ArticlePDF AvailableLiterature Review

Abstract and Figures

Sarcomeric dysfunction plays a central role in reduced cardiac pump function in heart failure. This review focuses on the alterations in sarcomeric proteins in diseased myocardium that range from altered isoform expression to post-translational protein changes such as proteolysis and phosphorylation. Recent studies in animal models of heart failure and human failing myocardium converge and indicate that sarcomeric dysfunction, including altered maximum force development, Ca(2+) sensitivity, and increased passive stiffness, largely originates from altered protein phosphorylation, caused by neurohumoral-induced alterations in the kinase-phosphatase balance inside the cardiomyocytes. Novel therapies, which specifically target phosphorylation sites within sarcomeric proteins or the kinases and phosphatases involved, might improve cardiac function in heart failure.
Content may be subject to copyright.
Review
Sarcomeric dysfunction in heart failure
Nazha Hamdani1, Viola Kooij1, Sabine van Dijk1, Daphne Merkus2, Walter J. Paulus1,
Cris dos Remedios3, Dirk J. Duncker2, Ger J.M. Stienen1, and Jolanda van der Velden1*
1
Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7,
1081 BT Amsterdam, The Netherlands;
2
Experimental Cardiology, Thoraxcenter, Cardiovascular Research School COEUR,
Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; and
3
Muscle Research Unit,
Institute for Biomedical Research, The University of Sydney, Sydney, Australia
Received 30 July 2007; revised 9 October 2007; accepted 24 October 2007; online publish-ahead-of-print 30 November 2007
Time for primary review: 27 days
Sarcomeric dysfunction plays a central role in reduced cardiac pump function in heart failure. This
review focuses on the alterations in sarcomeric proteins in diseased myocardium that range from
altered isoform expression to post-translational protein changes such as proteolysis and phosphoryl-
ation. Recent studies in animal models of heart failure and human failing myocardium converge and
indicate that sarcomeric dysfunction, including altered maximum force development, Ca
2þ
sensitivity,
and increased passive stiffness, largely originates from altered protein phosphorylation, caused by
neurohumoral-induced alterations in the kinase
phosphatase balance inside the cardiomyocytes.
Novel therapies, which specifically target phosphorylation sites within sarcomeric proteins or the
kinases and phosphatases involved, might improve cardiac function in heart failure.
KEYWORDS
Sarcomere;
Cardiomyocyte;
Contractility;
Heart failure
1. Sarcomeric dysfunction
The failing heart is characterized by reduced contractility
(systolic dysfunction) and/or impaired filling (diastolic dys-
function). A number of factors, including changes in
cardiac structure (dilation and hypertrophy), apoptotic and
necrotic cell death, maladaptive remodelling of the extra-
cellular matrix, abnormal energy metabolism, impaired
calcium handling, and neurohumoral disturbances have
been implicated in the initiation and progression of heart
failure.
1
4
Recent studies revealed that alterations in sarco-
meric function play a prominent role in reduced cardiac
pump function.
Sarcomeric function is determined by the expression
levels of multiple isoforms and by post-translational modifi-
cations of sarcomeric proteins. During muscle contraction a
molecular interaction takes place between the thin (actin)
and thick (myosin) filament of the sarcomeres, which is trig-
gered by a rise in the intracellular calcium and is driven by
the energy from ATP hydrolysis.
5
The tropomyosin
troponin
complex inhibits the actin
myosin interaction at low intra-
cellular free calcium (Figure 1A). This inhibition is released
when intracellular free calcium increases and binds to
troponin C (Figure 1B). Alterations in sarcomeric protein
composition under pathological conditions will influence
contractile performance of the heart. Within the first part
of this review, we discuss the functional role of individual
sarcomeric protein isoforms and of post-translational
protein modifications such as proteolysis and phosphoryl-
ation. In the second part, we highlight the major changes
in sarcomeric function reported in failing myocardium and
discuss the most likely underlying protein modifications.
2. Isoform composition and sarcomeric
dysfunction
2.1 Myosin heavy chains
The thick filament is composed of myosin, which consists of
two myosin heavy chains (MHC), and two pairs of myosin
light chains (MLCs) (Figure 1). One of the major isoform
changes which has been observed in hypertrophied and
failing ventricular myocardium is the shift from the fast
a-MHC to the slow b-MHC.
6
8
The magnitude of the MHC
shift largely depends on the amount of endogenous a-MHC
present in ventricular tissue, which is species-dependent,
being largest in small rodents and smallest in human.
6
11
Hence, the functional significance of the shift in MHC com-
position in diseased human ventricles is still a matter of
debate.
8,10,12
The MHCs carry the site for ATP hydrolysis and are import-
ant determinants of the rate of energy consumption and the
speed of contraction of the sarcomeres, which are closely
related.
13
In vitro studies have shown that the a-MHC
isoform has a higher ATPase activity
14
and a higher actin fila-
ment sliding velocity compared with the b-MHC isoform.
15
*Corresponding author. Tel: þ31 20 4448123; fax: þ31 20 4448255.
E-mail address: j.vandervelden@vumc.nl
Published on behalf of the European Society of Cardiology. All rights reserved. &The Author 2007.
For permissions please email: journals.permissions@oxfordjournals.org.
Cardiovascular Research (2008) 77, 649
658
doi:10.1093/cvr/cvm079
by guest on June 5, 2013http://cardiovascres.oxfordjournals.org/Downloaded from
From experiments in permeabilized cardiac preparations
from small mammals and humans it followed that the
b-MHC is 3
5 times more economical,
11,16,17
but is associ-
ated with reduced power output
18
and shortening vel-
ocity
17,18
compared with the a-MHC isoform. Consequently,
in rats a shift from a-tob-MHC coincided with significant
reductions in ATPase activity,
7,19
tension cost,
19
and power
output.
18
Evidence that a shift in MHC may be of pathophy-
siological relevance for human sarcomeric dysfunction was
already provided in 1962 by Alpert and Gordon
20
who
reported reduced myofibrillar ATPase activity in human con-
gestive heart failure. However, subsequent human studies
failed to unequivocally link this observation with an MHC
isoform shift in failing ventricular myocardium. First, no
differences were found in functional properties of myosin
isolated from non-failing and failing hearts,
10,21,22
indicating
that protein alterations other than a change in myosin com-
position are responsible for the reduction in myofilament
ATPase activity.
20,23
Moreover, various expression levels of
a-MHC (ranging from 0 to 30%) have been reported in the
ventricles of different individuals.
8,10,12,24
This may be due
to age-dependent changes in MHC composition and hetero-
geneous expression of MHC isoforms within the ventricular
wall.
25,26
Both in human
25
and rat
26
ventricular tissue, a
regional difference in MHC expression has been observed,
characterized by a higher expression of the fast a-MHC in
the subepicardial than in the subendocardial layer,
consistent with the shorter action potential and contraction
duration in the subepi- when compared with the sub-
endocardium. Thus, in human ventricular myocardium the
change in MHC isoform composition during heart failure
will be variable and this may have obscured significant
effects on sarcomeric function. However, recent studies
have shown that even a small shift will have a significant
impact on cardiomyocyte contractility.
11,27
In contrast to
ventricular human tissue, human atria contain 80% of
a-MHC.
12,24
In atrial fibrillation, the b-MHC expression
increased almost two-fold, which coincided with a reduction
in kinetics of force re-development.
28
Overall, the shift towards the slow and more economical
b-MHC isoform occurs in human diseased atria and to a
lesser extent in diseased human ventricular myocardium.
The MHC shift may be beneficial under pathological con-
ditions, since less energy is required to maintain cardiac
pump function at rest, though at the expense of reduced
speed of contraction and power output.
2.2 Myosin light chains
Apart from the shift in MHC, changes may occur in the
expression pattern of MLCs within the heart. In particular,
isoform changes have been reported for MLC-1 (or essential
MLC), both in atrial and ventricular tissue. MLC-1 not only
interacts with MHC, but also binds to actin with its N-
terminus. It exists in two forms, a ventricular (VLC-1) and
an atrial (ALC-1) form.
29
The latter is expressed in the
entire heart muscle during foetal and early life and is sub-
sequently replaced by the ventricular form. In the ventricles
of patients with hypertrophic obstructive cardiomyopathy
relatively high amounts of ALC-1 were found, which corre-
lated with the maximal rate of force development.
30
Apart
from its positive effect on dynamics of contraction, replace-
ment of VLC-1 by ALC-1 increased isometric force develop-
ment of ventricular preparations at maximal and
submaximal activation.
29
Morano et al.
31
hypothesized that
MLC-1 tethers MHC to actin and thereby restrains cross-
bridge cycling and reduces force generation. As actin-
binding affinity of ALC-1 is less than of VLC-1, improved
cardiac function in ventricular tissue expressing ALC-1 may
be explained by weakening of the interaction between
actin and MLC-1. However, although up-regulation of ALC-1
may represent a compensatory mechanism to improve
cardiac function, it is not a consistent protein alteration in
ischaemic and idiopathic cardiomyopathy (IDCM), as large
individual differences exist.
29,30,32
Alternatively, addition of the N-terminal region of MLC-1
may be used to promote sarcomeric function. Cardiac func-
tion may be enhanced by disruption of the actin
MLC-1
interaction by additional expression of N-terminal MLC-1
fragments, which competitively bind to actin.
29,31
Another
explanation for MLC-1 induced increase in function was
given by Rarick et al.
33
who reported increased myofibrillar
MgATPase activity at submaximal [Ca
2þ
] upon addition of an
N-terminal VLC-1 fragment. These authors suggested that
the MLC-1 N-terminal peptide directly affects protein inter-
actions and exerts an inotropic effect via cooperative mech-
anisms, which activate the entire thin filament. Hence, the
N-terminus of MLC-1 seems to exert a beneficial effect on
sarcomeric function as increased systolic force generation
and rates of contraction and relaxation were observed in
Figure 1 Schematic representation of the cardiac sarcomere during diastole
(A) and systole (B). (A) The thin filament is composed of actin, tropomyosin,
and the troponin complex (composed of troponin T, cTnT; troponin C, cTnC;
and troponin I, cTnI). The thick filament is composed of myosin, an asym-
metric dimer composed of a globular head portion (S1), a hinged stalk
region (S2), and a rod section. The S1 portion of myosin contains both the
ATP hydrolysis domain and the actin-binding domain. Each myosin head is
associated with a pair of myosin light chains, which consists of an essential
light chain (MLC-1) and a regulatory light chain (MLC-2). MyBP-C, myosin
binding protein C. (B) Calcium binding to cTnC induces a conformational
rearrangement in the troponin
tropomyosin complex. Movement of tropo-
myosin exposes a myosin-binding site on actin allowing cross-bridge formation
to take place. This results in force development and/or shortening of the
sarcomere (Modified from de Tombe, J Biomech 2003;36:721
730, with
permission).
N. Hamdani et al.650
by guest on June 5, 2013http://cardiovascres.oxfordjournals.org/Downloaded from
hearts from transgenic rats harbouring minigenes encoding
the N-terminal domain of MLC-1.
34
Thus, although endogen-
ous heterogeneous expression of MLCs may be of minor rel-
evance in failing human myocardium, up-regulation of the
MLC-1 N-terminal fragment might provide a therapeutic
tool to enhance cardiac performance.
2.3 Troponin T
Anderson et al.
35
have proposed that re-expression of foetal
troponin T (cTnT) may also contribute to the reduced ATPase
activity in human heart failure. A significant inverse nega-
tive relationship was found between (re-)expression of
foetal cTnTand myofibrillar ATPase activity in human ventri-
cular tissue from normal and end-stage failing hearts.
However, this observation has not been confirmed by
others, and similar to the expression of a-MHC, the
expression levels of foetal cTnT is variable among individ-
uals.
36
38
While Anderson et al.
35
observed re-expression
of foetal cTnT in all end-stage failing human hearts, Solaro
et al.
36
only observed this foetal cTnT isoform in one out
of 10 failing heart samples. Similarly, we have observed
foetal cTnT in only one out of 24 patients with end-stage
heart failure.
38
Mesnard-Rouiller et al.
37
found expression
of foetal cTnT in half of the failing ventricles and suggested
that re-expression of foetal cTnT isoforms is not a common
characteristic of heart failure and most likely depends on
other factors such as intensity and duration of the elevation
of wall stress.
The consequences of foetal cTnT on sarcomeric contrac-
tion were studied upon exchange of endogenous cTnT with
foetal cTnT in rat cardiomyocytes.
39
No differences were
found in myofilament force development at maximal and
submaximal Ca
2þ
concentrations at a sarcomere length of
2.2 mm between cardiomyocytes exchanged with troponin
complex containing adult or foetal cTnT.
39
Akella et al.
40
observed a decrease in Ca
2þ
-responsiveness at low
(1.9 mm), but not at high (2.4 mm) sarcomere length in
skinned cardiac trabeculae from diabetic rats which
coincided with alterations in cTnT composition. More
recently, it was shown by Gomes et al.
41
that foetal
cTnT modulates Ca
2þ
sensitivity in the presence of foetal
(skeletal) TnI. Hence, the effect of foetal cTnT on sarco-
meric function seems to be dependent on sarcomere
length and protein background in the heart.
In conclusion, most isoform changes might be an intrinsic
part of the so-called ‘fetal’ (hypertrophic) program yet their
expression appears to be highly variable within human ven-
tricular tissue. Minor shifts in MHC and MLC composition do
not explain sarcomeric dysfunction in heart failure, but
are of compensatory nature.
3. Proteolysis and sarcomeric dysfunction
3.1 Troponin I
There is ample evidence that proteolytic activity is
enhanced after an acute ischaemic insult. Degradation pro-
ducts of several sarcomeric proteins
42
47
have been
observed, which may subsequently induce sarcomeric
dysfunction. One of the main proteins thought to be res-
ponsible for impaired cardiac function upon ischaemia/
reperfusion is cardiac troponin I (cTnI) (Figure 1).
42
45
In
rodents, McDonough et al.
44
showed that with moderate
ischaemia/reperfusion, cTnI is cleaved at its C-terminus,
which results in a truncated cTnI product (cTnI
1
193
,in
human cTnI
1
192
). More recently, it was postulated that
degradation of cTnI might also impair cardiomyocyte func-
tion and contribute to reduced pump function in heart
failure.
48
Myocardial infarction in pigs induced a reduction
in the maximal force generating capacity of single permea-
bilized cardiomyocytes isolated from remodelled non-
infarcted left ventricular tissue, in which minor degradation
of cTnI (4%) was observed.
49
In addition, independent of
ischaemia, cTnI degradation has been demonstrated in
human myocardium from coronary artery disease patients
with different degrees of heart failure.
45,50,51
To investigate
if truncated cTnI may contribute to depressed cardiac pump
function in human ischaemic cardiomyopathy and heart
failure, we recently investigated the direct functional
effects of cTnI
1
192
in human cardiomyocytes. Force
measurements were performed in non-failing human cardio-
myocytes permeabilized with Triton-X 100 and exchanged
with troponin complex containing either full length
(cTnI
FL
) or truncated cTnI.
52
Surprisingly, truncated cTnI
did not significantly alter maximal force development
(Figure 2A). Likewise, passive force was not different
between cells containing cTnI
FL
and cTnI
1
192
. However,
myofilament Ca
2þ
-sensitivity was significantly higher in
cTnI
1
192
exchanged preparations compared with cTnI
FL
cells (Figure 2B). This implicates that in humans truncation
of cTnI may limit relaxation of the heart muscle, while sys-
tolic function would benefit from the increase in Ca
2þ
-
responsiveness of the myofilaments.
4. Role of protein phosphorylation in
sarcomeric dysfunction
4.1 Protein kinase A-mediated phosphorylation
Not only truncation of cTnI, but also its phosphorylation
status is a prominent determinant of sarcomeric function,
both in health and disease. Upon b-adrenergic stimulation,
protein kinase A (PKA)-mediated cTnI phosphorylation at
serines 23/24 is associated with a decrease in myofilament
Ca
2þ
sensitivity
32,53,54
and contributes to an acceleration
of cardiac relaxation.
55,56
Since b-adrenergic signalling is
reduced in heart failure due to down-regulation and desen-
sitization of b-adrenoceptors,
57
59
PKA-mediated cTnI phos-
phorylation might be less pronounced in failing myocardium.
In agreement with reduced b-adrenergic signalling, reduced
phosphorylation levels of cTnI have been reported in failing
human myocardium compared with non-failing donor
hearts.
60
62
More specifically, phosphorylation at PKA sites
23/24 was significantly reduced in end-stage failing com-
pared with donor human myocardium.
61,62
At the functional
level, reduced PKA-mediated cTnI phosphorylation would
result in an increase in Ca
2þ
sensitivity of the myofilaments,
as was observed in single permeabilized human cardiomyo-
cytes isolated from end-stage failing hearts.
32,54,61,63
In
comparison with cells from non-failing donor myocardium,
Ca
2þ
sensitivity was significantly higher in cardiomyocytes
from patients with idiopathic or ischaemic cardiomyopathy
(Figure 3A). This sarcomeric defect was normalized upon
treatment of cardiomyocytes with the catalytic subunit of
PKA (Figure 3B), as the reduction in pCa
50
was larger in car-
diomyocytes from failing compared with donor hearts.
32
Sarcomeric dysfunction in heart failure 651
by guest on June 5, 2013http://cardiovascres.oxfordjournals.org/Downloaded from
Recently, Messer et al.
62
have shown that altered cTnI phos-
phorylation most likely underlies the increased Ca
2þ
-
responsiveness as isolated thin filaments from failing
human hearts displayed higher Ca
2þ
-responsiveness com-
pared with filaments from donor myocardium in an in vitro
motility assay. One should be careful when using non-failing
donor myocardium as ‘normal’, because of the high blood
catecholamine levels at the time of tissue procurement.
The high level of cTnI phosphorylation and relatively small
effect of PKA on myofilament Ca
2þ
-responsiveness might
reflect over-stimulation of the b-adrenergic pathway in
donors and thereby augment the difference between
healthy and failing samples. However, a similar increase
in myofilament Ca
2þ
-responsiveness has been observed in
several animal models of heart failure.
49,64
66
To minimize variable receptor stimulation at the time of
biopsy procurement, we recently conducted a series of
experiments on single cardiomyocytes from pigs with a myo-
cardial infarction or sham-operated animals isolated from
transmural needle biopsies, which were instantly frozen in
liquid nitrogen. Biopsies were taken from remote left ventri-
cular tissue 3 weeks after myocardial infarction induced by
ligation of the left circumflex coronary artery or from
sham-operated animals. Consistent with previous obser-
vations (Figure 4),
49
Ca
2þ
-responsiveness was significantly
higher in cells from infarct compared with sham animals,
while the shift upon PKA was smaller in sham than in post-
infarct remodelled myocardium (unpublished data). These
data clearly show that alterations in b-adrenergic signalling
and the concomitant reduction in PKA-mediated cTnI phos-
phorylation impair sarcomeric function. The increased
Ca
2þ
sensitivity of the myofilaments might contribute to
diastolic dysfunction via impaired relaxation of failing
myocardium.
4.2 Protein kinase C and D
Apart from the b-adrenergic pathway, other signalling
routes might be involved in the alterations in phosphoryl-
ation and function of sarcomeric proteins in heart failure.
Figure 2 Isometric force was measured in single permeabilized human cardiomyocytes upon activation in solutions containing maximal (pCa 4.5) and submax-
imal Ca
2þ
concentrations (pCa 5 to 6) and in relaxing solution (pCa 9) to determine maximal (F
max
) and passive (F
pas
) force development (A) and Ca
2þ
sensitivity of
force development (B). Force at submaximal activation was normalized to the force obtained during maximal activation. (A) The maximal force generating
capacity (F
max
) and F
pas
did not differ between cardiomyocytes containing truncated cTnI (cTn
1
192
) or full-length cTnI (cTn
FL
). (B)Ca
2þ
sensitivity increased
in cTn
1
192
cardiomyocytes compared with cTn
FL
exchanged cells (From Narolska et al.Circ Res 2006;99:1012
1020: Figures printed with permission).
Figure 3 (A)Ca
2þ
sensitivity was significantly higher in cardiomyocytes isolated from end-stage failing (n¼10) compared with donor (n¼6) hearts. (B) Incu-
bation of cells with the catalytic subunit of PKA reduced pCa
50
(DpCa
50
: 0.20 +0.01 and 0.03 +0.01 in failing and donor cells, respectively) and abolished the
difference between both groups (Modified from van der Velden et al.Cardiovasc Res 2003;57:37
47, with permission).
N. Hamdani et al.652
by guest on June 5, 2013http://cardiovascres.oxfordjournals.org/Downloaded from
Noteworthy, overall phosphorylation status of cTnI, deter-
mined on ProQ Diamond stained gels,
67
did not significantly
differ between sham-operated and MI pigs (unpublished
data), while the shift in Ca
2þ
sensitivity upon PKA treatment
was larger in infarct compared with sham animals (Figure
4B). This implies that, whereas PKA-mediated cTnI phos-
phorylation is down-regulated in infarct animals, cTnI phos-
phorylation by other kinases should be increased. One of the
most likely candidates is protein kinase C (PKC). Its activity
and expression levels are increased in heart failure
66,68
71
and both cTnI and cTnT contain specific PKC phosphorylata-
ble sites (i.e. serine 43/45 and threonine 144 in cTnI and
serine 201 and threonine 197/206/287 in cTnT).
72
74
In
addition to PKC, an increase in protein kinase D (PKD) has
been reported in heart failure.
75
PKD can be activated
upon phosphorylation by PKC, and thus may act down-
stream of PKC, or it also may be directly activated upon
receptor stimulation by, e.g. endothelin 1.
76
It has been
shown that PKD is able to reduce myofilament Ca
2þ
sensi-
tivity via phosphorylation of PKA-sites (serine 23/24) in
cTnI.
77
Consequently, the regulatory window of PKC and
PKD in sarcomeric function might be widened in diseased
myocardium.
The functional consequences of PKC-mediated protein
phosphorylation have been investigated in rodent models
and indicated a central role for cTnI and cTnT in reducing
maximal myofilament force development.
73,74,78
Apart
from its effect on maximal force, PKC has been shown to
reduce myofilament Ca
2þ
-sensitivity in rodent and human
myocardium.
61,71,73,74,78
The possible involvement of
PKC-mediated protein phosphorylation in sarcomeric func-
tion in failing myocardium was shown recently in rat
models of end-stage heart failure resulting from chronic
pressure overload (aortic banding) and myocardial infarc-
tion.
71
In both models increased expression and activation
of PKCawere observed in the late, but not in the early
phase of heart failure. Maximal force generating capacity
and Ca
2þ
-sensitivity of permeabilized cardiomyocytes were
significantly reduced in end-stage failing animals compared
with age-matched controls,
71,79
and both parameters
increased upon treatment with protein phosphatase 1
(PP-1). In contrast, PKCadid not significantly alter cardio-
myocyte function of failing cardiomyocytes, while it
reduced both maximal force and its Ca
2þ
sensitivity in
cells from the control group. In a previous study, the same
group performed experiments where in failing cardiomyo-
cytes the endogenous Tn-complex was exchanged by unpho-
sphorylated troponin complex, while control cells were
exchanged with troponin complex extracted from failing
hearts.
79
Upon exchange, Ca
2þ
sensitivity of failing cardio-
myocytes was restored towards the value observed in con-
trols, while failing troponin complex induced a significant
reduction in Ca
2þ
sensitivity in control cells. However, tro-
ponin exchange did not affect maximal tension, indicating
that PKC-mediated phosphorylation of troponin is not
involved in the reduced force generating capacity. Overall,
the data confirm that altered myofilament Ca
2þ
sensitivity
can be attributed to altered troponin phosphorylation,
while changes in maximal force generating capacity most
likely rely on the permissive action of other sarcomeric
proteins.
Whether PKC- and PKD-mediated phosphorylation and a
concurrent reduction in Ca
2þ
-responsiveness is detrimental
for cardiac function or represents an alternative mechanism
to preserve positive lusitropy during exercise and compen-
sates for reduced PKA-mediated cardiac relaxation requires
further investigation.
5. Sarcomeric dysfunction in heart failure
5.1 Increased vs. decreased Ca
21
sensitivity
Opposite to the increased myofilament Ca
2þ
sensitivity
observed in human end-stage failing myocardium
(Figure 3)
32,54,61,63
and in several animal models of cardiac
disease (Figure 4),
49,64
66
a decrease in myofilament Ca
2þ
-
sensitivity was reported in rodent models of heart failure
resulting from chronic pressure overload (aortic banding)
and myocardial infarction.
71,79
One possible explanation
for these contrasting observations might be the level of neu-
rohumoral stimulation present at the time of tissue procure-
ment. An intricate balance exists between kinase and
phosphatase activities within the cardiomyocyte as was
shown recently by Braz et al.
68
They reported that both
PKA and PKC may alter phosphorylation status of proteins
indirectly via phosphorylation of protein phosphatase
inhibitor-1 (I-1). Opposite to PKA, which suppresses PP-1
activity,
80
PKC enhances PP-1 activity via phosphorylation
of I-1. This illustrates the delicate balance between
kinases and phosphatases within a cell. An increase in PKC
Figure 4 Isometric force measurements were performed in single Triton-
permeabilized cardiomyocytes isolated from remote left ventricular tissue
from pigs 3 weeks after myocardial infarction. Maximal force development
(F
max
) was significantly lower in myocardial infarction compared with
sham-operated animals (A). Similar to the observations in human heart
failure, Ca
2þ
sensitivity was significantly higher (A) and the PKA-mediated
reduction in Ca
2þ
sensitivity was larger (B) in cells from myocardial infarction
compared with sham animals (Modified from van der Velden et al.Circ Res
2004;95:e85-e95, with permission).
Sarcomeric dysfunction in heart failure 653
by guest on June 5, 2013http://cardiovascres.oxfordjournals.org/Downloaded from
and a decrease in PKA-mediated phosphorylation of I-1
would enhance PP-1 activity
81
and thereby induce hypopho-
sphorylation of sarcomeric proteins.
Apart from differences in neurohumoral status when
tissue is retrieved from the heart, diverse alterations in
the signalling pathways known to alter sarcomeric protein
phosphorylation upon neurohumoral stimulation most likely
underlie diverse functional properties of the sarcomeres.
Already in 1991, Bristow et al.
82
have shown different
alterations in the b-adrenergic pathway in hearts with
ischaemic heart disease (ISHD) and IDCM. Analysis of sarco-
meric protein phosphorylation on ProQ Diamond stained
gradient gels (Figure 5A)
67
revealed significant differences
between left ventricular myocardial tissue from end-stage
failing patients with IDCM and ISHD (Figure 5B). Phosphoryl-
ation of cTnI was significantly higher in non-failing donor
compared with end-stage failing myocardium. In addition,
myosin binding protein C, which is phosphorylated upon
b-adrenergic stimulation is lower in failing compared with
donor hearts.
67,83
Noteworthy, MLC-2 phosphorylation was
significantly higher in ISHD compared with donor and IDCM
myocardium, and statistical analysis revealed significant
different phosphorylation of cTnI between ISHD and IDCM
samples. In line with a higher level of cTnI phosphorylation
in ISHD samples, myofilament Ca
2þ
sensitivity was signifi-
cantly lower in ISHD compared with IDCM myocardium.
32,54
These data provide evidence that diverse alterations in
sarcomeric protein composition and function in failing
hearts are related to underlying cause or phenotype.
5.2 Reduction in maximal force generating capacity
Reduced maximal force has been observed in diverse models
of cardiomyopathy (Figure 4A).
49,65,79,84
In rat with
pressure-overload and infarction-induced cardiomyopathy,
the reduction in F
max
amounted to 35 and 42%, respect-
ively.
79
As reduced F
max
was only partly reversed by PP-1
(15%),
71
and as described earlier, may not directly involve
the troponin complex,
79
alternative signalling routes, and
other sarcomeric proteins may be of relevance. Moreover,
depressed cardiomyocyte force development was also
observed in enzymatically isolated preparations from
failing rat hearts,
84
in which the isolation procedure most
likely reduced phosphorylation status of most sarcomeric
proteins. Therefore, part of the reduction in maximal
force might at least in part be related to altered isoform
composition and/or proteolysis of sarcomeric proteins. A
recent study in transgenic mice by Vahebi et al.,
85
in
which p38 MAPK (mitogen activated protein kinase) was
constitutively active in the heart, revealed a possible role
for tropomyosin dephosphorylation in the depression of
maximal force of the sarcomeres. Activation of p38 MAPK,
as occurs in pressure overload-induced hypertrophy, has
been shown to exert a negative effect on cardiomyocyte
contractility without altering Ca
2þ
-handling.
86
The study in
transgenic mice
85
indicated that apart from its role in remo-
delling and apoptosis, activated p38 MAPK leads to sarco-
meric dysfunction, possibly via activation of phosphatases
and a subsequent dephosphorylation of tropomyosin. The
level of tropomyosin phosphorylation appears to be species-
dependent, being relatively high in mice
85
and lower in
human myocardium (Figure 5A). However, similar to
isoform changes in MHC, small changes in phosphorylation
may exert a significant effect on sarcomeric function.
Therefore, the (patho)physiological role of tropomyosin
phosphorylation for sarcomeric function requires further
investigation.
In conclusion, depressed force development cannot be
explained by a single protein alteration, though seems to
be the result of complex interactions between various sarco-
meric proteins.
5.3 Increased cardiomyocyte stiffening
Subtle, though functionally important changes in protein
phosphorylation, induced by kinases and phosphatases
other than PKA, may have been obscured in failing human
myocardium in comparison with donor hearts. Separation
of patients with heart failure into subgroups, based on
severity, cause or phenotype, represents a powerful
approach to reveal the causes and functional implications
of alterations in sarcomeric function in human heart
failure. Comparison of patients with diastolic (DHF) and sys-
tolic heart failure (SHF) revealed an increased passive force
Figure 5 (A) Human heart samples (D, donor and IDCM, idiopathic dilated cardiomyopathy; 20 mg/lane) separated on 4
15% gradient gels. Phosphorylation of
myosin binding protein C (MyBP-C), desmin, troponin T (cTnT), troponin I (cTnI), and myosin light chain 2 (MLC-2) was determined with Pro-Q Diamond stain. (B)
Bar graph to illustrate differences in sarcomeric protein phosphorylation between human donor hearts and failing myocardium (ISHD, ischaemic heart failure).
*P,0.05 in one-way ANOVA. n, number of hearts [Modified from Zaremba et al.Proteomics Clin Appl 2007;1:1285
1290, with permission].
N. Hamdani et al.654
by guest on June 5, 2013http://cardiovascres.oxfordjournals.org/Downloaded from
development in cardiomyocytes from DHF compared with
SHF patients (Figure 6).
87
A significant positive correlation
was present between in vivo left ventricular end-diastolic
pressure (LVEDP) and F
pas
,
88
indicating that cardiomyocyte
stiffening contributes to high filling pressures in DHF.
Increased cardiomyocyte passive force was corrected to
values observed in hearts with preserved ejection fraction
and normal LVEDP
88
upon incubation with PKA (Figure 6),
indicative for hypophosphorylation of sarcomeric proteins.
The hypophosphorylated sarcomeric protein, possibly
titin,
89,90
could be a specific myocardial target for drug
therapy to lower LVEDP in DHF.
6. Future perspectives
The question if depressed cardiomyocyte contractility is
involved in heart failure has been positively answered.
Overall, there seems to be general consensus that sarco-
meric dysfunction in heart failure results from altered
protein phosphorylation, which is the result of complex
changes in kinase and phosphatase expression and activity.
The balance between kinases and phosphatases in the cardi-
omyocyte are humoral and heart rate-dependent and as a
consequence the activities of kinases and phosphatases
vary in time. Apart from temporal changes, spatial
changes occur, as complex interactions have been shown
between kinases and phosphatases regulating calcium
homeostasis within cardiomyocytes.
91,92
Such complex sig-
nalling may also apply to the myofilaments. Moreover,
within the complex pattern of sarcomeric protein phos-
phorylation each protein and its phosphorylation status
influences the behaviour of other sarcomeric proteins.
93,94
As sarcomeric function most likely reflects differences in
phosphorylation status and depend on the neurohumoral
status and heart rate at the time of tissue procurement,
investigation of the functional properties of the sarcomeres
should be performed in cardiac tissue, which is obtained
under standardized conditions. The use of catheter biopsies
has proved to be a major leap forward in unravelling
sarcomeric dysfunction in human myocardium.
87,88
Linkage
of in vivo haemodynamic data with cardiomyocyte force
measurements revealed that stiffening of the sarcomeres
contributes to impaired filling of the heart in DHF patients.
88
To obtain insight in dynamic signalling cardiac samples could
be retrieved upon receptor stimulation.
67
This approach
allows determination of the direct relation between func-
tional, structural, and protein characteristics at the cellular
level with in vivo haemodynamics measured at the time of
tissue harvesting.
The sarcomeric proteome will be even more complex than
described in the present review, since other signalling
routes, apart from the b-adrenergic pathway, may be trig-
gered under pathological conditions and affect sarcomeric
function.
68,75,76,86,95,96
Only recently, Yuan et al.
97
discov-
ered novel phosphorylation sites within the N-terminus of
MyBP-C, which were differentially phosphorylated upon
stunning in canine and rat myocardium. Apart from phos-
phorylation, post-translational modifications resulting from
oxidative stress might impair sarcomeric function. Moreover,
mutant sarcomeric proteins as found in inherited cardiomyo-
pathies
98,99
further complicate analysis of causality between
protein alterations and function of the sarcomere. Over the
past years knowledge on mutated sarcomeric proteins
present in cardiomyopathies increased swiftly. However,
the exact consequences of these mutations on cardiomyo-
cyte function in human cardiac tissue are still unclear and
knowledge concerning additional (mal)adaptive changes in
sarcomeric proteins is scarce. Hence, it remains to be eluci-
dated if and to what extent altered sarcomeric protein
expression and/or post-translational changes impair sarco-
meric function in inherited cardiomyopathies.
The combination of sarcomeric force measurements with
proteomic analysis (i.e. functional proteomics) will reveal
(novel) post-translational modifications involved in cardio-
myocyte dysfunction in heart failure. The use of transgenic
animal models and protein exchange experiments in
cardiac preparations are required to define the specific
role of post-translational protein modifications and mutant
sarcomeric proteins in cardiac function.
7. Clinical implications
The recently obtained data in human myocardium (Figures 5
and 6) indicate that divergent disturbance of receptor-
signalling cascades depend on underlying cause and pheno-
type. Diverse alterations in signalling pathways might alter
the responsiveness of patients to drug therapy and therefore
the current strategy of treating heart failure should be
re-evaluated. Large randomized, double-blind, placebo-
controlled multicentre trials have shown that treatment of
heart failure patients (classified according to the New York
Heart Association into class II
IV) with neurohumoral recep-
tor blockers, such as ACE-inhibitors and beta-blockers,
reduces both morbidity and mortality. However, it remains
to be investigated if and to what extent reversal of sarco-
meric dysfunction contributes to the beneficial effects of
beta-blocker and ACE-inhibitor therapy in different patient
groups.
Interestingly, exercise in mice with a myocardial infarc-
tion reversed depressed sarcomeric function to values
observed in controls.
65
The beneficial effects appeared to
be the result of improved b-adrenergic signalling. Future
studies should investigate if the combination of currently
used neurohumoral blockers with exercise yield added
benefit. Novel therapy may include drugs targeted to
mediators down-stream of the adrenergic and angiotensin
receptors. Cardiac performance may be improved by target-
ing a specific myofilament protein
34,100
to directly modulate
Figure 6 Cardiomyocyte stiffening in human heart failure. Cardiomyocyte
passive force (F
pas
) was significantly higher in DHF than in SHF (*P,0.05).
PKA treatment reduced F
pas
in SHF and DHF (
#
P,0.05 in paired t-test)
(From van Heerebeek et al. Circulation 2006;113:1966
1973, with
permission).
Sarcomeric dysfunction in heart failure 655
by guest on June 5, 2013http://cardiovascres.oxfordjournals.org/Downloaded from
sarcomeric function. Further exploration of the complex sig-
nalling routes underlying defects in sarcomeric function is
required in order to develop more precise, individualized
therapy in heart failure patients.
Conflict of interest: none declared.
Funding
Netherlands Organisation for Scientific Research (VENI grant
916.36.013 to J.v.d.V.); Netherlands Heart Foundation
(grant 2000T042 to D.J.D., grant 2005B220 to J.v.d.V.).
References
1. De Tombe PP. Altered contractile function in heart failure. Cardiovasc
Res 1998;37:367
380.
2. Houser SR, Margulies KB. Is depressed myocyte contractility centrally
involved in heart failure? Circ Res 2003;92:350
358.
3. Nakayama H, Chen X, Baines CP, Klevitsky R, Zhang X, Zhang H et al.
Ca
2þ
- and mitochondrial-dependent cardiomyocyte necrosis as a
primary mediator of heart failure. J Clin Invest 2007;117:2431
2444.
4. Jane-Lise S, Corda S, Chassagne C, Rappaport L. The extracellular
matrix and the cytoskeleton in heart hypertrophy and failure. Heart
Fail Rev 2000;5:239
250.
5. De Tombe PP. Myofilaments: mechanics regulation. J Biomech 2003;36:
721
730.
6. Lompre AM, Schwartz K, d’Albis A, Lacombe G, Van Thiem N,
Swynghedauw B. Myosin isoenzyme redistribution in chronic heart
overload. Nature 1979;282:105
107.
7. Mercadier JJ, Lompre AM, Wisnewsky C, Samuel JL, Bercovici J,
Swynghedauw B et al. Myosin isoenzyme changes in several models of
rat cardiac hypertrophy. Circ Res 1981;49:525
532.
8. Miyata S, Minobe W, Bristow MR, Leinwand LA. Myosin heavy chain
isoform expression in the failing and nonfailing human heart. Circ Res
2000;86:386
390.
9. Lompre AM, Mercadier JJ, Wisnewsky C, Bouveret P, Pantaloni C,
d’Albis A et al. Species- and age-dependent changes in the relative
amounts of cardiac myosin isoenzymes in mammals. Develop Biol
1981;84:286
290.
10. Mercadier JJ, Bouveret P, Gorza L, Schiaffino S, Clark WA, Zak R et al.
Myosin isoenzymes in normal and hypertrophied human ventricular
myocardium. Circ Res 1983;53:52
62.
11. Narolska NA, van Loon RB, Boontje NM, Zaremba R, Penas SE, Russell J
et al. Myocardial contraction is 5-fold more economical in ventricular
than in atrial human tissue. Cardiovasc Res 2005;65:221
229.
12. Narolska NA, Eiras S, van Loon RB, Boontje NM, Zaremba R,
Spiegelenberg SR et al. Myosin heavy chain composition and the
economy of contraction in healthy and diseased human myocardium.
J Muscle Res Cell Motil 2005;26:39
48.
13. Barany M. ATPase activity of myosin correlated with speed of muscle
shortening. J Gen Physiol 1967;50(Suppl.):197
218.
14. Pope B, Hoh JF, Weeds A. The ATPase activities of rat cardiac myosin
isoenzymes. FEBS Lett 1980;118:205
208.
15. Harris DE, Work SS, Wright RK, Alpert NR, Warshaw DM. Smooth, cardiac
and skeletal muscle myosin force and motion generation assessed by
cross-bridge mechanical interactions in vitro. J Muscle Res Cell Motil
1994;15:11
19.
16. Van der Velden J, Moorman AF, Stienen GJM. Age-dependent changes in
myosin composition correlate with enhanced economy of contraction in
guinea-pig hearts. J Physiol 1998;507:497
510.
17. Rundell VL, Manaves V, Martin AF, de Tombe PP. Impact of beta-myosin
heavy chain isoform expression on cross-bridge cycling kinetics. Am J
Physiol Heart Circ Physiol 2005;288:H896
H903.
18. Korte FS, Herron TJ, Rovetto MJ, McDonald KS. Power output is linearly
related to MyHC content in rat skinned myocytes and isolated working
hearts. Am J Physiol Heart Circ Physiol 2005;289:H801
H812.
19. Rundell VL, Geenen DL, Buttrick PM, de Tombe PP. Depressed cardiac
tension cost in experimental diabetes is due to altered myosin heavy
chain isoform expression. Am J Physiol Heart Circ Physiol 2004;287:
H408
H413.
20. Alpert NR, Gordon MS. Myofibrillar adenosine triphosphatase activity in
congestive heart failure. Am J Physiol 1962;202:940
946.
21. Nguyen TT, Hayes E, Mulieri LA, Leavitt BJ, ter Keurs HE, Alpert NR et al.
Maximal actomyosin ATPase activity and in vitro myosin motility are
unaltered in human mitral regurgitation heart failure. Circ Res 1996;
79:222
226.
22. Noguchi T, Camp P Jr, Alix SL, Gorga JA, Begin KJ, Leavitt BJ et al.
Myosin from failing and non-failing human ventricles exhibit similar
contractile properties. J Mol Cell Cardiol 2003;35:91
97.
23. Pagani ED, Alousi AA, Grant AM, Older TM, Dziuban SW Jr, Allen PD.
Changes in myofibrillar content and Mg-ATPase activity in ventricular
tissues from patients with heart failure caused by coronary artery
disease, cardiomyopathy, or mitral valve insufficiency. Circ Res 1988;
63:380
385.
24. Reiser PJ, Portman MA, Ning XH, Schomisch MC. Human cardiac myosin
heavy chain isoforms in fetal and failing adult atria and ventricles. Am J
Physiol Heart Circ Physiol 2001;280:H1814
H1820.
25. Kuro-o M, Tsuchimochi H, Ueda S, Takaku F, Yazaki Y. Distribution of
cardiac myosin isozymes in human conduction system. Immunohisto-
chemical study using monoclonal antibodies. J Clin Invest 1986;77:
340
347.
26. Carnes CA, Geisbuhler TP, Reiser PJ. Age-dependent changes in contrac-
tion and regional myocardial myosin heavy chain isoform expression in
rats. J Appl Physiol 2004;97:446
453.
27. Herron TJ, McDonald KS. Small amounts of alpha-myosin heavy chain
isoform expression significantly increase power output of rat cardiac
myocyte fragments. Circ Res 2002;90:1150
1152.
28. Eiras S, Narolska NA, van Loon RB, Boontje NM, Zaremba R, Jimenez CR
et al. Alterations in contractile protein composition and function in
human atrial dilatation and atrial fibrillation. J Mol Cell Cardiol 2006;
41:467
477.
29. Morano I. Tuning the human heart molecular motors by myosin light
chains. J Mol Med 1999;77:544
555.
30. Ritter O, Luther HP, Haase H, Baltas LG, Baumann G, Schulte HD et al.
Expression of atrial myosin light chains but not alpha-myosin heavy
chains is correlated in vivo with increased ventricular function in
patients with hypertrophic obstructive cardiomyopathy. J Mol Med
1999;77:677
685.
31. Morano I, Ritter O, Bonz A, Timek T, Vahl CF, Michel G. Myosin light
chain-actin interaction regulates cardiac contractility. Circ Res 1995;
76:720
725.
32. Van der Velden J, Papp Z, Zaremba R, Boontje NM, de Jong JW, Owen VJ
et al. Increased Ca
2þ
sensitivity of the contractile apparatus in
end-stage human heart failure results from altered phosphorylation of
contractile proteins. Cardiovasc Res 2003;57:37
47.
33. Rarick HM, Opgenorth TJ, von Geldern TW, Wu-Wong JR, Solaro RJ. An
essential myosin light chain peptide induces supramaximal stimulation
of cardiac myofibrillar ATPase activity. J Biol Chem 1996;271:
27039
27043.
34. Haase H, Dobbernack G, Tunnemann G, Karczewski P, Cardoso C,
Petzhold D et al. Minigenes encoding N-terminal domains of human
cardiac myosin light chain-1 improve heart function of transgenic rats.
FASEB J 2006;20:865
873.
35. Anderson PA, Malouf NN, Oakeley AE, Pagani ED, Allen PD. Troponin T
isoform expression in the normal and failing human left ventricle: a cor-
relation with myofibrillar ATPase activity. Basic Res Cardiol 1992;
87(Suppl. 1):117
127.
36. Solaro RJ, Powers FM, Gao L, Gwathmey JK. Control of myofilament
activation in heart failure. Circulation 1993;87(Suppl. VII):VII38
VII43.
37. Mesnard-Rouiller L, Mercadier JJ, Butler-Browne G, Heimburger M,
Logeart D, Allen PD et al. Troponin T mRNA and protein isoforms in
the human left ventricle: pattern of expression in failing and control
hearts. J Mol Cell Cardiol 1997;29:3043
3055.
38. Van der Velden J, Klein LJ, van der Bijl M, Huybregts MA, Stooker W,
Witkop J et al. Isometric tension development and its calcium sensitivity
in skinned myocyte-sized preparations from different regions of the
human heart. Cardiovasc Res 1999;42:706
719.
39. Van der Velden J, Chandra M, Stienen GJM, Solaro RJ, de Tombe PP.
Exchange of troponin T in single cardiomyocytes from rat. (Abstract).
J Muscle Res Cell Motil 2000;21:803.
40. Akella AB, Ding XL, Cheng R, Gulati J. Diminished Ca
2þ
sensitivity of
skinned cardiac muscle contractility coincident with troponin T-band
shifts in the diabetic rat. Circ Res 1995;76:600
606.
41. Gomes AV, Venkatraman G, Davis JP, Tikunova SB, Engel P, Solaro RJ
et al. Cardiac troponin T isoforms affect the Ca
2þ
sensitivity of force
development in the presence of slow skeletal troponin I: insights into
the role of troponin T isoforms in the fetal heart. J Biol Chem 2004;
279:49579
49587.
N. Hamdani et al.656
by guest on June 5, 2013http://cardiovascres.oxfordjournals.org/Downloaded from
42. Gao WD, Atar D, Liu Y, Perez NG, Murphy AM, Marban E. Role of troponin
I proteolysis in the pathogenesis of stunned myocardium. Circ Res 1997;
80:393
399.
43. Van Eyk JE, Powers F, Law W, Larue C, Hodges RS, Solaro RJ. Breakdown
and release of myofilament proteins during ischemia and ischemia/
reperfusion in rat hearts: identification of degradation products and
effects on the pCa-force relation. Circ Res 1998;82:261
271.
44. McDonough JL, Arrell DK, Van Eyk JE. Troponin I degradation and
covalent complex formation accompanies myocardial ischemia/reperfu-
sion injury. Circ Res 1999;84:9
20.
45. Murphy AM, Kogler H, Georgakopoulos D, McDonough JL, Kass DA, Van
Eyk JE et al. Transgenic mouse model of stunned myocardium. Science
2000;287:488
491.
46. Papp Z, van der Velden J, Stienen GJM. Calpain-I induced alterations in
the cytoskeletal structure and impaired mechanical properties of single
myocytes of rat heart. Cardiovasc Res 2000;45:981
993.
47. Decker RS, Decker ML, Kulikovskaya I, Nakamura S, Lee DC, Harris K
et al. Myosin-binding protein C phosphorylation, myofibril structure,
and contractile function during low-flow ischemia. Circulation 2005;
111:906
912.
48. Van der Laarse A. Hypothesis: troponin degradation is one of the factors
responsible for deterioration of left ventricular function in heart failure.
Cardiovasc Res 2002;56:8
14.
49. Van der Velden J, Merkus D, Klarenbeek BR, James AT, Boontje NM,
Dekkers DH et al. Alterations in myofilament function contribute to
left ventricular dysfunction in pigs early after myocardial infarction.
Circ Res 2004;95:e85
e95.
50. McDonough JL, Labugger R, Pickett W, Tse MY, MacKenzie S, Pang SC
et al. Cardiac troponin I is modified in the myocardium of bypass
patients. Circulation 2001;103:58
64.
51. Neagoe C, Kulke M, del Monte F, Gwathmey JK, de Tombe PP, Hajjar RJ
et al. Titin isoform switch in ischemic human heart disease. Circulation
2002;106:1333
1341.
52. Narolska NA, Piroddi N, Belus A, Boontje NM, Scellini B, Deppermann S
et al. Impaired diastolic function after exchange of endogenous tropo-
nin I with C-terminal truncated troponin I in human cardiac muscle.
Circ Res 2006;99:1012
1020.
53. Solaro RJ, Moir AJ, Perry SV. Phosphorylation of troponin I and the ino-
tropic effect of adrenaline in the perfused rabbit heart. Nature 1976;
262:615
617.
54. Wolff MR, Buck SH, Stoker SW, Greaser ML, Mentzer RM. Myofibrillar
calcium sensitivity of isometric tension is increased in human dilated
cardiomyopathies. J Clin Invest 1996;98:167
176.
55. Zhang R, Zhao J, Mandveno A, Potter JD. Cardiac troponin I phosphoryl-
ation increases the rate of cardiac muscle relaxation. Circ Res 1995;76:
1028
1035.
56. Metzger JM, Westfall MV. Covalent and noncovalent modification of thin
filament action. The essential role of troponin in cardiac muscle regu-
lation. Circ Res 2004;94:146
158.
57. Bristow MR, Ginsburg R, Fowler M, Minobe W, Rasmussen R, Zera P et al.
b
1
- and b
2
-adrenergic receptor subpopulations in normal and failing
human ventricular myocardium: coupling of both receptor subtypes to
muscle contraction and selective b
1
-receptor downregulation in heart
failure. Circ Res 1986;59:297
309.
58. Brodde OE, Schuler S, Kretsch R, Brinkmann M, Borst HG, Hetzer R et al.
Regional distribution of b-adrenoceptors in the human heart: coexis-
tence of functional b
1
- and b
2
-adrenoceptors in both atria and ventri-
cles in severe congestive cardiomyopathy. J Cardiovasc Pharmacol
1986;8:1235
1242.
59. Harding SE, Jones SM, Vescovo G, Del Monte F, Poole-Wilson PA. Reduced
contractile responses to forskolin and a cyclic AMP analogue in myocytes
from failing human ventricle. Eur J Pharmacol 1992;223:39
48.
60. Bodor GS, Oakeley AE, Allen PD, Crimmins DL, Ladenson JH,
Anderson PA. Troponin I phosphorylation in the normal and failing
adult human heart. Circulation 1997;96:1495
1500.
61. Van der Velden J, Narolska NA, Lamberts RR, Boontje NM, Borbely A,
Zaremba R et al. Functional effects of protein kinase C-mediated myo-
filament phosphorylation in human myocardium. Cardiovasc Res 2006;
69:876
887.
62. Messer AE, Jacques AM, Marston SB. Troponin phosphorylation and regu-
latory function in human heart muscle: dephosphorylation of Ser23/24
on troponin I could account for the contractile defect in end-stage
heart failure. J Mol Cell Cardiol 2007;42:247
259.
63. Van der Velden J, Klein LJ, Zaremba R, Boontje NM, Huybregts MA,
Stooker W et al. Effects of calcium, inorganic phosphate, and pH on
isometric force in single skinned cardiomyocytes from donor and
failing human hearts. Circulation 2001;104:1140
1146.
64. Wolff MR, Whitesell LF, Moss RL. Calcium sensitivity of isometric tension
is increased in canine experimental heart failure. Circ Res 1995;76:
781
789.
65. De Waard MC, van der Velden J, Bito V, Ozdemir S, Biesmans L,
Boontje NM et al. Early exercise training normalizes myofilament func-
tion and attenuates left ventricular pump dysfunction in mice with a
large myocardial infarction. Circ Res 2007;100:1079
1088.
66. Lamberts RR, Hamdani N, Soekhoe TW, Boontje NM, Zaremba R,
Walker LA et al. Frequency-dependent myofilament Ca
2þ
desensitisa-
tion in failing rat myocardium. J Physiol 2007;582:695
709.
67. Zaremba R, Merkus D, Hamdani N, Lamers JMJ, Paulus WJ, dos
Remedios C et al. Quantitative analysis of myofilament protein phos-
phorylation in small cardiac biopsies. Proteomics Clin Appl 2007;1:
1285
1290.
68. Braz JC, Gregory K, Pathak A, Zhao W, Sahin B, Klevitsky R et al. PKC-a
regulates cardiac contractility and propensity toward heart failure.
Nature 2004;10:248
254.
69. Bowling N, Walsh RA, Song G, Estridge T, Sandusky GE, Fouts RL et al.
Increased protein kinase C activity and expression of Ca
2þ
-sensitive
isoforms in the failing human heart. Circulation 1999;99:384
391.
70. Noguchi T, Hunlich M, Camp PC, Begin KJ, El-Zaru M, Patten R et al. Thin
filament-based modulation of contractile performance in human heart
failure. Circulation 2004;110:982
987.
71. Belin RJ, Sumandea MP, Allen EJ, Schoenfelt K, Wang H, John Solaro R
et al. Augmented protein kinase C-a-induced myofilament protein phos-
phorylation contributes to myofilament dysfunction in experimental
congestive heart failure. Circ Res 2007;101:195
204.
72. Noland TA, Raynor RL, Kuo JF. Identification of sites phosphorylated in
bovine cardiac troponin I and troponin T by protein kinase C and
comparative substrate activity of synthetic peptides containing the
phosphorylation sites. J Biol Chem 1989;264:20778
20785.
73. Burkart EM, Sumandea MP, Kobayashi T, Nili M, Martin AF, Homsher E
et al. Phosphorylation or glutamic acid substitution at protein kinase
C sites on cardiac troponin I differentially depress myofilament
tension and shortening velocity. J Biol Chem 2003;278:11265
11272.
74. Sumandea MP, Pyle WG, Kobayashi T, de Tombe PP, Solaro RJ. Identifi-
cation of a functionally critical protein kinase C phosphorylation
residue of cardiac troponin T. J Biol Chem 2003;278:35135
35144.
75. Bossuyt J, Wu X, Avkiran M, Martin JL, Pogwizd SM, Bers DM. CaMKII and
PKD overexpression seen in heart failure maintains the HDAC5 redistri-
bution from the nucleus to the cytosol. (Abstract). Circulation 2006;
114:396.
76. Cuello F, Bardswell SC, Haworth RS, Yin X, Lutz S, Wieland T et al.
Protein kinase D selectively targets cardiac troponin I and regulates
myofilament Ca
2þ
sensitivity in ventricular myocytes. Circ Res 2007;
100:864
873.
77. Haworth RS, Cuello F, Herron TJ, Franzen G, Kentish JC, Gautel M et al.
Protein kinase D is a novel mediator of cardiac troponin I phosphoryl-
ation and regulates myofilament function. Circ Res 2004;95:1091
1099.
78. Montgomery DE, Chandra M, Huang QQ, Jin JP, Solaro RJ. Transgenic
incorporation of skeletal TnT into cardiac myofilaments blunts
PKC-mediated depression of force. Am J Physiol 2001;260:
H1011
H1018.
79. Belin RJ, Sumandea MP, Kobayashi T, Walker LA, Rundell VL, Urboniene D
et al. Left ventricular myofilament dysfunction in rat experimental
hypertrophy and congestive heart failure. Am J Physiol Heart Circ
Physiol 2006;291:H2344
H2353.
80. Neumann J, Gupta RC, Schmitz W, Scholz H, Nairn AC, Watanabe AM.
Evidence for isoproterenol-induced phosphorylation of phosphatase
inhibitor-1 in the intact heart. Circ Res 1991;69:1450
1457.
81. Neumann J, Eschenhagen T, Jones LR, Linck B, Schmitz W, Scholz H
et al. Increased expression of cardiac phosphatases in patients with
end-stage heart failure. J Mol Cell Cardiol 1997;29:265
272.
82. Bristow MR, Anderson FL, Port JD, Skerl L, Hershberger RE, Larrabee P
et al. Differences in b-adrenergic neuroeffector mechanisms in ischemic
versus idiopathic dilated cardiomyopathy. Circulation 1991;84:
1024
1039.
83. El-Armouche A, Pohlmann L, Schlossarek S, Starbatty J, Yeh YH, Nattel S
et al. Increased phosphorylation levels of cardiac myosin-binding
protein-C in human and experimental heart failure. J Mol Cell Cardiol
2007;43:223
229.
84. Fan D, Wannenburg T, de Tombe PP. Decreased myocyte tension devel-
opment and calcium responsiveness in rat right ventricular pressure
overload. Circulation 1997;95:2312
2317.
Sarcomeric dysfunction in heart failure 657
by guest on June 5, 2013http://cardiovascres.oxfordjournals.org/Downloaded from
85. Vahebi S, Ota A, Li M, Warren CM, de Tombe PP, Wang Y et al. p38-MAPK
induced dephosphorylation of alpha-tropomyosin is associated with
depression of myocardial sarcomeric tension and ATPase activity. Circ
Res 2007;100:408
415.
86. Liao P, Wang SQ, Wang S, Zheng M, Zheng M, Zhang SJ et al. p38
Mitogen-activated protein kinase mediates a negative inotropic effect
in cardiac myocytes. Circ Res 2002;90:190
196.
87. Van Heerebeek L, Borbely A, Niessen HW, Bronzwaer JG, van der
Velden J, Stienen GJM et al. Myocardial structure and function differ
in systolic and diastolic heart failure. Circulation 2006;113:1966
1973.
88. Borbe
´ly A, van der Velden J, Papp Z, Bronzwaer JG, Edes I, Stienen GJM
et al. Cardiomyocyte stiffness in diastolic heart failure. Circulation
2005;111:774
781.
89. Yamasaki R, Wu Y, McNabb M, Greaser M, Labeit S, Granzier H. Protein
kinase A phosphorylates titin’s cardiac-specific N2B domain and
reduces passive tension in rat cardiac myocytes. Circ Res 2002;90:
1181
1188.
90. Kruger M, Linke WA. Protein kinase-A phosphorylates titin in human
heart muscle and reduces myofibrillar passive tension. J Muscle Res
Cell Motil 2006;27:435
444.
91. Wehrens XH, Lehnart SE, Reiken SR, Marks AR. Ca
2þ
/calmodulin-
dependent protein kinase II phosphorylation regulates the cardiac
ryanodine receptor. Circ Res 2004;94:e61
e70.
92. Huke S, Bers DM. Temporal dissociation of frequency-dependent accel-
eration of relaxation and protein phosphorylation by CaMKII. J Mol
Cell Cardiol 2007;42:590
599.
93. van der Velden J, Papp Z, Boontje NM, Zaremba R, de Jong JW,
Janssen PM et al. The effect of myosin light chain 2 dephosphorylation
on Ca
2þ
sensitivity of force is enhanced in failing human hearts.
Cardiovasc Res 2003;57:505
514.
94. Verduyn SC, Zaremba R, van der Velden J, Stienen GJ. Effects of con-
tractile protein phosphorylation on force development in permeabilized
rat cardiac myocytes. Basic Res Cardiol 2007;102:476
487.
95. Buscemi N, Foster DB, Neverova I, Van Eyk JE. p21-activated kinase
increases the calcium sensitivity of rat triton-skinned cardiac muscle
fiber bundles via a mechanism potentially involving novel phosphoryl-
ation of troponin I. Circ Res 2002;91:509
516.
96. Chen Y, Rajashree R, Liu Q, Hofmann P. Acute p38 MAPK activation
decreases force development in ventricular myocytes. Am J Physiol
Heart Circ Physiol 2003;285:H2578
H2586.
97. Yuan C, Guo Y, Ravi R, Przyklenk K, Shilkofski N, Diez R et al. Myosin
binding protein C is differentially phosphorylated upon myocardial stun-
ning in canine and rat hearts: evidence for novel phosphorylation sites.
Proteomics 2006;6:4176
4186.
98. Chang AN, Potter JD. Sarcomeric protein mutations in dilated cardio-
myopathy. Heart Fail Rev 2005;10:225
235.
99. Tardiff JC. Sarcomeric proteins and familial hypertrophic cardiomyopa-
thy: linking mutations in structural proteins to complex cardiovascular
phenotypes. Heart Fail Rev 2005;10:237
248.
100. Day SM, Westfall MV, Metzger JM. Tuning cardiac performance in
ischemic heart disease and failure by modulating myofilament function.
J Mol Med 2007;85:911
921.
N. Hamdani et al.658
by guest on June 5, 2013http://cardiovascres.oxfordjournals.org/Downloaded from
... Types of economic growth: (Hamdani, 2008(Hamdani, /2009 [37] . Economic growth can be classified into several types, including a) Natural economic growth: It is the growth that occurs because of the transition from a feudal society to a capitalist society over a historical period, and during that period many social changes occur, b) Planned economic growth: It is the growth that occurs because of comprehensive planning processes for a community's resources and needs, but its effectiveness is linked to the ability of planners, implementation and follow-up, and the interaction of community members with those plans. ...
... Types of economic growth: (Hamdani, 2008(Hamdani, /2009 [37] . Economic growth can be classified into several types, including a) Natural economic growth: It is the growth that occurs because of the transition from a feudal society to a capitalist society over a historical period, and during that period many social changes occur, b) Planned economic growth: It is the growth that occurs because of comprehensive planning processes for a community's resources and needs, but its effectiveness is linked to the ability of planners, implementation and follow-up, and the interaction of community members with those plans. ...
... Exploring the molecular details reveals that the two HF phenotypes exhibit unique patterns, with cardiac remodeling being a common feature characterized by changes in the phosphorylation of myofilament proteins, particularly regulatory proteins [75,90,91]. While there is a depth of understanding regarding these changes in HFrEF, insights into HFpEF remain relatively limited [75,90,91]. ...
... Exploring the molecular details reveals that the two HF phenotypes exhibit unique patterns, with cardiac remodeling being a common feature characterized by changes in the phosphorylation of myofilament proteins, particularly regulatory proteins [75,90,91]. While there is a depth of understanding regarding these changes in HFrEF, insights into HFpEF remain relatively limited [75,90,91]. For instance, the protein TnI, pivotal for myofilament Ca 2+ sensitivity, has been subject to varying results across studies. ...
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.
... At the cellular level, relaxation impairment is characterized by a decrease in the rate of relaxation of the sarcomeres. The relaxation behavior of sarcomeres, as basic contractile units of the heart 25 , is critical for effective diastolic behavior 26 . Relaxation impairment at the cellular level is often associated with abnormalities in calcium cycling, which is critical for regulating cardiac contraction and relaxation. ...
... Relaxation impairment at the cellular level is often associated with abnormalities in calcium cycling, which is critical for regulating cardiac contraction and relaxation. Impaired calcium cycling can result in elevated levels of intracellular calcium during diastole, which, in turn, can lead to delayed relaxation 26,27 . ...
... Journal of General Physiology development of diastolic pathology (Gilbert et al., 2020). However, based on studies employing either animal models or human samples, it has become evident that depressed myofilament function also contributes to the depressed cardiac function seen in both systolic and diastolic heart failure (de Tombe, 1998;Hamdani et al., 2008;van der Velden and de Tombe, 2014;van der Velden and Stienen, 2019). It should be noted that while the present study focused on female guinea pigs, it has been reported that AOB in male guinea pigs also results in cardiac hypertrophy and heart failure (Siri et al., 1989), a finding we confirmed in preliminary studies leading up to the current study. ...
... The initial factors leading to cardiac and associated myofilament dysfunction may be acquired, for example, pressure or volume cardiac overload or chronic myocardial ischemia (de Tombe, 1998;Hamdani et al., 2008;van der Velden and de Tombe, 2014;van der Velden and Stienen, 2019). More recently, it has also become evident that inherited factors, such as mutations in cardiac myofilament proteins, may be causal to myofilament dysfunction that, in turn, and only in some patients, lead to cardiac dysfunction and failure (van der Velden and Stienen, 2019). ...
Article
Full-text available
Cardiac hypertrophy is associated with diastolic heart failure (DHF), a syndrome in which systolic function is preserved but cardiac filling dynamics are depressed. The molecular mechanisms underlying DHF and the potential role of altered cross-bridge cycling are poorly understood. Accordingly, chronic pressure overload was induced by surgically banding the thoracic ascending aorta (AOB) in ∼400 g female Dunkin Hartley guinea pigs (AOB); Sham-operated age-matched animals served as controls. Guinea pigs were chosen to avoid the confounding impacts of altered myosin heavy chain (MHC) isoform expression seen in other small rodent models. In vivo cardiac function was assessed by echocardiography; cardiac hypertrophy was confirmed by morphometric analysis. AOB resulted in left ventricle (LV) hypertrophy and compromised diastolic function with normal systolic function. Biochemical analysis revealed exclusive expression of β-MHC isoform in both sham control and AOB LVs. Myofilament function was assessed in skinned multicellular preparations, skinned single myocyte fragments, and single myofibrils prepared from frozen (liquid N2) LVs. The rates of force-dependent ATP consumption (tension-cost) and force redevelopment (Ktr), as well as myofibril relaxation time (Timelin) were significantly blunted in AOB, indicating reduced cross-bridge cycling kinetics. Maximum Ca²⁺ activated force development was significantly reduced in AOB myocytes, while no change in myofilament Ca²⁺ sensitivity was observed. Our results indicate blunted cross-bridge cycle in a β-MHC small animal DHF model. Reduced cross-bridge cycling kinetics may contribute, at least in part, to the development of DHF in larger mammals, including humans.
... The oxidative stress can also disrupt the transmission of calcium-dependent contractile responses [54][55][56] . A highly oxidative environment seems to modulate phosphorylation in sarcomere proteins altering its functioning 57-59 . ...
Preprint
The renin-angiotensin system (RAS) plays a critical role in the regulation of the cardiovascular system. The Mas-related G protein receptor member D (MrgD) is the receptor of alamandine, and both are components of the RAS noncanonical arm. Alamandine/MrgD induces vasodilation, anti-inflammatory, anti-fibrotic and anti-oxidative effects. In contrast, Mrgd gene deletion leads to a remarkable dilated cardiomyopathy (DCM) in mice. Here, we aimed to investigate the molecular mechanisms of DCM triggered by the deletion of MrgD in the left ventricle and isolated ventricular cardiomyocytes from 8-12 weeks old mice using phosphoproteomics. Our findings revealed an increased oxidative stress not caused by angiotensin II/AT1 hyperactivation but instead due to the up-regulation of the monoamine oxidase B (MAO-B), leading to a higher catabolism of dopamine and epinephrine in the MrgD-KO cardiac tissues. The oxidative environment induced by MAO-B hyperactivation seems to be the cause of the observed alteration in ionic dynamics - altered Ca2+ transient and Na+/K+-ATPase activity - leading to altered resting membrane potential (RMP) and decreased contraction of MrgD-KO cardiomyocytes. In addition, cardiac Troponin-I phosphorylation, and Titin dephosphorylation seem to contribute to the contractile dysfunction observed in MrgD-KO. The treatment of cardiomyocytes from MrgD-KO mice with the MAO-B inhibitor Pargyline reverted the observed impaired contraction, corroborating the hypothesis that MAO-B hyperactivation is, at least partially, the cause of the failing heart observed in MrgD-KO mouse. The findings reported here provide important insights into the pathogenesis of heart failure and suggest a potential therapeutic target (MrgD activation) for managing failing hearts.
... There is ample evidence that HF degrades sarcomeric proteins that ultimately leads to sarcomere dysfunction [60][61][62] . Upon β-adrenergic stimulation, protein kinase A (PKA)mediated TnI phosphorylation at S23/24 is associated with a decrease in myofilament Ca 2+ sensitivity and contributes to an accelerated rate of cardiac relaxation 63 . ...
Preprint
Full-text available
Diastolic dysfunction is a key feature of the aging heart. We have shown that late-life treatment with mTOR inhibitor, rapamycin, reverses age-related diastolic dysfunction in mice but the molecular mechanisms of the reversal remain unclear. To dissect the mechanisms by which rapamycin improves diastolic function in old mice, we examined the effects of rapamycin treatment at the levels of single cardiomyocyte, myofibril and multicellular cardiac muscle. Compared to young cardiomyocytes, isolated cardiomyocytes from old control mice exhibited prolonged time to 90% relaxation (RT90) and time to 90% Ca2+ transient decay (DT90), indicating slower relaxation kinetics and calcium reuptake with age. Late-life rapamycin treatment for 10 weeks completely normalized RT90 and partially normalized DT90, suggesting improved Ca2+ handling contributes partially to the rapamycin-induced improved cardiomyocyte relaxation. In addition, rapamycin treatment in old mice enhanced the kinetics of sarcomere shortening and Ca2+ transient increase in old control cardiomyocytes. Myofibrils from old rapamycin-treated mice displayed increased rate of the fast, exponential decay phase of relaxation compared to old controls. The improved myofibrillar kinetics were accompanied by an increase in MyBP-C phosphorylation at S282 following rapamycin treatment. We also showed that late-life rapamycin treatment normalized the age-related increase in passive stiffness of demembranated cardiac trabeculae through a mechanism independent of titin isoform shift. In summary, our results showed that rapamycin treatment normalizes the age-related impairments in cardiomyocyte relaxation, which works conjointly with reduced myocardial stiffness to reverse age-related diastolic dysfunction.
Preprint
During heart failure, gene and protein expression profiles undergo extensive compensatory and pathological remodeling. We previously observed that fast skeletal myosin binding protein-C (fMyBP-C) is upregulated in diseased mouse hearts. While fMyBP-C shares significant homology with its cardiac paralog, cardiac myosin binding protein-C (cMyBP-C), there are key differences that may affect cardiac function. However, it is unknown if the expression of fMyBP-C expression in the heart is a pathological or compensatory response. We aim to elucidate the cardiac consequence of either increased or knockout of fMyBP-C expression. To determine the sufficiency of fMyBP-C to cause cardiac dysfunction, we generated cardiac-specific fMyBP-C over-expression mice. These mice were further crossed into a cMyBP-C null model to assess the effect of fMyBP-C in the heart in the complete absence of cMyBP-C. Finally, fMyBP-C null mice underwent transverse aortic constriction (TAC) to define the requirement of fMyBP-C during heart failure development. We confirmed the upregulation of fMyBP-C in several models of cardiac disease, including the use of lineage tracing. Low levels of fMyBP-C caused mild cardiac remodeling and sarcomere dysfunction. Exclusive expression of fMyBP-C in a heart failure model further exacerbated cardiac pathology. Following 8 weeks of TAC, fMyBP-C null mice demonstrated greater protection against heart failure development. Mechanistically, this may be due to the differential regulation of the myosin super-relaxed state. These findings suggest that the elevated expression of fMyBP-C in diseased hearts is a pathological response. Targeted therapies to prevent upregulation of fMyBP-C may prove beneficial in the treatment of heart failure. Significance Statement Recently, the sarcomere – the machinery that controls heart and muscle contraction - has emerged as a central target for development of cardiac therapeutics. However, there remains much to understand about how the sarcomere is modified in response to disease. We recently discovered that a protein normally expressed in skeletal muscle, is present in the heart in certain settings of heart disease. How this skeletal muscle protein affects the function of the heart remained unknown. Using genetically engineered mouse models to modulate expression of this skeletal muscle protein, we determined that expression of this skeletal muscle protein in the heart negatively affects cardiac performance. Importantly, deletion of this protein from the heart could improve heart function suggesting a possible therapeutic avenue.
Article
Full-text available
Microphysiological systems (MPS) are drawing increasing interest from academia and from biomedical industry due to their improved capability to capture human physiology. MPS offer an advanced in vitro platform that can be used to study human organ and tissue level functions in health and in diseased states more accurately than traditional single cell cultures or even animal models. Key features in MPS include microenvironmental control and monitoring as well as high biological complexity of the target tissue. To reach these qualities, cross-disciplinary collaboration from multiple fields of science is required to build MPS. Here, we review different areas of expertise and describe essential building blocks of heart MPS including relevant cardiac cell types, supporting matrix, mechanical stimulation, functional measurements, and computational modelling. The review presents current methods in cardiac MPS and provides insights for future MPS development with improved recapitulation of human physiology.
Article
Impaired relaxation of cardiomyocytes leads to diastolic dysfunction in the left ventricle. Relaxation velocity is regulated in part by intracellular calcium (Ca2+) cycling, and slower outflux of Ca2+ during diastole translates to reduced relaxation velocity of sarcomeres. Sarcomere length transient and intracellular calcium kinetics are integral parts of characterizing the relaxation behavior of the myocardium. However, a classifier tool that can separate normal cells from cells with impaired relaxation using sarcomere length transient and/or calcium kinetics remains to be developed. In this work, we employed nine different classifiers to classify normal and impaired cells, using ex-vivo measurements of sarcomere kinematics and intracellular calcium kinetics data. The cells were isolated from wild-type mice (referred to as normal) and transgenic mice expressing impaired left ventricular relaxation (referred to as impaired). We utilized sarcomere length transient data with a total of n = 126 cells (n = 60 normal cells and n = 66 impaired cells) and intracellular calcium cycling measurements with a total of n = 116 cells (n = 57 normal cells and n = 59 impaired cells) from normal and impaired cardiomyocytes as inputs to machine learning (ML) models for classification. We trained all ML classifiers with cross-validation method separately using both sets of input features, and compared their performance metrics. The performance of classifiers on test data showed that our soft voting classifier outperformed all other individual classifiers on both sets of input features, with 0.94 and 0.95 area under the receiver operating characteristic curves for sarcomere length transient and calcium transient, respectively, while multilayer perceptron achieved comparable scores of 0.93 and 0.95, respectively. However, the performance of decision tree, and extreme gradient boosting was found to be dependent on the set of input features used for training. Our findings highlight the importance of selecting appropriate input features and classifiers for the accurate classification of normal and impaired cells. Layer-wise relevance propagation (LRP) analysis demonstrated that the time to 50% contraction of the sarcomere had the highest relevance score for sarcomere length transient, whereas time to 50% decay of calcium had the highest relevance score for calcium transient input features. Despite the limited dataset, our study demonstrated satisfactory accuracy, suggesting that the algorithm can be used to classify relaxation behavior in cardiomyocytes when the potential relaxation impairment of the cells is unknown.
Article
Identification and proteomic characterization of rare cell types within complex organ-derived cell mixtures is best accomplished by label-free quantitative mass spectrometry. High throughput is required to rapidly survey hundreds to thousands of individual cells to adequately represent rare populations. Here we present parallelized nanoflow dual-trap single-column liquid chromatography (nanoDTSC) operating at 15 min of total run time per cell with peptides quantified over 11.5 min using standard commercial components, thus offering an accessible and efficient LC solution to analyze 96 single cells per day. At this throughput, nanoDTSC quantified over 1000 proteins in individual cardiomyocytes and heterogeneous populations of single cells from the aorta.
Article
Full-text available
It is now apparent that compared with cardiac myofilaments from normal subjects, cardiac myofilaments from patients in end-stage heart failure have depressed maximum Ca2+-dependent actomyosin MgATPase activity. We show evidence that confirms these results. This depression cannot be explained by shifts in the population of myosin heavy chain isoforms. The depression could be due to expression of mutant myosins or to loss of myosin light chain 2. There have also been reports showing that there is a fetal isoform of TnT, the Tm (tropomyosin) binding unit of troponin, expressed in the myopathic myofilaments. In addition, the depression in actomyosin ATPase may be due to changes in the level of phosphorylation of sites on TnT or TnI, the inhibitory unit of troponin, associated with myopathy. We discuss how these changes in the thin filament might affect activation of the myofilaments by modulating the ability of cross-bridges and/or Ca2+ to reverse thin filament inhibition by Tn-Tm. Our discussion emphasizes the role of force-generating cross-bridges as determinants of myofilament activation and considers the possibility that activation by strong cross-bridges could be altered by changes in the thin filament as well as in the thick filament.
Article
The N-terminal region of skeletal myosin light chain-1 (MLC-1) binds to the C terminus of actin, yet the functional significance of this interaction is unclear. We studied a fragment (MLC-pep; residues 5-14) of the ventricular MLC-1. When added to rat cardiac myofibrils, 10 nM MLC-pep induced a supramaximal increase in the MgATPase activity at submaximal Ca2+ levels with no effect at low and maximal Ca2+ levels. A nonsense, scrambled sequence peptide had no effect at any pCa value. MLC-pep did not affect myosin KEDTA and CaATPase activities or actin-activated MgATPase activities in the absence or presence of tropomyosin, The MLC-pep did not alter the ability of troponin I to inhibit MgATPase activity. Moreover, when troponin I and troponin C were extracted from the myofibrils, the MLC-pep lost its ability to stimulate the ATPase rate, This effect was fully restored upon reconstitution of the extracted myofibrils with troponin I-troponin C complex, Thus, activation of MgATPase activity by the peptide required a full complement of thin filament regulatory proteins. Interestingly, the stimulatory effect occurred at a ratio of 4 peptides to 1 thin filament, suggesting that the peptide engages in a highly cooperative process that may involve activation of the entire thin filament.
Article
β-Adrenergic stimulation of cardiac muscle activates protein kinase A (PKA), which is known to phosphorylate proteins on the thin and thick filaments of the sarcomere. Cardiac muscle sarcomeres contain a third filament system composed of titin, and here we demonstrate that titin is also phosphorylated by the β-adrenergic pathway. Titin phosphorylation was observed after β-receptor stimulation of intact cardiac myocytes and incubation of skinned cardiac myocytes with PKA. Mechanical experiments with isolated myocytes revealed that PKA significantly reduces passive tension. In vitro phosphorylation of recombinant titin fragments and immunoelectron microscopy suggest that PKA targets a subdomain of the elastic segment of titin, referred to as the N2B spring element. The N2B spring element is expressed only in cardiac titins, in which it plays an important role in determining the level of passive tension. Because titin-based passive tension is a determinant of diastolic function, these results suggest that titin phosphorylation may modulate cardiac function in vivo.
Article
Phosphorylation of myofilament proteins by kinases such as cAMP-dependent protein kinase and protein kinase C has been shown to lead to altered thin-filament protein-protein interactions and modulation of cardiac function in vitro. In the present study, we report that a small GTPase-dependent kinase, p21-activated kinase (PAK), increases the calcium sensitivity of Triton-skinned cardiac muscle fiber bundles. Constitutively active PAK3 caused an average 1.25-fold (25.06.0%, n6) increase in force at pCa 5.75, 1.44-fold (44.07.78%, n6) at pCa 6.25, and 2.41-fold (141.223.7%, n4) at pCa 6.5, representing a change in pCa50 value of approximately 0.25. Constitutively active PAK3 produced no change in force under conditions of relaxation (pCa 8.0) or maximal contraction (pCa 4.5). Furthermore, an inactive, kinase-dead form of PAK3 failed to produce any change in force development at any pCa value. The myofilament proteins phosphorylated by PAK3, at pCa 6.5, are desmin, troponin T, troponin I, and an unidentified 70-kDa protein. Importantly, cardiac troponin I was found to be phosphorylated at serine 149 of human cardiac troponin I, representing a novel phosphorylation site. These findings suggest a novel mechanism of modulating the calcium sensitivity of cardiac muscle contraction. (Circ Res. 2002;91:509-516.)
Article
p38 Mitogen-activated protein kinase (MAPK) is one of the most ancient signaling molecules and is involved in multiple cellular processes, including cell proliferation, cell growth, and cell death. In the heart, enhanced activation of p38 MAPK is associated with ischemia/reperfusion injury and the onset of heart failure. In the present study, we investigated the function of p38 MAPK in regulating cardiac contractility and its underlying mechanisms. In cultured adult rat cardiomyocytes, activation of p38 MAPK by adenoviral gene transfer of an activated mutant of its upstream kinase, MKK3bE, led to a significant reduction in baseline contractility, compared with uninfected cells or those infected with a control adenoviral vector (Adv-β-galactosidase). The inhibitory effect of MKK3bE on contractility was largely prevented by coexpressing a dominant-negative mutant of p38 MAPK or treating cells with a p38 MAPK inhibitor, SB203580. Conversely, inhibition of endogenous p38 MAPK activity by SB203580 rapidly and reversibly enhanced cell contractility in a dose-dependent manner, without altering L-type Ca²⁺ currents or Ca²⁺i transients. MKK3bE-induced p38 activation had no significant effect on pHi, whereas SB203580 had a minor effect to elevate pHi. Furthermore, activation of p38 MAPK was unable to increase troponin I phosphorylation. Thus, we conclude that the negative inotropic effect of p38 MAPK is mediated by decreasing myofilament response to Ca²⁺, rather than by altering Ca²⁺i homeostasis and that the reduced myofilament Ca²⁺ sensitivity is unlikely attributable to troponin I phosphorylation or alterations in pHi. These findings reveal a novel function of p38 MAPK and shed a new light on our understanding of the coincidence of p38 MAPK activation and the onset of heart failure.
Article
Summary We evaluated the amount of β1- and β2-adrenoceptors in human right and left atrium as well as in right and left ventricular wall obtained from heart transplant recipients who suffered from end-stage congestive cardiomyopathy. The total number of myocardial β-adrenoceptors was assessed with the nonsubtype selective β-adrenoceptor radioligand (-)[‘25I]iodocyanopindolol (ICYP); concomitantly, the number of β1-adrenoceptors was determined with the selective β1-adrenoceptor radioligand (-)[3H]bisoprolol. The number of β2-adrenoceptors was calculated by subtracting (-)[3H]bisoprolol binding sites from ICYP binding sites. With this technique, a β1/β2-ratio of ∼65/35% for both atria and of ∼75/25% for both ventricles was found. Identical results were obtained when the β1/β2-ratio was calculated indirectly by nonlinear regression analysis of competition curves of the selective β1-adrenoceptor antagonist bisoprolol and the selective β2-adrenoceptor antagonist ICI 118,551 with ICYP binding. In addition, on atria and on ventricles, adenylate cyclase was activated by norepinephrine (presumably by β1- and β2-adrenoceptor stimulation) and by procaterol (by β2-adrenoceptor stimulation). It is concluded that in the human heart functional β1- and β2-adrenoceptors coexist on both atria and both ventricles. In end-stage congestive cardiomyopathy, there appears to be a selective down-regulation of cardiac β1-adrenoceptors, whereas β2-adrenoceptors are obviously not affected. This may explain the beneficial effects of β2-adrenoceptor agonists in severe heart failure.
Article
We and others have previously cloned several cDNAs of human cardiac troponin T (cTnT), demonstrating the multiplicity of cTnT isoforms in the human heart. Four of them named cTnT1, 2, 3 and 4 result from a combinatorial alternative inclusion of 30-and 15-nucleotides in the 5′ coding region of the cDNAs. In failing human ventricles, increased expression of cTnT4 has been reported at the protein level. More recent RT-PCR experiments showed increased expression of fetal-type splicing products in the 5′ region, one of them corresponding to cTnT1. To clarify this issue, we examined the accumulation of the 4 cTnT mRNA and protein species in left ventricular specimens at the time of heart transplantation, and in control left ventricular samples using RNase protection and Western blotting. In all samples, cTnT3 was the major mRNA isoform, cTnT4 a minor isoform while cTnT1 and cTnT2 mRNAs were present but barely detectable. At the protein level, cTnT3, 4 and 1 were detected with the same relative abundance as that seen at the mRNA level. In addition, we detected a fourth TnT species of very low abundance corresponding either to a skeletal or to a “short” cardiac TNT isoform. Compared to controls, increased levels of cTnT4 mRNA and protein were detected in only half the failing ventricles independently of the cause of failure, suggesting that this increase may not be a general characteristic of left ventricular failure but instead could be related to stress. Unexpectedly, we found a decrease in cTnT1 protein expression in all failing ventricular samples studied, compared to controls.
Article
The cardiac myofilaments are composed of highly ordered arrays of proteins that coordinate cardiac contraction and relaxation in response to the rhythmic waves of [Ca(2+)] during the cardiac cycle. Several cardiac disease states are associated with altered myofilament protein interactions that contribute to cardiac dysfunction. During acute myocardial ischemia, the sensitivity of the myofilaments to activating Ca(2+) is drastically reduced, largely due to the effects of intracellular acidosis on the contractile machinery. Myofilament Ca(2+) sensitivity remains compromised in post-ischemic or "stunned" myocardium even after complete restoration of blood flow and intracellular pH, likely because of covalent modifications of or proteolytic injury to contractile proteins. In contrast, myofilament Ca(2+) sensitivity can be increased in chronic heart failure, owing in part to decreased phosphorylation of troponin I, the inhibitory subunit of the troponin regulatory complex. We highlight, in this paper, the central role of the myofilaments in the pathophysiology of each of these distinct disease entities, with a particular focus on the molecular switch protein troponin I. We also discuss the beneficial effects of a genetically engineered cardiac troponin I, with a histidine button substitution at C-terminal residue 164, for a variety of pathophysiologic conditions, including hypoxia, ischemia, ischemia-reperfusion and chronic heart failure.
Article
Protein kinase D (PKD) is a serine kinase whose myocardial substrates are unknown. Yeast 2-hybrid screening of a human cardiac library, using the PKD catalytic domain as bait, identified cardiac troponin I (cTnI), myosin-binding protein C (cMyBP-C), and telethonin as PKD-interacting proteins. In vitro phosphorylation assays revealed PKD-mediated phosphorylation of cTnI, cMyBP-C, and telethonin, as well as myomesin. Peptide mass fingerprint analysis of cTnI by liquid chromatography-coupled mass spectrometry indicated PKD-mediated phosphorylation of a peptide containing Ser22 and Ser23, the protein kinase A (PKA) targets. Ser22 and Ser23 were replaced by Ala, either singly (Ser22Ala or Ser23Ala) or jointly (Ser22/23Ala), and the troponin complex reconstituted in vitro, using wild-type or mutated cTnI together with wild-type cardiac troponin C and troponin T. PKD-mediated cTnI phosphorylation was reduced in complexes containing Ser22Ala or Ser23Ala cTnI and completely abolished in the complex containing Ser22/23Ala cTnI, indicating that Ser22 and Ser23 are both targeted by PKD. Furthermore, troponin complex containing wild-type cTnI was phosphorylated with similar kinetics and stoichiometry (approximately 2 mol phosphate/mol cTnI) by both PKD and PKA. To determine the functional impact of PKD-mediated phosphorylation, Ca2+ sensitivity of tension development was studied in a rat skinned ventricular myocyte preparation. PKD-mediated phosphorylation did not affect maximal tension but produced a significant rightward shift of the tension-pCa relationship, indicating reduced myofilament Ca2+ sensitivity. At submaximal Ca2+ activation, PKD-mediated phosphorylation also accelerated isometric crossbridge cycling kinetics. Our data suggest that PKD is a novel mediator of cTnI phosphorylation at the PKA sites and may contribute to the regulation of myofilament function.
Article
Phosphorylation of cardiac myofilament proteins represents one of the main post-translational mechanisms that regulate cardiac pump function. Human studies are often limited by the amount of available tissue as biopsies taken during cardiac catheterization weigh only 1 mg (dry weight). Similarly, investigation of time- (or dose-) dependent changes in protein phosphorylation in animal studies is often hampered by tissue availability. The present study describes quantitative analysis of phosphorylation status of multiple myofilament proteins by 2-DE and Pro-Q® Diamond stained gradient gels using minor amounts (˜0.5 mg dry weight) of human and pig cardiac tissue.