canonical transient receptor poten-
tial (TRPC) channels. TRPC consists of a family of seven mem-
brane-spanning nonselective cation channels that have been
implicated in the nonvoltage-gated influx of calcium in response
to G protein-coupled receptor signaling, receptor tyrosine
kinase signaling, and depletion of internal calcium stores.
TRPC3 expression is up-regulated in multiple rodent models of
pathological cardiac hypertrophy, whereas TRPC5 expression is
induced in failing human heart. We demonstrate that TRPC
promotes cardiomyocyte hypertrophy through activation of cal-
cineurin and its downstream effector, the nuclear factor of acti-
vated T cells transcription factor. These results define a novel
role for TRPC channels in the control of cardiac growth, and
suggest that a TRPC-derived pool of calcium contributes to
selective activation of calcineurin in diseased heart.
Cardiac hypertrophy is an adaptive response of the heart to
many forms of cardiac disease, including hypertension,
mechanical load abnormalities, myocardial infarction, valvular
dysfunction, cardiac arrhythmias, endocrine disorders, and
genetic mutations in cardiac contractile protein genes. Whereas
the hypertrophic response is thought to be an initially compensa-
tory mechanism that augments cardiac performance, sustained
hypertrophy is maladaptive and frequently leads to ventricular
dilation and the clinical syndrome of heart failure. Accordingly,
cardiac hypertrophy has been established as an independent risk
factor for cardiac morbidity and mortality (1).
Abnormal calcium handling, characterized by elevated intra-
cellular diastolic calcium levels, is a hallmark of cardiac hyper-
trophy and heart failure. Elevated intracellular calcium not only
impairs the contractile performance of the heart, but also acti-
vates calcium-dependent signaling pathways that mediate mal-
adaptive cardiac remodeling (2). One such pathway is regu-
lated by the calcium-calmodulin-dependent phosphatase
calcineurin, which has been shown to be sufficient, and in some
cases, necessary for pathological hypertrophy (3–5). Activated
calcineurin dephosphorylates the transcription factor nuclear
factor of activated T-cells (NFAT),
4
facilitating translocation of
NFAT to the nucleus where it acts in concert with other pro-
teins to mediate expression of prohypertrophic genes. The
activity of calcineurin is tightly regulated in vivo by a negative
feedback mechanism; one of the most highly sensitive NFAT
target genes encodes a potent calcineurin inhibitor, modulatory
calcineurin-interacting protein 1 (MCIP1) (68), also known
as Down syndrome critical region 1.
How pathological stress is translated into a calcium stimulus
capable of activating cardiac calcineurin, and why calcineurin
is spared from activation in response to the calcium that drives
muscle contraction, remain important unanswered questions.
In lymphocytes, influx of extracellular calcium through calcium
release-activated calcium channels provides the sustained cal-
cium signal required to activate the calcineurin/NFAT pathway
(9). Unlike voltage-gated L-type calcium channels, calcium
entry via calcium release-activated calcium channels is trig-
gered by the release of calcium from intracellular stores, a
mechanism referred to as store-operated calcium or capacita-
tive calcium entry. Recent work has confirmed that hyper-
trophic agonists stimulate capacitative calcium entry in cardiac
myocytes, leading to activation of the calcineurin/NFAT path-
way and cardiac hypertrophy (10, 11). However, the identity of
* The costs of publication of this article were defrayed in part by the payment
of page charges. This article must therefore be hereby marked advertise-
ment in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
1
To whom correspondence may be addressed. Tel.: 303-464-5234; Fax: 303-
410-6669; E-mail: erik.bush@myogen.com.
2
Supported by grants from the National Institutes of Health, The Donald W.
Reynolds Clinical Cardiovascular Research Center, The Robert A. Welch
Foundation, and the Texas Advanced Technology Program.
3
To whom correspondence may be addressed. Tel.: 303-533-1736; Fax: 303-
410-6669; E-mail: timothy.mckinsey@myogen.com.
4
The abbreviations used are: NFAT, nuclear factor of activated T-cells; MCIP1,
modulatory calcineurin-interacting protein 1; TRPC, canonical transient
receptor potential channels; PAMH, pyridine activator of myocyte hyper-
trophy; BTP2, N-{4-[3,5-bis(trifluoromethyl)-1H-pyrazol-1-yl]phenyl}-4-
methyl-1,2,3-thiadiazole-5-carboxamide; LV, left ventricle; OAG, oleoyl-2-
acetyl-sn-glycerol; 2APB, 2-aminoethoxydiphenylborane; GFP, green
fluorescent protein; NRVM, neonatal rat ventricular myocyte; DMEM, Dul-
becco’s modified Eagle’s medium; SHHF, spontaneous hypertensive heart
failure; TAB, thoracic aortic banding; PBS, phosphate-buffered saline; RT,
reverse transcriptase; ANF, atrial natriuretic factor.
THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 281, NO. 44, pp. 33487–33496, November 3, 2006
© 2006 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.
NOVEMBER 3, 2006 VOLUME 281 NUMBER 44 JOURNAL OF BIOLOGICAL CHEMISTRY 33487
by guest on January 13, 2016http://www.jbc.org/Downloaded from
the cardiac channel(s) responsible for capacitative calcium
entry remains unknown.
Members of the canonical transient receptor potential
(TRPC) family of channels have been implicated in the control
of capacitative calcium entry in non-cardiac cell types (12). The
seven members of the TRPC family (TRPC1–7) are nonselec-
tive calcium-permeable cation channels. Most family members
can be activated directly by diacylglycerol, a second messenger
product of phospholipase C. Furthermore, TRPC channels
have recently been shown to function as stretch receptors,
admitting calcium influx in response to mechanical stress (13,
14), a pathological stimulus known to contribute to the devel-
opment of cardiac hypertrophy (15). Other signaling molecules
known to regulate TRPC channel activity include the receptor
tyrosine kinase Src (16) and the cGMP-dependent protein
kinase PKG (17).
Previously, we reported results of microarray studies with
RNA from cultured cardiac myocytes exposed to distinct
hypertrophic stimuli (18). Among the genes most potently up-
regulated by two distinct hypertrophic agonists, the
-adrener-
gic receptor agonist, phenylephrine (PE), and the serotonin
receptor agonist, pyridine activator of myocyte hypertrophy
(PAMH), was that encoding TRPC3. Here we demonstrate that
cardiacTRPC3expressionisup-regulatedthroughacalcineurin-
dependent mechanism by discrete pharmacologic, genetic, and
physiologic stimuli that trigger pathological cardiac hypertro-
phy in rodent models. In contrast, TRPC5 is selectively induced
in failing human heart. Gain and loss of function studies dem-
onstrate that TRPC channels positively regulate cardiac hyper-
trophy through activation of calcineurin/NFAT signaling.
These results suggest a role for TRPC channels in the transmis-
sion of pathological calcium signals in the heart.
EXPERIMENTAL PROCEDURES
Chemical Reagents, Plasmids, and Adenoviral Constructs—PE,
endothelin-1, oleoyl-2-acetyl-sn-glycerol (OAG), and 2-aminoe-
thoxydiphenylborane (2-APB) were obtained from Sigma. N-{4-
[3,5-Bis(trifluoromethyl)-1H-pyrazol-1-yl]phenyl}-4-methyl-1,2,3-
thiadiazole-5-carboxamide (BTP2) was obtained from Calbiochem.
The cDNA encoding full-length, human TRPC3 (accession number
NM_003305) was cloned by reverse transcriptase-PCR with the One
Step RT-PCR kit (Invitrogen). For adenovirus production, this cDNA
was subcloned into pShuttle-cytomegalovirus (QBiogen) and adeno-
virus was generated in HEK293 cells according to the manufacturer’s
recommendations. Clonal populations of virus were obtained using
the agar overlay method and titered with the Adeno-X Rapid
Titer Kit (Clontech). The adenovirus encoding GFP-NFATc
(NFAT2) was provided by Dr. Martin Schneider (University of
Maryland, Baltimore, MD).
Cardiomyocyte Culture—Neonatal rat ventricular myocytes
(NRVMs) were prepared from 1–2-day-old Sprague-Dawley
rats as described previously (19). Cells were plated on gelatin-
coated plates in DMEM containing fetal bovine serum (10%),
penicillin-streptomycin, and
L-glutamine (DMEM complete).
After 12 h, plating medium was replaced with serum-free
DMEM supplemented with Nutridoma-SP (0.1%; Roche
Applied Sciences), which contains albumin, insulin, transferrin,
and other defined organic and inorganic compounds the fol-
lowing day. Cells were treated with agonists/inhibitors for
48–72 h.
Animal Models and Human Heart Samples—The Institu-
tional Animal Care and Use Committee approved all animal
protocols. The spontaneous hypertensive heart failure (SHHF)
rat is a well documented genetic model of hypertension, cardiac
hypertrophy, and heart failure (20). For the isoproterenol
model, male Sprague-Dawley rats were implanted subcutane-
ously with Alzet Mini-Pumps filled with either vehicle control
(0.5 mg/ml sodium bisulfite in 0.9% sterile NaCl) or isoproter-
enol (dissolved in vehicle sufficient to administer 5 mg/kg/day
of isoproterenol for 4 days). For thoracic aortic banding (TAB),
male Sprague-Dawley rats (8 –9 weeks of age, 200 –225 g) were
anesthetized with 5% isoflurane (v/v 100% O
2
), intubated, and
maintained at 2.0% isoflurane with positive pressure ventila-
tion. A left thoracotomy through the third intercostal space was
performed and the descending thoracic aorta 3–4 mm cranial
to the intersection of the aorta and azygous vein was isolated. A
segment of 5-0 silk suture was then positioned around the iso-
lated aorta to function as a ligature. A blunted hypodermic nee-
dle (gauge determined by weight) was placed between the aorta
and the suture to prevent complete aortic occlusion when the
suture was tied. When tying was completed, the needle was
removed from between the aorta and ligature, re-establishing
flow through the vessel. The thorax was then closed and the
pneumothorax evacuated. After 7 days of recovery, animals
were sacrificed and left ventricular tissue processed for West-
ern blot analysis. Average heart weight to body weight ratios in
banded versus sham-operated rats increased 22% at 1 week. The
construction and characterization of transgenic mice express-
ing constitutively active calcineurin have been described else-
where (3).
Ventricular samples from human patients with end-stage idi-
opathic dilated cardiomyopathy or nonfailing human cardiac
tissue samples were obtained through the University of Colo-
rado Health Sciences Center Cardiac Transplantation Pro-
gram. End-stage failing hearts came from individuals who
underwent heart transplantation because of idiopathic dilated
cardiomyopathy (n 6; 3 male, 3 female, average age 49.8
years). Nonfailing controls were obtained from organ donors
whose hearts were unsuitable for donation due to blood type or
size incompatibilities (n 6; 4 male, 2 female; average age 51.8
years). Tissue samples were taken immediately upon explanta-
tion and rapidly frozen in liquid nitrogen.
Immunoblotting—NRVMs and in vivo tissue samples were
homogenized in Tris buffer (50 m
M, pH 7.5) containing EDTA
(5 m
M), Triton X-100 (1%), protease inhibitor mixture (Com-
plete, Roche), phenylmethylsulfonyl fluoride (1 m
M), and phos-
phatase inhibitors (sodium pyrophosphate, 1 m
M; sodium flu-
oride, 2 m
M;
-glycerolphosphate, 10 mM; sodium molybdate, 1
m
M; and sodium orthovanadate, 1 mM). Homogenates were
briefly sonicated and cleared by centrifugation. Protein concen-
trations were determined by BCA assay (Pierce) and 15
gof
total protein was resolved by SDS-PAGE with gradient gels
(4–20% polyacrylamide; Invitrogen). Proteins were transferred
to nitrocellulose membranes (Bio-Rad) and probed with
TRPC3- or TRPC5-specific antibodies, according to the manu-
facturer’s instructions (Alomone Labs). Experiments were per-
TRPC Channels Promote Cardiomyocyte Hypertrophy
33488 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 NUMBER 44 NOVEMBER 3, 2006
by guest on January 13, 2016http://www.jbc.org/Downloaded from
formed with freshly diluted antibodies, because in our experi-
ence we have found that the lyophilized antibodies, once
resuspended, lose significant immunoreactivity in storage.
Negative controls for specificity were performed by preincubat-
ing the antibody with the corresponding antigen peptide (0.75
g of peptide/1
g of antibody) for 1 h. Antibodies against
MCIP1 and calnexin have been described previously (18). Pro-
teins were visualized using an enhanced chemiluminescence
system (Pierce). For immunoprecipitation experiments, 3
gof
antibody was incubated with homogenate in a total volume of
0.5 ml for 1 h. Protein G-Sepharose beads (10
l, GE Health-
care) were added, and after a 1-h incubation, beads were
washed three times in homogenization buffer and prepared for
SDS-PAGE.
Hypertrophy Measurements—The atrial natriuretic factor
(ANF) competition enzyme-linked immunosorbent assay,
measurement of
-myosin heavy chain expression, RNA dot
blot assay for hypertrophic marker gene expression, and adeny-
late kinase release assay have been described previously (19).
For cell volume measurements, NRVM cultured in 6-well
dishes were harvested by treatment with trypsin (Cellgro).
After recovery by centrifugation, cell pellets were washed in
PBS, resuspended in 10 ml of IsoFlow electrolyte solution
(Beckman-Coulter), and analyzed with a Z2 Coulter Particle
Counter and Size Analyzer (Beckman-Coulter).
RT-PCR Analysis—RNA was isolated from NRVMs using
TRI Reagent (Sigma) according to the manufacturer’s instruc-
tions. RNA was quantified using both standard spectrophotom-
etry of nucleic acids at 260 nm using a DU64 spectrophotome-
ter (Beckman-Coulter) and fluorescence with a TD700
fluorometer (Turner Designs). RT-PCR of rat TRPC3 (acces-
sion number NM_021771) used primers that spanned the first
intron (7950 bp): forward primer, 5-ctggccaacatagagaaggagt-
3; and reverse primer, 5-caccgattccagatctccat-3. Rat 18 S
ribosomal RNA amplicons were used as input controls: forward
primer, 5-cgaggaattcccagtaagtgc-3, and reverse primer,
5-aagttcgaccgtcttctcagc-3. Reverse transcription and amplifi-
cation of 0.5
g of total RNA were done using the Super-
Script
TM
One-step RT-PCR System with Platinum Taq DNA
Polymerase (Invitrogen), on a I-cycler
TM
thermocyler (Bio-
Rad). Reverse transcription reactions were run on 2% agarose
gels containing 0.1
g/ml ethidium bromide. Imaging and den-
sitometry were performed with an Alpha Innotech gel imaging
system and ChemImage 5.5 software. RT-PCR amplified a sin-
FIGURE 1. Hypertrophic agonists stimulate TRPC3 expression in cultured cardiac myocytes. A, NRVMs (2 10
6
) were cultured in serum-free medium and
exposed to endothelin-1 (ET-1;50n
M), PE (20
M), or fetal bovine serum (FBS; 10%) for 72 h, as indicated. RT-PCR analysis was performed with primers specific
for TRPC3 or 18 S ribosomal RNA. B, TRPC3 mRNA levels were quantified by densitometry of ethidium bromide-stained gels and normalized to 18 S RNA to
control for differences in RNA input/loading. Values represent averages S.D. from three independent sets of cells per condition. C, NRVMs were treated with
PE (20
M) in the absence or presence of cyclosporine A (CsA; 500 nM) or FK506 (1 nM). TRPC3 mRNA expression was analyzed as described in B. Values represent
averages S.D. from three independent sets of cells per condition. D, total protein from NRVMs, rat left ventricle and mouse left ventricle was prepared and
analyzed by immunoblotting with a TRPC3-specific antibody. As a specificity control, the TRPC3 antibody was preincubated with the corresponding antigen
peptide. The immunoblot was exposed to film for 5 (left panel) and 30 s (right panel). E, NRVMs were treated with PE (20
M) as described in A. Total protein was
prepared and analyzed by immunoblotting with a TRPC3-specific antibody. Blots were reprobed with anti-calnexin antibodies to control for protein loading.
F, TRPC3 protein levels from E were quantified by densitometry. Values represent averages S.D. from three independent sets of cells per condition. *, p 0.01.
TRPC Channels Promote Cardiomyocyte Hypertrophy
NOVEMBER 3, 2006 VOLUME 281 NUMBER 44 JOURNAL OF BIOLOGICAL CHEMISTRY 33489
by guest on January 13, 2016http://www.jbc.org/Downloaded from
gle 141-bp band of the expected size, which sequencing
confirmed to be TRPC3. Negative control reactions excluding
RNA template, reverse transcriptase, or either primer did not
yield a PCR product. For analysis of human samples, RNA
was extracted from left ventricular tissue as described above
and subjected to TaqMan real-time quantitative RT-PCR
(PerkinElmer Life Sciences) as described by Riccio et al. (21).
Human TRPC5 primers were: forward, 5-cctctcatcagaaccatgc-
caa-3, and reverse, 5-gcgtttgcttgatgactcagc-3.
Indirect Immunofluorescence—NRVMs were plated on gela-
tin-coated 6-well dishes (2.5 10
5
cells/well) in DMEM com
-
plete and transferred 12 h subsequently to serum-free DMEM
supplemented with Nutridoma-SP (0.1%; Roche Applied Sci-
ences). Adenovirus was added to cells at the time of plating.
Unless otherwise indicated, virus was used at a multiplicity of
infection of 20. Following experimental treatment, cells were
washed two times in PBS, fixed with formalin (10%) in PBS,
permeabilized, and blocked with PBS containing Nonidet P-40
(0.1%) and bovine serum albumin (3%), and then incubated in
the same solution containing primary antibodies specific for
either sarcomeric
-actinin (Sigma; 1:200 dilution) or Myc
(Santa Cruz; 1:500 dilution) for1hatroom temperature. Cells
were washed 4 times in PBS and incubated in PBS/Nonidet
P-40/bovine serum albumin containing either a fluorescein- or
CY3-conjugated secondary antibody (Jackson Laboratories;
1:200 dilution) for 30 min at room temperature. Cells were
washed 4 times in PBS and then covered with mounting solu-
tion (SlowFade, Molecular Probes) and glass coverslips. Pro-
teins were visualized with an inverted fluorescence micro-
scope (Olympus model BH-2) at 40 magnification, and
images were captured using a digital camera (Photometrics;
Roper Scientific).
RESULTS
Calcineurin-dependent Induction of TRPC3 Expression in
Cultured Cardiac Myocytes—To gain insight into changes in
gene expression underlying stress-induced cardiac hypertro-
phy, RNA microarray studies were performed with cultured
NRVMs. NRVMs undergo hypertrophy in response to multiple
agonists, including PE, which stimulates the
1
-adrenergic
receptor, and PAMH, a novel 5-HT2B serotonin receptor ago-
nist. A complete summary of microarray results with RNA from
NRVMs stimulated with PE or PAMH for 48 h is found in
the NCBI Gene Expression Omnibus (GEO) and are accessible
through GEO Series accession number GSE925. Among the
genes most potently up-regulated by both agonists were those
encoding
-myosin heavy chain, a classical marker of cardiac
hypertrophy, and the transient receptor potential channel 3
(TRPC3) calcium channel.
Although calcium is known to play a key role in the regula-
tion of cardiac hypertrophy (2), the relative contribution of dis-
tinct calcium pools to this growth response remains unclear. As
such, we focus here on elucidating the potential role of TRPC
channels in the control of stress-induced cardiac hypertrophy.
To confirm the microarray results, semi-quantitative
RT-PCR was performed with RNA from freshly isolated
NRVMs treated with PE or two independent hypertrophic ago-
nists, endothelin-1 and fetal bovine serum, for 48 h. Each ago-
nist stimulated expression of TRPC3 mRNA transcripts, with
PE and fetal bovine serum inducing the highest levels (Fig. 1, A
and B). Similar results were obtained with RNA from PAMH-
treated cells (data not shown).
Prior studies have suggested that signaling by the calcineurin
phosphatase stimulates expression of TRPC3 in skeletal muscle
FIGURE 2. Induction of cardiac TRPC3 protein expression in vivo. A, male,
2-month-old Sprague-Dawley (SD) rats were given a sham operation or sub-
jected to thoracic aortic banding to trigger pressure overload hypertrophy of
the LV of the heart, as described under “Experimental Procedures.” Seven
days following the operation, animals were sacrificed and TRPC3 levels in LV
protein lysates analyzed by immunoblotting. Blots were reprobed with cal-
nexin-specific antibodies to control for protein loading. B, parallel samples of
LV protein lysates were run to assess MCIP1 protein levels by immunoblot. C,
male SHHF rats of the indicated ages were sacrificed and TRPC3 levels in LV
preparations assessed as described in A. D, male SD rats were implanted with
osmotic mini-pumps that release either vehicle or the
-adrenergic receptor
agonist isoproterenol (ISO, 4 mg/kg/day) for 4 days. TRPC3 levels in LV prep-
arations were assessed as described in A. E, male mice (2 months of age)
expressing a constitutively active calcineurin transgene (Cn-Tg) and control
nontransgenic littermates (WT) were sacrificed and TRPC3 and MCIP1 levels in
LV preparations assessed as described in A. F, TRPC3 protein levels in the
indicated models were quantified by densitometry. Values represent aver-
ages S.D. *, p 0.05.
TRPC Channels Promote Cardiomyocyte Hypertrophy
33490 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 NUMBER 44 NOVEMBER 3, 2006
by guest on January 13, 2016http://www.jbc.org/Downloaded from
(22). To determine the role of cal-
cineurin in TRPC3 gene regulation
in cardiac myocytes, RT-PCR was
performed with RNA from NRVMs
stimulated with PE in the absence or
presence of the calcineurin inhibi-
tors cyclosporine A or FK506. As
shown in Fig. 1C, agonist-depend-
ent induction of TRPC3 mRNA
transcripts was completely blocked
by either of the two calcineurin
inhibitors.
To evaluate expression of TRPC3
protein expression in NRVMs and
rat and mouse ventricles, we per-
formed immunoblot analysis with a
TRPC3-specific antibody. As a con-
trol for specificity, the TRPC3 anti-
body was preincubated with the
antigen peptide. The antibody
detected immunoreactive bands of
the expected molecular mass
(110–120 kDa) (Fig. 1D, left
panel). The most abundant band
observed in NRVM was 115 kDa,
with a slightly smaller band of 110
kDa predominating in rat and
mouse ventricular samples. Nota-
bly, detection of these immunoreac-
tive bands was efficiently competed
by preincubation of the TRPC3
antibody with the antigen peptide.
Intentional overexposure of the
same immunoblot (Fig. 1D, right
panel) revealed a number of non-
specific bands that were not com-
peted in the antigen peptide-blocked
controls. Immunoblot analysis veri-
fied that the observed induction of
TRPC3 mRNA correlated with mod-
est (between 1.5- and 2-fold) but sig-
nificant increases in TRPC3 protein
levels in PE-treated NRVMs (Fig. 1, E
and F).
Induction of TRPC3 Expression
in Rodent Models of Cardiac Hy-
pertrophy—Experiments were per-
formed to determine whether
TRPC3 expression is elevated in
animal models of pathological car-
diac hypertrophy. TAB places a
pressure overload on the heart and
triggers left ventricular (LV) hy-
pertrophy. Immunoblotting studies
revealed that TRPC3 protein ex-
pression was consistently elevated in
LVs of rats subjected to TAB for 7
days (Fig. 2A).
FIGURE 3. Ectopic TRPC3 stimulates cardiomyocyte hypertrophy. A, NRVMs (2 10
6
) were cultured in
10-cm dishes, and infected with Myc-tagged TRPC3 adenovirus (Ad-TRPC3) for 48 h. Total protein was prepared
and analyzed by immunoblotting with antibodies to TRPC3 (left panel, two independent clones shown) and the
Myc epitope tag (right panel). The apparent molecular mass of endogenous and recombinant TRPC3 was 115
kDa. B, samples prepared as in panel A were immunoprecipitated with either the anti-Myc antibody or the
anti-TRPC3 antibody and immunoblotted with the other antibody, confirming that both antibodies immuno-
precipitated recombinant TRPC3. C, NRVMs were cultured in 6-well dishes (2.5 10
5
/well) and infected with
adenoviruses encoding
-galactosidase (Ad-
-GAL) or Myc-tagged TRPC3 (Ad-TRPC3). Cells were cultured for
48 h in serum-free medium in the absence or presence of the TRPC agonist OAG (100
M). Cells were fixed and
sarcomeres were visualized by indirect immunofluorescence with anti-sarcomeric actinin antibodies (top row).
Higher magnification images of the same cells are shown (middle row). Cells were co-stained with anti-Myc
antibody to reveal the subcellular distribution of TRPC3 (bottom row). D, NRVMs prepared as in panel B were
harvested by trypsinization, and mean cell volumes were determined by Coulter analysis. Values represent aver-
ages S.D. for four independent sets of cells per condition. *, p 0.01 versus uninfected control; #, p 0.05
versus Ad-TRPC3 alone. E, NRVMs (2 10
6
) were cultured in 10-cm dishes, infected with either Ad-
-galactosidase or
Ad-TRPC3, and maintained in serum-free medium for 72 h. Total RNA was prepared and atrial natriuretic factor (ANF)
and
-skeletal actin (
-SK-actin) mRNA abundance determined by dot blot analysis with radiolabeled probes. Values
represent averages S.D. for three independent sets of cells per condition. *, p 0.05.
TRPC Channels Promote Cardiomyocyte Hypertrophy
NOVEMBER 3, 2006 VOLUME 281 NUMBER 44 JOURNAL OF BIOLOGICAL CHEMISTRY 33491
by guest on January 13, 2016http://www.jbc.org/Downloaded from
To determine whether induction of TRPC3 expression cor-
relates with stimulation of calcineurin signaling, we quantified
expression of the endogenous NFAT target gene, modulatory
calcineurin-interacting protein-1 (MCIP1). The MCIP1 pro-
tein is a negative-feedback regulator of calcineurin signaling
(8), and is expressed as 28- and 38-kDa isoforms (18). Expres-
sion of 28-kDa MCIP1 is regulated by an alternative promoter
that harbors 15 NFAT binding sites, and thus this lower molec-
ular weight form of the protein serves as a sensitive marker of
NFAT activity (23). We observed significantly increased
expression of 28-kDa MCIP1 in TAB ventricles, indicating
enhanced calcineurin/NFAT activation (Fig. 2B). These obser-
vations are consistent with a role of calcineurin in the control of
TRPC3 gene expression.
SHHF rats provide a genetic
model of pathological cardiac
remodeling and heart failure (20).
Due to a mutation in the leptin
receptor gene, SHHF rats develop
hypertension that results in cardiac
hypertrophy and eventual heart fail-
ure. As shown in Fig. 2C, TRPC3
protein levels were elevated in LVs
from failing hearts of 20-month-
old SHHF rats. However, TRPC3
expression was normal in 8–9-
month-old rats with compensated
LV hypertrophy. TRPC3 protein
expression was also increased in
LVs of rats infused for 4 days with
the
-adrenergic receptor agonist
isoproterenol, which stimulates car-
diac hypertrophy that is accompa-
nied by fibrosis (Fig. 2D). Similar to
the TAB model, increased expres-
sion of MCIP1 protein was observed
in LV samples from both ISO-in-
fused and 20-month-old SHHF
rats (data not shown).
To address the role of calcineurin
signaling in the control of cardiac
TRPC3 expression, TRPC3 protein
levels were measured in LVs of
transgenic mice expressing consti-
tutively active calcineurin in the
heart under the control of the
-my-
osin heavy chain promoter (Cn-Tg)
(3). As shown in Fig. 2E, TRPC3
expression was dramatically ele-
vated in Cn-Tg mice compared with
wild-type littermates. Levels of
TRPC3 protein in LV samples from
each model of cardiac hypertrophy
were quantified by densitometry
(Fig. 2F). Together, the results dem-
onstrate that TRPC3 protein
expression is induced by diverse
cues for pathological cardiac growth
in vivo, and suggest a role for calcineurin signaling in the induc-
tion of cardiac TRPC3 expression.
Regulation of Cardiomyocyte Hypertrophy by TRPC Chan-
nels—Cardiac hypertrophy is associated with enhanced protein
synthesis, cell growth in the absence of mitosis, and increased
sarcomere assembly. In addition, fetal cardiac genes, including
those that encode ANF and
-skeletal actin, and
-myosin
heavy chain become reactivated during the hypertrophic
response. To begin to address the role of TRPC channels in the
regulation of cardiac hypertrophy, we developed an adenovirus
that encodes full-length Myc epitope-tagged TRPC3 (Ad-
TRPC3) (Fig. 3A). As an additional control for antibody selec-
tivity, we determined that both anti-Myc and anti-TRPC3 anti-
bodies effectively immunoprecipitated recombinant TRPC3
FIGURE 4. Inhibition of hypertrophic gene expression by 2-APB and BTP2. NRVMs were cultured in 96-well
dishes and exposed to increasing doses of two different TRPC channel inhibitors, 2-APB or BTP2, in the absence
or presence of PE (20
M). After 48 h of stimulation, ANF abundance in culture supernatants, (A and B) cellular
-myosin heavy chain expression (C and D), and adenylate kinase release (E and F) (cytoxicity marker) were
measured as described under “Experimental Procedures.”
TRPC Channels Promote Cardiomyocyte Hypertrophy
33492 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 NUMBER 44 NOVEMBER 3, 2006
by guest on January 13, 2016http://www.jbc.org/Downloaded from
(Fig. 3B). NRVMs were infected with Ad-TRPC3 or control
virus expressing
-galactosidase (Ad-
-galactosidase) and cul-
tured for 48 h in the absence or presence of OAG, a diacylglyc-
erol analog that binds to and stimulates TRPC channel activity
(24, 25). As shown in Fig. 3C, Ad-TRPC3 overexpression led to
modest increases in NRVM sarcomere organization compared
with control cells infected with Ad-
-galactosidase. The posi-
tive effect of TRPC3 overexpression on sarcomere assembly
was dramatically enhanced by stimulation of cells with the
TRPC channel agonist, OAG. Enhanced sarcomere assembly
correlated with expression of ectopic TRPC3, which localized
to the perinuclear and punctuate pan-cellular regions of the
NRVMs. The perinuclear localization of TRPC3 likely repre-
sents TRPC3 transiting through the Golgi network, because
indirect immunofluorescence revealed co-localization of this
pool of TRPC3 with p155 golgin, a Golgi marker (data not
shown).
We observed that ectopic TRPC3 expression significantly
increased mean cell volume (Fig. 3D); the presence of OAG
produced a further increase. As shown in Fig. 3E, TRPC3
induced expression of ANF and
-skeletal actin mRNA tran-
scripts. Of note, OAG did not appear to enhance TRPC3-me-
diated induction of these genes.
Thus, basal TRPC3 activity appears
to be sufficient to increase cell vol-
ume and induce fetal cardiac genes,
whereas full sarcomere altering
effects of TRPC3 require activation
by OAG. Together, the results sug-
gest that ectopic TRPC3 is capable
of stimulating hypertrophy of cul-
tured cardiac myocytes.
As an alternative approach to
study the role of TRPC channels in
the control of cardiac hypertrophy,
we employed two distinct small
molecule inhibitors of calcium
release-activated calcium channels,
2-APB (25) and the pyrazole deriva-
tive BTP2 (28). Whereas these
inhibitors block TRPC3-mediated
calcium influx, we cannot exclude
the possibility that 2-APB and BTP2
may also be acting on other chan-
nels including TRPVs and TRPMs.
As shown in Fig. 4, AD, both
2-APB and BTP2 exhibited dose-
dependent inhibition of ANF secre-
tion and
-myosin heavy chain
expression in NRVMs stimulated
with PE. Only cells treated with the
highest concentrations of 2-APB
and BTP2 showed evidence of cyto-
toxicity, as determined by assessing
the presence of adenylate kinase in
culture supernatants (Fig. 4, E and
F). Treatment with 2-APB also
resulted in suppression of PE-medi-
ated increases in NRVM size and protein synthesis (data not
shown). Together, the data from these gain and loss of function
experiments suggest a role for TRPC channels in the control of
cardiac hypertrophy.
TRPC3 Activates Calcineurin/NFAT Signaling—The cal-
cineurin phosphatase plays an essential role in the regulation of
cardiac hypertrophy, and has recently been shown to be regu-
lated by TRPC3 in skeletal muscle. As such, we hypothesized
that the mechanism whereby TRPC channels stimulate cardiac
hypertrophy involves calcineurin. As an initial step to address
this hypothesis, we examined the effect of TRPC3 overexpres-
sion on the subcellular distribution of the calcineurin target,
NFAT. The NFAT transcription factor is a phosphoprotein that
resides in the cytosol and, upon dephosphorylation by cal-
cineurin, translocates to the nucleus. NFAT was cytosolic in
unstimulated NRVMs, and infection of cells with Ad-
-galac-
tosidase did not alter its subcellular distribution. In contrast,
TRPC3 overexpression partially stimulated nuclear import of
NFAT, and this effect was enhanced by treatment of the cells
with OAG (Fig. 5A). Of note, we observed modest induction of
NFAT nuclear import in Ad-
-galactosidase-infected cells
treated with OAG.
FIGURE 5. TRPC channels stimulate cardiac calcineurin/NFAT signaling. A, NRVMs were infected with
adenoviruses encoding GFP-tagged NFAT and either Ad-
-galactosidase (GAL) or Ad-TRPC3. Cells were cul-
tured in serum-free medium and exposed to OAG (100
M) for 12 h, as indicated. Cells were fixed and GFP-NFAT
localization assessed by fluorescence microscopy (top panels). TRPC3 was detected in the same cells by indirect
immunofluorescence with anti-Myc antibodies (bottom panels). B, NRVMs were treated as described in A. After 12 h
of OAG treatment, protein lysates were prepared and NFAT phosphorylation status assessed by immunoblotting
with GFP-specific antibodies (upper panel). The blot was reprobed with anti-Myc antibody to reveal ectopic TRPC3
(lower panel). C, NRVMs were left uninfected or infected with increasing amounts of Ad-TRPC3 (multiplicity of infec-
tion 1, 5, and 25). Cells were harvested after 48 h of culture and MCIP1 expression analyzed by immunoblotting
(upper panel). Phosphorylated NFAT is indicated by circled P. The blot was reprobed to assess ectopic TRPC3 levels
(lower panel). D, NRVMs were cultured in serum-free medium and exposed to PE (20
M) in the absence or presence
of 2-APB (30
M). Following 48 h in culture, protein lysates were prepared and MCIP1 levels determined by immu-
noblotting. The 28-kDa isoform of MCIP1 is calcineurin responsive.
TRPC Channels Promote Cardiomyocyte Hypertrophy
NOVEMBER 3, 2006 VOLUME 281 NUMBER 44 JOURNAL OF BIOLOGICAL CHEMISTRY 33493
by guest on January 13, 2016http://www.jbc.org/Downloaded from
Immunoblotting studies were performed to confirm that
TRPC3 stimulates calcineurin-dependent dephosphorylation
of NFAT. Hypophosphorylated NFAT migrates more rapidly
than hyperphosphorylated NFAT in denaturing polyacrylam-
ide gels. As shown in Fig. 5B, overexpression of TRPC3 in
NRVMs triggered dephosphorylation of NFAT, as evidenced by
the appearance of the rapidly migrating NFAT species. Consist-
ent with the results presented in Fig. 5A, OAG enhanced
TRPC3-mediated dephosphorylation of NFAT.
To determine whether TRPC channels regulate endogenous
NFAT target genes, we monitored expression of the 28-kDa
isoform of MCIP1, which is dependent on calcineurin/NFAT
signaling. As shown in Fig. 5C, exposure of NRVMs to increas-
ing doses of Ad-TRPC3 led to selective up-regulation of 28-kDa
MCIP1,indicatingthatTRPC3overexpressionstimulatesNFAT-
dependent transcription. In contrast, 2-APB blocked PE-medi-
ated induction of 28-kDa MCIP1 (Fig. 5D). Together, the
results demonstrate that TRPC channels stimulate cardiac cal-
cineurin/NFAT signaling.
Induction of TRPC5 Expression in Failing Human Heart—To
begin to address whether TRPC channels play a role in the
pathogenesis of human heart failure, quantitative RT-PCR was
employed to survey expression of TRPC1, -3, -4, -5, and -6 in
nonfailing human heart versus hearts of patients with idio-
pathic dilated cardiomyopathy. TRPC5 mRNA expression was
significantly elevated in failing human heart relative to nonfail-
ing controls (Fig. 6A). Immunoblot analysis confirmed that
increased TRPC5 mRNA levels in failing heart correlated with
enhanced TRPC5 protein expression (Fig. 6B). We observed
two immunoreactive bands in the human heart; both were
effectively competed by preincubating the TRPC5 antibody
with antigen peptide. The lower band, of 110 kDa, is in agree-
ment with the predicted molecular mass of TRPC5. Expression
of a larger specific band of 180 kDa was also elevated in failing
human hearts. Consistent with previous observations (21),
TRPC3 mRNA transcripts and protein were undetectable in
nonfailing or failing human heart (Fig. 6A, and data not shown).
Levels of TRPC1, -4, and -6 were unchanged in normal versus
failing heart (data not shown).
DISCUSSION
The current study suggests that TRPC channels contribute to
the maintenance of cardiac hypertrophy via a positive feedback
mechanism involving activation of calcineurin/NFAT signaling
(Fig. 7). Pathological stress stimuli cause phospholipase C acti-
vation and store depletion, which potentiate TRPC activity and
FIGURE 6. Induction of TRPC5 protein expression in failing human heart.
A, total RNA was prepared from left ventricles of end-stage failing heart from
individuals with idiopathic dilated cardiomyopathy (n 6) and nonfailing
controls (n 6). Levels of mRNA transcripts for individual TRPC isoforms were
determined by quantitative RT-PCR. Values depict averages of the ratio of
TRPC mRNA to GAPDH mRNA (ng/ng), S.D., p 0.05. B, protein lysates were
prepared from the same LV samples and TRPC5 levels determined by immu-
noblotting (top panels). Specificity of the TRPC5 antibody was confirmed by
preincubating the antibody with the corresponding antigen peptide (middle
panels). Blots were reprobed with calnexin-specific antibody to control for
protein loading (bottom panels).
FIGURE 7. A model for regulation of cardiac hypertrophy by TRPC chan-
nels. Stress signals, such as those emanating from G protein-coupled recep-
tors (GPCRs), trigger phospholipase C (PLC) activation, with subsequent for-
mation of diacylglycerol (DAG) and inositol trisphosphate (IP3). Inositol
trisphosphate promotes release of calcium (Ca
2
) stores from internal stores,
which potentiates TRPC channel activity and produces a calcium signal suffi-
cient to activate calcineurin/NFAT. TRPC channels can also be activated by
direct diacylglycerol binding. Among the genes modulated by the cal-
cineurin/NFAT pathway are TRPCs themselves, suggesting the existence of a
positive-feedback circuit that stabilizes a state of hypertrophic gene expres-
sion. NFAT regulates cardiac gene expression through association with other
transcription factors, including myocyte enhancer factor-2 (MEF2) and GATA.
TRPC Channels Promote Cardiomyocyte Hypertrophy
33494 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 NUMBER 44 NOVEMBER 3, 2006
by guest on January 13, 2016http://www.jbc.org/Downloaded from
produce a calcium signal sufficient to activate calcineurin/
NFAT. Because TRPC channels are mechanosensitive, it is also
possible that abnormal mechanical stress in the hypertrophied
and failing heart might activate TRPC channels directly.
Among the genes modulated by the calcineurin/NFAT path-
way are TRPCs themselves, completing a positive feedback
circuit that stabilizes a state of hypertrophic gene expression.
Calcineurin signaling may also stabilize TRPC3 protein post-
translationally. The interplay between calcineurin and TRPC
channels may contribute to the selective activation of cardiac
calcineurin by calcium in diseased myocardium.
A similar regulatory mechanism has been hypothesized to
operate in skeletal muscle, where calcineurin/NFAT-depend-
ent up-regulation of TRPC3 provides positive feedback rein-
forcing the slow oxidative muscle fiber phenotype (22). Inter-
play between TRPC channels and calcineurin was also recently
described in a prostate cancer cell line (26). In addition, inde-
pendent pharmacologic evidence of the central role TRPC
channels play in the regulation of calcineurin/NFAT signaling
has been provided by recent small molecule screens for NFAT
inhibitors. A high-throughput screen for compounds capable
of suppressing activation of the NFAT-responsive interleu-
kin-2 promoter (27) identified a series of 3,5-bistrifluoromethyl
pyrazole store-operated calcium inhibitors (including BTP2)
(28), which have subsequently been shown to be TRPC antag-
onists (29). An alternative high-throughput screen for com-
pounds that block nuclear translocation of NFAT also pro-
duced a series of store-operated calcium inhibitors (30).
Aberrant TRPC expression or activity is thought to contrib-
ute to the pathogenesis of a number of human diseases (31).
Recent studies indicate that TRPC channels mediate the abnor-
mal calcium influx and overload observed in skeletal myotubes
from Duchenne muscular dystrophy patients (32). The patho-
logic hyperplasia of pulmonary arterial smooth muscle cells
that occurs in idiopathic pulmonary hypertension is driven, in
part, by increased calcium influx through TRPC channels (33).
Bosentan, a non-selective endothelin receptor antagonist used
for the treatment of idiopathic pulmonary arterial hyperten-
sion, may exert antiproliferative effects via down-regulation of
TRPC6 (34).
We observed that TRPC expression is up-regulated in animal
models of cardiac hypertrophy and human heart failure, and
that pharmacologic inhibition of channel activity suppressed
hypertrophy in vitro. However, the relative contribution of this
channel family to the abnormal calcium handling seen in the
hypertrophied and failing human heart remains to be further
elucidated. Intriguingly, down-regulation of cardiac SERCA2 in
vitro (a hallmark of hypertrophy in vivo) is associated with coor-
dinate up-regulation of TRPC channels and calcineurin activa-
tion, suggesting a link between the regulation of calcium
reuptake and extracellular calcium influx (35). Because TRPC
family members can form heteromultimers with distinct elec-
trophysiological properties, it will be particularly important to
assess the relative contributions of individual family members
to the activation of cardiac calcineurin/NFAT signaling. Our
own observations indicate that TRPC5, rather than TRPC3, is
up-regulated in the failing human heart, perhaps revealing a
crucial species difference. In addition, cardiac-specific overex-
pression of TRPC6 has been shown to induce heart failure in
mice.
5
As regulators of pro-hypertrophic calcium signaling in car-
diomyocytes, TRPC channels may represent novel pharmaco-
logic targets for modulating pathologic calcineurin/NFAT sig-
naling in the heart. Future work focused on the development of
selective inhibitors of TRPC channel activity or expression will
provide essential tools for the functional characterization of
these channels in cardiomyocytes, and may yield therapeuti-
cally useful compounds for the treatment of cardiac hypertro-
phy and heart failure.
Acknowledgments—We thank M. Schneider (University of Maryland)
for the NFAT-GFP adenovirus, S. McCune for SHHF rats, T. K. Pope
for rat TAB samples, M. A. McNiven (Mayo Clinic) for the antibody
against p155 golgin, and A. Tizenor for graphics. We are grateful to K.
Pitts and G. Lundgaard for assistance and advice with NRVMs, and
all members of Myogen R&D for support during the course of this
work.
REFERENCES
1. Levy, D., Garrison, R. J., Savage, D. D., Kannel, W. B., and Castelli, W. P.
(1990) N. Engl. J. Med. 322, 1561–1566
2. Frey, N., McKinsey, T. A., and Olson, E. N. (2000) Nat. Med. 6,
1221–1227
3. Molkentin, J. D., Lu, J. R., Antos, C. L., Markham, B., Richardson, J.,
Robbins, J., Grant, S. R., and Olson, E. N. (1998) Cell 93, 215–228
4. Frey, N., and Olson, E. N. (2003) Annu. Rev. Physiol. 65, 45–79
5. Wilkins, B. J., and Molkentin, J. D. (2004) Biochem. Biophys. Res. Commun.
322, 1178 –1191
6. Rothermel, B., Vega, R. B., Yang, J., Wu, H., Bassel-Duby, R., and Williams,
R. S. (2000) J. Biol. Chem. 275, 8719 8725
7. Rothermel, B. A., McKinsey, T. A., Vega, R. B., Nicol, R. L., Mammen, P.,
Yang, J., Antos, C. L., Shelton, J. M., Bassel-Duby, R., Olson, E. N., and
Williams, R. S. (2001) Proc. Natl. Acad. Sci. U. S. A. 98, 3328–3333
8. Rothermel, B. A., Vega, R. B., and Williams, R. S. (2003) Trends Cardio-
vasc. Med. 13, 15–21
9. Crabtree, G. R., and Olson, E. N. (2002) Cell 109, (suppl) S67–S79
10. Hunton, D. L., Lucchesi, P. A., Pang, Y., Cheng, X., Dell’Italia, L. J., and
Marchase, R. B. (2002) J. Biol. Chem. 277, 14266–14273
11. Hunton, D. L., Zou, L., Pang, Y., and Marchase, R. B. (2004) Am. J. Physiol.
286, H1124 –H1132
12. Putney, J. W. (2005) Pflugers Arch. 451, 29 –34
13. Maroto, R., Raso, A., Wood, T. G., Kurosky, A., Martinac, B., and Hamill,
O. P. (2005) Nat. Cell Biol. 7, 179–185
14. Barritt, G., and Rychkov, G. (2005) Nat. Cell Biol. 7, 105–107
15. Sadoshima, J., and Izumo, S. (1997) Annu. Rev. Physiol. 59, 551–571
16. Vazquez, G., Wedel, B. J., Kawasaki, B. T., Bird, G. S., and Putney, J. W., Jr.
(2004) J. Biol. Chem. 279, 40521– 40528
17. Kwan, H. Y., Huang, Y., and Yao, X. (2004) Proc. Natl. Acad. Sci. U. S. A.
101, 2625–2630
18. Bush, E., Fielitz, J., Melvin, L., Martinez-Arnold, M., McKinsey, T. A.,
Plichta, R., and Olson, E. N. (2004) Proc. Natl. Acad. Sci. U. S. A. 101,
2870–2875
19. Harrison, B. C., Roberts, C. R., Hood, D. B., Sweeney, M., Gould, J. M.,
Bush, E. W., and McKinsey, T. A. (2004) Mol. Cell. Biol. 24, 10636 –10649
20. Haas, G. J., McCune, S. A., Brown, D. M., and Cody, R. J. (1995) Am.
Heart J. 130, 806 811
21. Riccio, A., Medhurst, A. D., Mattei, C., Kelsell, R. E., Calver, A. R., Randall,
A. D., Benham, C. D., and Pangalos, M. N. (2002) Brain Res. Mol. Brain Res.
109, 95–104
5
E. Olson and K. Kuwahara, unpublished data.
TRPC Channels Promote Cardiomyocyte Hypertrophy
NOVEMBER 3, 2006 VOLUME 281 NUMBER 44 JOURNAL OF BIOLOGICAL CHEMISTRY 33495
by guest on January 13, 2016http://www.jbc.org/Downloaded from
22. Rosenberg, P., Hawkins, A., Stiber, J., Shelton, J. M., Hutcheson, K., Bassel-
Duby, R., Shin, D. M., Yan, Z., and Williams, R. S. (2004) Proc. Natl. Acad.
Sci. U. S. A. 101, 9387–9392
23. Yang, J., Rothermel, B., Vega, R. B., Frey, N., McKinsey, T. A., Olson, E. N.,
Bassel-Duby, R., and Williams, R. S. (2000) Circ. Res. 87, E61–E68
24. Hofmann, T., Obukhov, A. G., Schaefer, M., Harteneck, C., Gudermann,
T., and Schultz, G. (1999) Nature 397, 259–263
25. Lievremont, J. P., Bird, G. S., and Putney, J. W., Jr. (2005) Mol. Pharmacol.
68, 758 –762
26. Pigozzi, D., Ducret, T., Tajeddine, N., Gala, J. L., Tombal, B., and Gailly, P.
(2006) Cell Calcium 39, 401– 415
27. Trevillyan, J. M., Chiou, X. G., Chen, Y. W., Ballaron, S. J., Sheets, M. P.,
Smith, M. L., Wiedeman, P. E., Warrior, U., Wilkins, J., Gubbins, E. J.,
Gagne, G. D., Fagerland, J., Carter, G. W., Luly, J. R., Mollison, K. W., and
Djuric, S. W. (2001) J. Biol. Chem. 276, 48118 48126
28. Zitt, C., Strauss, B., Schwarz, E. C., Spaeth, N., Rast, G., Hatzelmann, A.,
and Hoth, M. (2004) J. Biol. Chem. 279, 12427–12437
29. He, L. P., Hewavitharana, T., Soboloff, J., Spassova, M. A., and Gill, D. L.
(2005) J. Biol. Chem. 280, 10997–11006
30. Venkatesh, N., Feng, Y., Decker, B., Yacono, P., Golan, D., Mitchison,
T., and McKeon, F. (2004) Proc. Natl. Acad. Sci. U. S. A. 101,
8969 8974
31. Nilius, B., Voets, T., and Peters, J. (2005) Sci. STKE 2005, re8
32. Vandebrouck, C., Martin, D., Colson-Van, S. M., Debaix, H., and Gailly, P.
(2002) J. Cell Biol. 158, 1089 –1096
33. Yu, Y., Fantozzi, I., Remillard, C. V., Landsberg, J. W., Kunichika, N., Pla-
toshyn, O., Tigno, D. D., Thistlethwaite, P. A., Rubin, L. J., and Yuan, J. X.
(2004) Proc. Natl. Acad. Sci. U. S. A. 101, 13861–13866
34. Kunichika, N., Landsberg, J. W., Yu, Y., Kunichika, H., Thistlethwaite,
P. A., Rubin, L. J., and Yuan, J. X. (2004) Am. J. Respir. Crit. Care Med. 170,
1101–1107
35. Seth, M., Sumbilla, C., Mullen, S. P., Lewis, D., Klein, M. G., Hussain, A.,
Soboloff, J., Gill, D. L., and Inesi, G. (2004) Proc. Natl. Acad. Sci. U. S. A.
101, 16683–16688
TRPC Channels Promote Cardiomyocyte Hypertrophy
33496 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 NUMBER 44 NOVEMBER 3, 2006
by guest on January 13, 2016http://www.jbc.org/Downloaded from
Michael R. Bristow, Eric N. Olson and Timothy A. McKinsey
Erik W. Bush, David B. Hood, Philip J. Papst, Joseph A. Chapo, Wayne Minobe,
Hypertrophy through Activation of Calcineurin Signaling
Canonical Transient Receptor Potential Channels Promote Cardiomyocyte
doi: 10.1074/jbc.M605536200 originally published online September 1, 2006
2006, 281:33487-33496.J. Biol. Chem.
10.1074/jbc.M605536200Access the most updated version of this article at doi:
Alerts:
When a correction for this article is posted
When this article is cited
to choose from all of JBC's e-mail alertsClick here
http://www.jbc.org/content/281/44/33487.full.html#ref-list-1
This article cites 32 references, 16 of which can be accessed free at
by guest on January 13, 2016http://www.jbc.org/Downloaded from
... TRPC5 was originally identified in the central nervous system, but is also known to be located in peripheral tissues, including those of the cardiovascular system [5]. mRNA transcripts and protein have been detected in endothelial cells [6] and vascular myocytes [7,8], as well as in cardiomyocytes [5,9,10]. However, our understanding of the functional role of TRPC5 in the cardiovascular system is at an early stage. ...
... In vitro silencing of SERCA2a in cultured rat cardiomyocytes resulted in an increased TRPC5 expression [18], implicating a potential role in cardiac disease. In rodents, an increased expression has been noted in failing cardiac left ventricles of spontaneously hypertensive rats [19], but studies using a pressure overload model of cardiac hypertrophy have implicated TRPC1 [10], TRPC3 [9,20], and TRPC6 [21] in pathogenesis, whilst TRPC5's expression was reported to be unchanged [9,10]. In humans, its expression was increased in left ventricle biopsies from patients with end-stage dilated cardiomyopathy [9], suggesting that TRPC5 may be more relevant in humans or severe cardiac dysfunction. ...
... In vitro silencing of SERCA2a in cultured rat cardiomyocytes resulted in an increased TRPC5 expression [18], implicating a potential role in cardiac disease. In rodents, an increased expression has been noted in failing cardiac left ventricles of spontaneously hypertensive rats [19], but studies using a pressure overload model of cardiac hypertrophy have implicated TRPC1 [10], TRPC3 [9,20], and TRPC6 [21] in pathogenesis, whilst TRPC5's expression was reported to be unchanged [9,10]. In humans, its expression was increased in left ventricle biopsies from patients with end-stage dilated cardiomyopathy [9], suggesting that TRPC5 may be more relevant in humans or severe cardiac dysfunction. ...
Article
Full-text available
TRPC5 is a non-selective cation channel that is expressed in cardiomyocytes, but there is a lack of knowledge of its (patho)physiological role in vivo. Here, we examine the role of TRPC5 on cardiac function under basal conditions and during cardiac hypertrophy. Cardiovascular parameters were assessed in wild-type (WT) and global TRPC5 knockout (KO) mice. Despite no difference in blood pressure or activity, heart rate was significantly reduced in TRPC5 KO mice. Echocardiography imaging revealed an increase in stroke volume, but cardiac contractility was unaffected. The reduced heart rate persisted in isolated TRPC5 KO hearts, suggesting changes in basal cardiac pacing. Heart rate was further investigated by evaluating the reflex change following drug-induced pressure changes. The reflex bradycardic response following phenylephrine was greater in TRPC5 KO mice but the tachycardic response to SNP was unchanged, indicating an enhancement in the parasympathetic control of the heart rate. Moreover, the reduction in heart rate to carbachol was greater in isolated TRPC5 KO hearts. To evaluate the role of TRPC5 in cardiac pathology, mice were subjected to abdominal aortic banding (AAB). An exaggerated cardiac hypertrophy response to AAB was observed in TRPC5 KO mice, with an increased expression of hypertrophy markers, fibrosis, reactive oxygen species, and angiogenesis. This study provides novel evidence for a direct effect of TRPC5 on cardiac function. We propose that (1) TRPC5 is required for maintaining heart rate by regulating basal cardiac pacing and in response to pressure lowering, and (2) TRPC5 protects against pathological cardiac hypertrophy.
... have low levels of TRPC expression; however, in cases of pathological hypertrophy and heart failure, there appears to be an increase in both expression and activity (12,25, 26). It is thought that TRPC6 starts the Ca 2+ -dependent signaling pathways that result in heart failure, myocardial infarction, and pathological hypertrophy (27-30). ...
Preprint
Full-text available
Cell and body damage are encouraged by myocardial ischaemia/reperfusion injury (MI/RI). The pathophysiology of cardiac I/R damage is mostly caused by oxidative stress, calcium excess, and inflammation. Cardiomyocytes' MI/RI is significantly influenced by autophagy. Through lysosome-mediated degradation, autophagy is a dynamic process that eliminates damaged organelles and long-lived proteins. This process has an antioxidant impact that lowers oxidative stress. An essential part of MI/RI is played by canonical transient receptor potential channel 6 (TRPC6), a nonselective cation channel that permits passage of Ca ²⁺ . It is yet unknown, nevertheless, how TRPC6 and autophagy relate to each other in MI/RI. In this work, we observed that following myocardial/ischaemia-reperfusion, autophagy and apoptosis increased, and that the production of autophagosomes and autophagy-related proteins, the Bax/Bcl-2 ratio, and cell damage decreased under TRPC6 suppression or knockout. Myocardial ischaemia/reperfusion injury may benefit from targeting TRPC6-mediated autophagy as a potential novel therapeutic target in the future.
... Conversely, the use of CN and NFAT inhibitors in diverse experimental models of HF has shown to attenuate the maladaptive response [14,29,30]. Notably, among TRP channels, various TRPCs have been involved in the development of pathological remodeling through activation of the CN/NFATc3 pathway in CM [8,31]. Although TRPV4 channels are known to activate the same pathway in other cell types [15], to the best of our knowledge, ours is the first study to demonstrate their role in the activation of the Ca 2+ /CN/NFATc3 pathway in cardiac FB as a mechanism to explain the development of maladaptive remodeling. ...
Article
Full-text available
TRPV4 channels, which respond to mechanical activation by permeating Ca2+ into the cell, may play a pivotal role in cardiac remodeling during cardiac overload. Our study aimed to investigate TRPV4 involvement in pathological and physiological remodeling through Ca2+-dependent signaling. TRPV4 expression was assessed in heart failure (HF) models, induced by isoproterenol infusion or transverse aortic constriction, and in exercise-induced adaptive remodeling models. The impact of genetic TRPV4 inhibition on HF was studied by echocardiography, histology, gene and protein analysis, arrhythmia inducibility, Ca2+ dynamics, calcineurin (CN) activity, and NFAT nuclear translocation. TRPV4 expression exclusively increased in HF models, strongly correlating with fibrosis. Isoproterenol-administered transgenic TRPV4−/− mice did not exhibit HF features. Cardiac fibroblasts (CFb) from TRPV4+/+ animals, compared to TRPV4−/−, displayed significant TRPV4 overexpression, elevated Ca2+ influx, and enhanced CN/NFATc3 pathway activation. TRPC6 expression paralleled that of TRPV4 in all models, with no increase in TRPV4−/− mice. In cultured CFb, the activation of TRPV4 by GSK1016790A increased TRPC6 expression, which led to enhanced CN/NFATc3 activation through synergistic action of both channels. In conclusion, TRPV4 channels contribute to pathological remodeling by promoting fibrosis and inducing TRPC6 upregulation through the activation of Ca2+-dependent CN/NFATc3 signaling. These results pose TRPV4 as a primary mediator of the pathological response.
... However, in HFpEF-related models, cardiomyocyte Ca 2+ transients are often found to be normal or even enhanced [104][105][106][107][108][109][110], suggesting an adaptive phase where Ca 2+ flux shifts towards cardiomyocyte Ca 2+ accumulation [111]. This adaptation may involve excessive Ca 2+ entry through CaV1.2 and transient receptor potential (TRP) channels [112][113][114], along with increased SR Ca 2+ release through RyR [104,105,110] and IP3R2 [109], enhancing Ca 2+ cycling and contraction. Yet, without a simultaneous enhancement of SERCA activity, this leads to elevated diastolic Ca 2+ , preserved or enhanced Ca 2+ transient amplitude, but slower Ca 2+ reuptake kinetics and impaired relaxation. ...
Article
Full-text available
Heart failure (HF) presents a significant clinical challenge, with current treatments mainly easing symptoms without stopping disease progression. The targeting of calcium (Ca2+) regulation is emerging as a key area for innovative HF treatments that could significantly alter disease outcomes and enhance cardiac function. In this review, we aim to explore the implications of altered Ca2+ sensitivity, a key determinant of cardiac muscle force, in HF, including its roles during systole and diastole and its association with different HF types—HF with preserved and reduced ejection fraction (HFpEF and HFrEF, respectively). We further highlight the role of the two rate constants kon (Ca2+ binding to Troponin C) and koff (its dissociation) to fully comprehend how changes in Ca2+ sensitivity impact heart function. Additionally, we examine how increased Ca2+ sensitivity, while boosting systolic function, also presents diastolic risks, potentially leading to arrhythmias and sudden cardiac death. This suggests that strategies aimed at moderating myofilament Ca2+ sensitivity could revolutionize anti-arrhythmic approaches, reshaping the HF treatment landscape. In conclusion, we emphasize the need for precision in therapeutic approaches targeting Ca2+ sensitivity and call for comprehensive research into the complex interactions between Ca2+ regulation, myofilament sensitivity, and their clinical manifestations in HF.
... 83 This study confirms a strong expression of the TRPC1, TRPC4, TRPC6, and TRPC2 channels and all members of the TRPML subfamily (MCOLN1, MCOLN2, and MCOLN3) (Figure 2(G)). TRPC channels appear to play a significant role in cardiac dysfunction, and nearly all of them are upregulated in HCFs, [84][85][86] except for TRPC3, TRPC7, and TRPC5. The study conducted by Yamaguchi et al. revealed the significance of TRPC3/6/7 channels in the cellular response to mechanical stressors such as flow, stretch, and osmotic pressure. ...
Article
Full-text available
Human cardiac fibroblasts (HCFs) have mRNA transcripts that encode different mechanosensitive ion channels and channel regulatory proteins whose functions are not known yet. The primary goal of this work was to define the mechanosensitive ion channelome of HCFs. The most common type of cationic channel is the transient receptor potential (TRP) family, which is followed by the TWIK-related K ⁺ channel (TREK), transmembrane protein 63 (TMEM63), and PIEZO channel (PIEZO) families. In the sodium-dependent NON-voltage-gated channel (SCNN) subfamily, only SCNN1D was shown to be highly expressed. Particular members of the acid-sensing ion channel (ASIC) (ASIC1 and ASIC3) subfamilies were also significantly expressed. The transcripts per kilobase million (TPMs) for Piezo 2 were almost 100 times less abundant than those for Piezo 1. The tandem of P domains in a weak inward rectifying K ⁺ channel (TWIK)-2 channel, TWIK-related acid-sensitive K ⁺ channel (TASK)-5, TASK-1, and the TWIK-related K1 (TREK-1) channel were the four most prevalent types in the K2P subfamily. The highest expression in the TRPP subfamily was found for PKD2 and PKD1, while in the TRPM subfamily, it was found for TRPM4, TRPM7, and TRPM3. TRPV2, TRPV4, TRPV3, and TRPV6 (all members of the TRPV subfamily) were also substantially expressed. A strong expression of the TRPC1, TRPC4, TRPC6, and TRPC2 channels and all members of the TRPML subfamily (MCOLN1, MCOLN2, and MCOLN3) was also shown. In terms of the transmembrane protein 16 (TMEM16) family, the HCFs demonstrated significant expression of the TMEM16H, TMEM16F, TMEM16J, TMEM16A, and TMEM16G channels. TMC3 is the most expressed channel in HCFs of all known members of the transmembrane channel-like protein (TMC) family. This analysis of the mechanosensitive ionic channel transcriptome in HCFs: (1) agrees with previously documented findings that all currently identified mechanosensitive channels play a significant and well recognized physiological function in elucidating the mechanosensitive characteristics of HCFs; (2) supports earlier preliminary reports that point to the most common expression of the TRP mechanosensitive family in HCFs; and (3) points to other new mechanosensitive channels (TRPC1, TRPC2, TWIK-2, TMEM16A, ASIC1, and ASIC3).
... In animal models of hypertension, however, TRPC6 inhibitor treatment can revert this transition and reduce blood pressure levels. 87,88 Prior research has indicated that PTEN, a phosphatase and tension homolog absent on chromosome 10, plays a regulatory role in TRPC6 (transient receptor potential canonical 6). The regulation of phenotypic differentiation in vascular smooth muscle cells (VSMC) is partially influenced by this association. ...
Article
Objective The purpose of this study is to develop an animal model of Chronic Intermittent Hypoxia (CIH) and investigate the role of the TRPC5 channel in cardiac damage in OSAHS rats. Methods Twelve male Sprague Dawley rats were randomly divided into the CIH group and the Normoxic Control (NC) group. Changes in structure, function, and pathology of heart tissue were observed through echocardiography, transmission electron microscopy, HE-staining, and TUNEL staining. Results The Interventricular Septum thickness at diastole (IVSd) and End-Diastolic Volume (EDV) of rats in the CIH group significantly increased, whereas the LV ejection fraction and LV fraction shortening significantly decreased. TEM showed that the myofilaments in the CIH group were loosely arranged, the sarcomere length varied, the cell matrix dissolved, the mitochondrial cristae were partly flocculent, the mitochondrial outer membrane dissolved and disappeared, and some mitochondria were swollen and vacuolated. The histopathological examination showed that the cardiomyocytes in the CIH group were swollen with granular degeneration, some of the myocardial fibers were broken and disorganized, and most of the nuclei were vacuolar and hypochromic. Conclusion CIH promoted oxidative stress, the influx of Ca²⁺, and the activation of the CaN/NFATc signaling pathway, which led to pathological changes in the morphology and ultrastructure of cardiomyocytes, the increase of myocardial apoptosis, and the decrease of myocardial contractility. These changes may be associated with the upregulation of TRPC5.
Chapter
Canonical or classical transient receptor potential (TRPC) proteins are nonselective cationic channels and a subset of TRP superfamily which are ubiquitously expressed in mammalian cells. Activation of TRPC channels permits the inflow of calcium (Ca2+) and other monovalent alkali cations such as Na+ into the cytosol resulting in cell depolarization and an increase in intracellular Ca2+ concentration. Hence, TRPC channels are multifunctional signaling protein complexes that play crucial roles in a vast range of human physiological and pathological processes such as cancer, neurological disorders, cardiovascular diseases, pain, and chronic kidney disease. Therefore, they serve as potential drug target for several human diseases and disorders. In this chapter, the TRPC structure, function, regulation, and recently identified potent and selective modulators of these channels will be discussed. Functional tetrameric TRPC channels assemble as homo- or heterotetrameric complexes. To date, seven TRPC subfamily members have been identified (TRPC1–TRPC7), and TRPC2 is a pseudogene in humans. Recent TRPC structural studies have offered new insights into channel architecture and assembly. High-resolution structures of TRPC3, TRPC4, TRPC5, and TRPC6 homotetramers have been reported. Together, potentially compound profiling will lead to discovering and characterizing novel drug candidates that are intended to generate innovative therapeutic therapies.
Chapter
Cardiac electrical activity is mainly determined by a set of voltage-dependent channels that control depolarizing currents carried by Na+ and Ca2+ cations and repolarizing currents carried by K+ and Cl− ions. Arrhythmia often results from a disorder in these currents or from the occurrence of additional inward currents such as those generated by the transient receptor potential (TRP) channels. In mammals, the TRP family comprises six major subfamilies; TRPC (canonical), TRPV (vanilloid), TRPA (ankyrin), TRPM (melastatin), TRPP (polycystin), and TRPML (mucolipin). In the heart, TRP channels are known to be involved in various diseases, including hypertrophy, heart failure, and arrhythmia. A large part of this chapter focuses on the potential contribution of TRP channels, namely TRPCs and TRPM4, that could carry an inward current at the resting potential to modulate cardiac rhythm, as well as on their potential arrhythmic effects. Specific attention is given to the proarrhythmic effects of ATP as a purinergic agonist. Under ischemia, a burst of ATP is released that stimulates P2Y2 receptors, which through phospholipase C activates heterotetrameric TRPC3/TRPC7 channels and, as well, releases Ca2+ from the sarcoplasmic reticulum to activate TRPM4. The subsequent inward currents could depolarize the cell and trigger anomalous activity. Furthermore, several other neurotransmitters that all induce through G protein or tyrosine kinase, the formation of DAG could, as well, modulate cardiac rhythm and trigger arrhythmia.
Article
Full-text available
The canonical transient receptor potential channel (TRPC) proteins form Ca²⁺-permeable cation channels that are involved in various heart diseases. However, the roles of specific TRPC proteins in myocardial ischemia/reperfusion (I/R) injury remain poorly understood. We observed that TRPC1 and TRPC6 were highly expressed in the area at risk (AAR) in a coronary artery ligation induced I/R model. Trpc1−/− mice exhibited improved cardiac function, lower serum Troponin T and serum creatine kinase level, smaller infarct volume, less fibrotic scars, and fewer apoptotic cells after myocardial-I/R than wild-type or Trpc6−/− mice. Cardiomyocyte-specific knockdown of Trpc1 using adeno-associated virus 9 mitigated myocardial I/R injury. Furthermore, Trpc1 deficiency protected adult mouse ventricular myocytes (AMVMs) and HL-1 cells from death during hypoxia/reoxygenation (H/R) injury. RNA-sequencing-based transcriptome analysis revealed differential expression of genes related to reactive oxygen species (ROS) generation in Trpc1−/− cardiomyocytes. Among these genes, oxoglutarate dehydrogenase-like (Ogdhl) was markedly downregulated. Moreover, Trpc1 deficiency impaired the calcineurin (CaN)/nuclear factor-kappa B (NF-κB) signaling pathway in AMVMs. Suppression of this pathway inhibited Ogdhl upregulation and ROS generation in HL-1 cells under H/R conditions. Chromatin immunoprecipitation assays confirmed NF-κB binding to the Ogdhl promoter. The cardioprotective effect of Trpc1 deficiency was canceled out by overexpression of NF-κB and Ogdhl in cardiomyocytes. In conclusion, our findings reveal that TRPC1 is upregulated in the AAR following myocardial I/R, leading to increased Ca²⁺ influx into associated cardiomyocytes. Subsequently, this upregulates Ogdhl expression through the CaN/NF-κB signaling pathway, ultimately exacerbating ROS production and aggravating myocardial I/R injury.
Article
Full-text available
Signaling events controlled by calcineurin promote cardiac hypertrophy, but the degree to which such pathways are required to transduce the effects of various hypertrophic stimuli remains uncertain. In particular, the administration of immunosuppressive drugs that inhibit calcineurin has inconsistent effects in blocking cardiac hypertrophy in various animal models. As an alternative approach to inhibiting calcineurin in the hearts of intact animals, transgenic mice were engineered to overexpress a human cDNA encoding the calcineurin-binding protein, myocyte-enriched calcineurin-interacting protein-1 (hMCIP1) under control of the cardiac-specific, α-myosin heavy chain promoter (α-MHC). In unstressed mice, forced expression of hMCIP1 resulted in a 5–10% decline in cardiac mass relative to wild-type littermates, but otherwise produced no apparent structural or functional abnormalities. However, cardiac-specific expression of hMCIP1 inhibited cardiac hypertrophy, reinduction of fetal gene expression, and progression to dilated cardiomyopathy that otherwise result from expression of a constitutively active form of calcineurin. Expression of the hMCIP1 transgene also inhibited hypertrophic responses to β-adrenergic receptor stimulation or exercise training. These results demonstrate that levels of hMCIP1 producing no apparent deleterious effects in cells of the normal heart are sufficient to inhibit several forms of cardiac hypertrophy, and suggest an important role for calcineurin signaling in diverse forms of cardiac hypertrophy. The future development of measures to increase expression or activity of MCIP proteins selectively within the heart may have clinical value for prevention of heart failure.
Article
Full-text available
A pattern of left ventricular hypertrophy evident on the electrocardiogram is a harbinger of morbidity and mortality from cardiovascular disease. Echocardiography permits the noninvasive determination of left ventricular mass and the examination of its role as a precursor of morbidity and mortality. We examined the relation of left ventricular mass to the incidence of cardiovascular disease, mortality from cardiovascular disease, and mortality from all causes in 3220 subjects enrolled in the Framingham Heart Study who were 40 years of age or older and free of clinically apparent cardiovascular disease, in whom left ventricular mass was determined echocardiographically. During a four-year follow-up period, there were 208 incident cardiovascular events, 37 deaths from cardiovascular disease, and 124 deaths from all causes. Left ventricular mass, determined echocardiographically, was associated with all outcome events. This relation persisted after we adjusted for age, diastolic blood pressure, pulse pressure, treatment for hypertension, cigarette smoking, diabetes, obesity, the ratio of total cholesterol to high-density lipoprotein cholesterol, and electrocardiographic evidence of left ventricular hypertrophy. In men, the risk factor-adjusted relative risk of cardiovascular disease was 1.49 for each increment of 50 g per meter in left ventricular mass corrected for the subject's height (95 percent confidence interval, 1.20 to 1.85); in women, it was 1.57 (95 percent confidence interval, 1.20 to 2.04). Left ventricular mass (corrected for height) was also associated with the incidence of death from cardiovascular disease (relative risk, 1.73 [95 percent confidence interval, 1.19 to 2.52] in men and 2.12 [95 percent confidence interval, 1.28 to 3.49] in women). Left ventricular mass (corrected for height) was associated with death from all causes (relative risk, 1.49 [95 percent confidence interval, 1.14 to 1.94] in men and 2.01 [95 percent confidence interval, 1.44 to 2.81] in women). We conclude that the estimation of left ventricular mass by echocardiography offers prognostic information beyond that provided by the evaluation of traditional cardiovascular risk factors. An increase in left ventricular mass predicts a higher incidence of clinical events, including death, attributable to cardiovascular disease.
Article
Full-text available
Here we describe a small family of proteins, termed MCIP1 and MCIP2 (for myocyte-enrichedcalcineurin interacting protein), that are expressed most abundantly in striated muscles and that form a physical complex with calcineurin A. MCIP1 is encoded byDSCR1, a gene located in the Down syndrome critical region. Expression of the MCIP family of proteins is up-regulated during muscle differentiation, and their forced overexpression inhibits calcineurin signaling to a muscle-specific target gene in a myocyte cell background. Binding of MCIP1 to calcineurin A requires sequence motifs that resemble calcineurin interacting domains found in NFAT proteins. The inhibitory action of MCIP1 involves a direct association with the catalytic domain of calcineurin, rather than interference with the function of downstream components of the calcineurin signaling pathway. The interaction between MCIP proteins and calcineurin may modulate calcineurin-dependent pathways that control hypertrophic growth and selective programs of gene expression in striated muscles.
Article
Calcium signaling activates the phosphatase calcineurin and induces movement of NFATc proteins into the nucleus, where they cooperate with other proteins to form complexes on DNA. Nuclear import is opposed by kinases such as GSK3, thereby rendering transcription continuously responsive to receptor occupancy. Disruptions of the genes involved in NFAT signaling are implicating this pathway as a regulator of developmental cell–cell interactions.
Article
This study uses echocardiography to characterize the pattern of left ventricular hypertrophy in a new hypertensive heart failure-prone rat strain designated SHHF/Mcc-cp (SHHF). M-mode echocardiograms of the left ventricle in nine 10- to 12-month old SHHF rats and nine age-matched spontaneously hypertensive rats (SHR) were compared. Wistar-Kyoto and Sprague-Dawley strains served as the normotensive control group. SHHF rats had significantly greater left ventricular mass than did rats in the normotensive control group. Although left ventricular mass was not different between SHHF and SHR, significant differences were seen in the pattern of left ventricular remodeling as determined by relative wall thickness. These differences in left ventricular remodeling may explain the earlier development of heart failure in SHHF. The different patterns of left ventricular hypertrophy in SHHF and SHR suggests that heart failure in SHHF is not mediated by hypertension alone.
Article
External load plays a critical role in determining muscle mass and its phenotype in cardiac myocytes. Cardiac myocytes have the ability to sense mechanical stretch and convert it into intracellular growth signals, which lead to hypertrophy. Mechanical stretch of cardiac myocytes in vitro causes activation of multiple second messenger systems that are very similar to growth factor-induced cell signaling systems. Stretch of neonatal rat cardiac myocytes stimulates a rapid secretion of angiotensin II which, together with other growth factors, mediates stretch-induced hypertrophic responses in vitro. In this review, various cell signaling mechanisms initiated by mechanical stress on cardiac myocytes are summarized with emphasis on potential mechanosensing mechanisms and the relationship between mechanical loading and the cardiac renin-angiotensin system.
Article
In response to numerous pathologic stimuli, the myocardium undergoes a hypertrophic response characterized by increased myocardial cell size and activation of fetal cardiac genes. We show that cardiac hypertrophy is induced by the calcium-dependent phosphatase calcineurin, which dephosphorylates the transcription factor NF-AT3, enabling it to translocate to the nucleus. NF-AT3 interacts with the cardiac zinc finger transcription factor GATA4, resulting in synergistic activation of cardiac transcription. Transgenic mice that express activated forms of calcineurin or NF-AT3 in the heart develop cardiac hypertrophy and heart failure that mimic human heart disease. Pharmacologic inhibition of calcineurin activity blocks hypertrophy in vivo and in vitro. These results define a novel hypertrophic signaling pathway and suggest pharmacologic approaches to prevent cardiac hypertrophy and heart failure.
Article
Eukaryotic cells respond to many hormones and neurotransmitters with increased activity of the enzyme phospholipase C and a subsequent rise in the concentration of intracellular free calcium ([Ca2+]i). The increase in [Ca2+]i occurs as a result of the release of Ca2+ from intracellular stores and an influx of Ca2+ through the plasma membrane; this influx of Ca2+ may or may not be store-dependent. Drosophila transient receptor potential (TRP) proteins and some mammalian homologues (TRPC proteins) are thought to mediate capacitative Ca2+ entry. Here we describe the molecular mechanism of store-depletion-independent activation of a subfamily of mammalian TRPC channels. We find that hTRPC6 is a non-selective cation channel that is activated by diacylglycerol in a membrane-delimited fashion, independently of protein kinases C activated by diacylglycerol. Although hTRPC3, the closest structural relative of hTRPC6, is activated in the same way, TRPCs 1, 4 and 5 and the vanilloid receptor subtype 1 are unresponsive to the lipid mediator. Thus, hTRPC3 and hTRPC6 represent the first members of a new functional family of second-messenger-operated cation channels, which are activated by diacylglycerol.
Article
Calcium is central in the regulation of cardiac contractility, growth and gene expression. Variations in the amplitude, frequency and compartmentalization of calcium signals are decoded by calcium/calmodulin-dependent enzymes, ion channels and transcription factors. Understanding the circuitry for calcium signaling creates opportunities for pharmacological modification of cardiac function.
Article
Calcineurin, a calcium/calmodulin-regulated protein phosphatase, modulates gene expression in cardiac and skeletal muscles during development and in remodeling responses such as cardiac hypertrophy that are evoked by environmental stresses or disease. Recently, we identified two genes encoding proteins (MCIP1 and MCIP2) that are enriched in striated muscles and that interact with calcineurin to inhibit its enzymatic activity. In the present study, we show that expression of MCIP1 is regulated by calcineurin activity in hearts of mice with cardiac hypertrophy, as well as in cultured skeletal myotubes. In contrast, expression of MCIP2 in the heart is not altered by activated calcineurin but responds to thyroid hormone, which has no effect on MCIP1. A approximately 900-bp intragenic segment located between exons 3 and 4 of the MCIP1 gene functions as an alternative promoter that responds to calcineurin. This region includes a dense cluster of 15 consensus binding sites for NF-AT transcription factors. Because MCIP proteins can inhibit calcineurin, these results suggest that MCIP1 participates in a negative feedback circuit to diminish potentially deleterious effects of unrestrained calcineurin activity in cardiac and skeletal myocytes. Inhibitory effects of MCIP2 on calcineurin activity may be pertinent to gene switching events driven by thyroid hormone in striated muscles. The full text of this article is available at http://www. circresaha.org.