ArticlePDF Available

Deranged Myofilament Phosphorylation and Function in Experimental Heart Failure with Preserved Ejection Fraction.

Authors:

Abstract and Figures

AimsHeart failure (HF) with preserved ejection fraction (HFpEF) is a major cause of morbidity and mortality. Key alterations in HFpEF include increased left ventricular (LV) stiffness and abnormal relaxation. We hypothesized that myofilament protein phosphorylation and function are deranged in experimental HFpEF vs. normal myocardium. Such alterations may involve the giant elastic protein titin, which contributes decisively to LV stiffness.Methods and resultsLV tissue samples were procured from normal dogs (CTRL) and old dogs with hypertension-induced LV hypertrophy and diastolic dysfunction (OHT/HFpEF). We quantified the expression and phosphorylation of myofilament proteins, including all-titin and site-specific titin phosphorylation, and assessed the expression/activity of major protein kinases (PKs) and phosphatases (PPs), myofilament calcium sensitivity (pCa50), and passive tension (F passive) of isolated permeabilized cardiomyocytes. In OHT vs. CTRL hearts, protein kinase-G (PKG) activity was decreased, whereas PKC activity and PP1/PP2a expression were increased. Cardiac MyBPC, TnT, TnI and MLC2 were less phosphorylated and pCa50 was increased in OHT vs. CTRL. The titin N2BA (compliant) to N2B (stiff) isoform-expression ratio was lowered in OHT. Hypophosphorylation in OHT was detected for all-titin and at serines S4010/S4099 within titin-N2Bus, whereas S11878 within proline, glutamate, valine, and lysine (PEVK)-titin was hyperphosphorylated. Cardiomyocyte Fpassive was elevated in OHT, but could be normalized by PKG or PKA, but not PKC, treatment.Conclusions This patient-mimicking HFpEF model is characterized by titin stiffening through altered isoform composition and phosphorylation, both contributing to increased LV stiffness. Hypophosphorylation of myofilament proteins and increased calcium sensitivity suggest that functional impairment at the sarcomere level may be an early event in HFpEF.
Content may be subject to copyright.
.....................................................................................................................................................................................
.....................................................................................................................................................................................
Deranged myofilament phosphorylation and
function in experimental heart failure with
preserved ejection fraction
Nazha Hamdani1, Kalkidan G. Bishu2, Marion von Frieling-Salewsky1,
Margaret M. Redfield2, and Wolfgang A. Linke1*
1
Department of Cardiovascular Physiology, Ruhr University, MA 3/56, D-44780 Bochum, Germany; and
2
Mayo Clinic and Foundation, Rochester, MN, USA
Received 16 April 2012; revised 14 November 2012; accepted 28 November 2012; online publish-ahead-of-print 4 December 2012
Time for primary review: 28 days
Aims Heart failure (HF) with preserved ejection fraction (HFpEF) is a major cause of morbidity and mortality. Key altera-
tions in HFpEF include increased left ventricular (LV) stiffness and abnormal relaxation. We hypothesized that myo-
filament protein phosphorylation and function are deranged in experimental HFpEF vs.normal myocardium. Such
alterations may involve the giant elastic protein titin, which contributes decisively to LV stiffness.
Methods
and results
LV tissue samples were procured from normal dogs (CTRL) and old dogs with hypertension-induced LV hypertrophy
and diastolic dysfunction (OHT/HFpEF). We quantified the expression and phosphorylation of myofilament proteins,
including all-titin and site-specific titin phosphorylation, and assessed the expression/activity of major protein kinases
(PKs) and phosphatases (PPs), myofilament calcium sensitivity (pCa
50
), and passive tension (F
passive
) of isolated per-
meabilized cardiomyocytes. In OHT vs.CTRL hearts, protein kinase-G (PKG) activity was decreased, whereas PKCa
activity and PP1/PP2a expression were increased. Cardiac MyBPC, TnT, TnI and MLC2 were less phosphorylated and
pCa
50
was increased in OHT vs.CTRL. The titin N2BA (compliant) to N2B (stiff) isoform-expression ratio was
lowered in OHT. Hypophosphorylation in OHT was detected for all-titin and at serines S4010/S4099 within titin-
N2Bus, whereas S11878 within proline, glutamate, valine, and lysine (PEVK)-titin was hyperphosphorylated. Cardio-
myocyte F
passive
was elevated in OHT, but could be normalized by PKG or PKA, but not PKCa, treatment.
Conclusions This patient-mimicking HFpEF model is characterized by titin stiffening through altered isoform composition and
phosphorylation, both contributing to increased LV stiffness. Hypophosphorylation of myofilament proteins and
increased calcium sensitivity suggest that functional impairment at the sarcomere level may be an early event in
HFpEF.
-----------------------------------------------------------------------------------------------------------------------------------------------------------
Keywords Diastolic heart failure Hypertrophy Titin Passive stiffness
1. Introduction
Heart failure (HF) is a major cause of mortality and morbidity and a
frequent reason for hospital admission in the USA and Europe.
1
More than 50% of HF patients have a left ventricular (LV) ejection
fraction (EF) .50% and are referred to as patients with HF with a
preserved EF (HFpEF). Typically, HFpEF patients show impaired LV
filling resulting from abnormal relaxation and increased LV diastolic
stiffness.
2,3
The factors contributing to the increased LV passive stiff-
ness include cardiac hypertrophy, fibrosis,
4
abnormal Ca
2+
-handling,
3
or deranged expression/phosphorylation of the elastic sarcomere
protein titin.
57
Cardiac titin is expressed as stiff N2B isoform (3000 kDa) and more
compliant N2BA isoform (3200– 3700 kDa),
8
and the N2BA:N2B ex-
pression ratio partly defines myofibrillar passive stiffness.
911
Patho-
logically increased N2BA:N2B ratios have been found in end-stage
human HF with reduced EF (HFrEF).
9,11
In human HFpEF, the propor-
tion of compliant N2BA titin is also increased, but cardiomyocytes
have been found to be stiffer than normal.
7
The increased cardio-
myocyte passive tension (F
passive
) may be due to deranged titin
*Corresponding author. Tel: +49 234 3229100; fax: +49 234 3214040, Email: wolfgang.linke@rub.de
Published on behalf of the European Society of Cardiology. All rights reserved. &The Author 2012. For permissions please email: journals.permissions@oup.com.
Cardiovascular Research (2013) 97, 464–471
doi:10.1093/cvr/cvs353
by guest on January 12, 2016Downloaded from
phosphorylation. A cardiac-specific segment in titin, the N2Bus, is
phosphorylated by protein kinase (PK)A
12,13
and cGMP-activated
protein kinase-G (PKG),
14
an effect augmented by PDE5A-inhibitor
sildenafil, in vivo.
15
This titin modification reduces cardiomyocyte
F
passive
,
6,7,12 15
whereas a deficit in phosphorylation at the N2Bus-titin
site would increase F
passive
.
7,14
However, if the elastic titin segment is
phosphorylated at a different site, the proline, glutamate, valine, and
lysine-(PEVK) domain, by PKCa, titin-based stiffness increases.
16
PKCais elevated in HF
17
and hyperphosphorylation of the PEVK
domain could increase F
passive
in failing hearts.
18
Thus, while titin phos-
phorylation changes most probably alter F
passive
in HF, we are only be-
ginning to understand which sites on titin become more or less
phosphorylated in failing hearts and how these modifications alter dia-
stolic stiffness, particularly in HFpEF.
Cardiac remodelling in HF also involves phosphorylation changes in
other myofilament proteins, notably the regulatory proteins.
1921
Little
is known about alterations in regulatory protein phosphorylation in
HFpEF, whereas HFrEF has been well studied for such alterations.
1921
A prominentexample is cardiac troponin I (cTnI), which is largely respon-
sible for myofilament calcium sensitivity. TnI has been found to be hyper-
phosphorylated in HFrEF in some studies, but hypophosphorylated in
others, and similar differences have been reported for other regulatory
proteins.
2227
A firm cause for these discrepancies is not known.
We speculated that derangements in phosphorylation and function
of cardiac myofilament proteins may occur in experimental dogs with
advanced age and hypertension-induced LV hypertrophy,
15,28
in com-
parison to normal dog hearts. Whereas the dog model has been
shown to mimic some forms of human HFpEF,
15,28
these dogs have
not been studied yet for biochemical and functional properties at
the level of the sarcomere proteins. We find hypophosphorylation
of regulatory proteins and increased Ca
2+
sensitivity of the contractile
apparatus in the experimental HFpEF model. We also detect elevated
F
passive
in HFpEF dog hearts owing to both titin-isoform switching and
altered titin phosphorylation, including site-specific phosphorylation
revealed by novel phospho-specific antibodies. While these altera-
tions are already apparent in the HFpEF model, in vivo cardiac mech-
anical function is still maintained. Our findings provide a novel
mechanistic insight into the remodelling processes during HFpEF de-
velopment and suggest new possibilities for therapeutic interventions
in this syndrome.
2. Methods
A detailed methods and additional data section is provided in the Supple-
mentary material online.
2.1 Animal model, in vivo mechanical analysis,
and tissue sampling
The study employed mongrel dogs (n¼39; Supplementary material
online, Table S1) that were divided into controls [CTRL; aged one year
(‘young’) or 8 12 years (‘old’)] and old dogs made hypertensive by bilat-
eral renal wrapping (OHT; aged 813 years).
28
All dogs underwent echo-
cardiography in the conscious unsedated state. Short-term haemodynamic
studies were performed in CTRL and OHT dogs 8 weeks after renal
wrapping or sham surgery.
28
Animals were anaesthetized using fentanyl
(0.25 mg kg
21
intravenous bolus followed by 0.18 mg kg
21
h
21
) and mid-
azolam (0.75 mg kg
21
intravenous bolus followed by 0.59 mg kg
21
h
21
).
Adequacy of anaesthesia was monitored from the disappearance of the
corneal reflex and jaw tone. Dogs were intubated, ventilated, and given
maintenance saline infusion (3 mL kg
21
min
21
), and they received
autonomic blockade with atropine (1 mg) and propranolol (2 mg kg
21
).
Thoracotomy and pericardiotomy were performed. Under fluoroscopic
guidance, animals were instrumented with a pulmonary artery catheter,
an LV integrated pressure-conductance catheter (Millar), a left atrial and
central aortic high-fidelity pressure transducer (Millar), a pneumatic oc-
cluding device around the thoracic inferior vena cava, and an atrial lead
for pacing at 10– 20 bpm above the sinus rate. LV tissue samples were
procured from CTRL and OHT (8 weeks after renal wrapping), either
by taking full-thickness LV biopsies from the beating heart (n¼7 per
group) or by excising tissue post-mortem (n¼7– 9 per group).
In the beating-heart biopsy group, serial samples were harvested fromdif-
ferent regions of the anterior or anterior lateral wall from seven CTRL and
seven OHT dogs subjected to an identical experimental protocol without
collection of haemodynamic data, because the biopsy and haemostatic
sutures would alter chamber diastolic properties. Biopsy samples were
frozen in liquid nitrogen within seconds and stored at 2808C until use.
The investigation conforms with the Guide for the Care and Use of Labora-
tory Animals published by the US National Institutes of Health (NIH Pub-
lication, 8th Edition, 2011) and was approved by the Mayo Institutional
Animal Care and Use Committee. Dogs were euthanized by intravenous
KCl under deep anaesthesia, consistent with the Panel on Euthanasia guide-
lines for the American Veterinary Medical Association. Hearts were
removed post-mortem, weighed, and the LV sectioned for samples that
were flash frozen in liquid nitrogen. This procedure occurred as quickly
as possible but could take up to 60 min. Samples were stored at 2808C
until use. All data shown for the CTRL post-mortem group were obtained
from old (aged) animals; however, young CTRL hearts procured post-
mortem revealed a similar myofilament protein expression and phosphor-
ylation and cardiomyocyte mechanical properties compared with old
CTRL post-mortem hearts (data not shown).
2.2 Protein analysis
Titin isoform separation. Homogenized myocardial samples were analysed
by 2% sodium dodecyl sulphatepolyacrylamide gel electrophoresis
(SDSPAGE).
9
Protein bands were visualized using Coomassie blue or
SYPRO Ruby, scanned, and analysed densitometrically.
Total protein phosphorylation assays. Tissue samples (20 mg dry weight/
lane) were separated on 2% SDS PAGE gels for titin, or on gradient
gels for other myofilament proteins. Gels were stained with Pro-Q
Diamond for 1 h and subsequently with SYPRO Ruby overnight. Phos-
phorylation signals for myofilament proteins on Pro-Q Diamond-stained
gels were normalized to SYPRO Ruby-stained total protein signals.
Immunoblotting. Expression of cMyBPC, phospho-cMyBPC (S282), cTnI,
phospho-cTnI (S23/S24), cMLC2, phospho-cMLC2 (S19), PKCa,
phospho-PKCa, PP1, and PP2a was measured by 15% SDS– PAGE and
western blot, expression of titin phospho-N2Bus (S4010; S4099) and
phospho-PEVK (S11878) by 2% SDS–PAGE and western blot. The position
of titin phosphosites is according to full-length human titin, UniProtKB identi-
fier, Q8WZ42. Affinity-purified phosphospecific and sequence-specific anti-
titin antibodies were custom-made by Eurogentec (Belgium).
2.3 Myocardial protein kinase G and A activity
tests
PKG activity (pmol/min/mg protein) was measured using radiolabeled
ATP. PKA activity (ng mL
21
) was measured using a nonradioactive PKA
kinase activity-assay kit.
2.4 Force measurements on isolated skinned
cardiomyocytes
Cardiomyocytes were demembranated and isolated cells were attached
between a force transducer and a motor.
29
F
passive
was recorded
between 1.8 and 2.4 mm sarcomere length (SL). Ca
2+
sensitivity of the
contractile apparatus (pCa
50
) was determined at 2.2 mm SL.
Myofilament alterations in HFpEF 465
by guest on January 12, 2016Downloaded from
2.5. Statistics
The values are given as mean +SEM in each group. Data were tested for
statistically significant differences using the Bonferroni-adjusted t-test,
apart from Figure 6and Supplementary material online, Figure S10,
where the paired Student’s t-test was used. A P-value of ,0.05 was con-
sidered significant.
3. Results
Clinical, haemodynamic (anaesthesia), and conscious echocardiog-
raphy data were available for all groups of dogs (Supplementary ma-
terial online, Table S1). OHT dogs had chronic hypertension as
described previously.
28,30
Both systolic and diastolic blood pressure
were significantly increased in OHT vs.CTRL, as were the LV end-
systolic pressure, the relaxation constant Tau, and the LV weight/
body weight ratio. Hypertrophy was present in OHT, as indicated
by an increased mean cardiomyocyte diameter in this group com-
pared with CTRL (Supplementary material online, Figure S1). LV EF
was unaltered in OHT vs.CTRL dogs. Thus, the OHT dogs showed
typical signs of early HFpEF.
3.1. Hypophosphorylation of myofilament
regulatory proteins in OHT
Myofilament regulatory proteins cMyBPC, cTnI, cTnT, and cMLC2
showed reduced phosphorylation in biopsies of OHT vs.CTRL
hearts, as detected by Pro-Q Diamond/SYPRO Ruby staining
(Figure 1). In post-mortem OHT hearts, cMyBPC, cTnI, and cTnT
were also hypophosphorylated compared with CTRL, whereas
cMLC2 phosphorylation was unaltered (Supplementary material
online, Figure S2). Using western blots for the detection of myofila-
ment protein expression and phosphorylation, we found cTnI phos-
phorylation at S23/S24 to be significantly reduced by .50% in
OHT vs.CTRL, in beating-heart biopsies (Figure 2B) and post-
mortem tissues (Supplementary material online, Figure S3). Further-
more, cMyBPC phosphorylation at S282 was reduced by 80% in
biopsied OHT vs.CTRL samples and cMLC2 phosphorylation at
S19 was reduced by 70% (Figure 2Aand C). Total cMyBPC, cTnI,
and cMLC2 expression remained unaltered in OHT (Figure 2).
3.2. Alterations in expression/activity of
major protein kinases and phosphatases
in OHT
The activity of PKG was reduced in OHT vs. CTRL (Supplementary
material online, Figure S4A and B). PKCaexpression was similar in
CTRL and OHT (Supplementary material online, Figure S4C and D),
but phosphorylation of PKCa(a measure of kinase activity) was
higher in OHT vs. CTRL (Supplementary material online, Figure S4E
and F). PKA activity was not different between sample groups (Sup-
plementary material online, Figure S5). Expression of PP1 (Supplemen-
tary material online, Figure S6A and B) and PP2a (Supplementary
material online, Figure S6C and D) was increased in OHT vs. CTRL.
3.3. Altered calcium sensitivity of
cardiomyocytes in OHT and effect of PKA
treatment
The forcepCa relationship of skinned single cardiomyocytes from
beating-heart biopsies (n, 1016 cells per group; from 2 to 3 hearts
per group) revealed significantly higher myofilament calcium
sensitivity (pCa
50
) in OHT (5.77 +0.01) vs.CTRL (5.64 +0.02)
(Figure 3A). Maximum Ca
2+
-activated tension was reduced in OHT
(Figure 3A, inset). In cardiomyocytes from post-mortem hearts,
pCa
50
was significantly lower in OHT (5.52 +0.01) than in CTRL
(5.61 +0.01), while maximum Ca
2+
-activated tension was also
reduced (Supplementary material online, Figure S7). The Hill coeffi-
cient indexing the steepness of the force– Ca
2+
curve was decreased
in OHT (1.9+0.2) vs.CTRL (2.5 +0.2) biopsy samples (Figure 3A),
but increased in OHT (3.9 +0.5) vs.CTRL (3.0 +0.8) post-
mortem samples (Supplementary material online, Figure S7). The
Ca
2+
sensitivity of the contractile apparatus was significantly
reduced (P¼0.012) in OHT cardiomyocytes upon incubation with
a PKA catalytic subunit (pCa
50
shift, 0.16 +0.02 units), whereas in
CTRL myocytes this effect was not significant ( pCa
50
shift, 0.06 +
0.01 units) (Figure 3B).
3.4. Titin isoform shift towards N2B
in OHT
By measuring the titin N2BA/N2B isoform composition (Figure 4A;
Supplementary material online, Figure S8A), we found that the mean
N2B proportion in biopsied samples increased from 63.3 +3.3% in
CTRL to 72.6 +3.3% in OHT (Figure 4B), and in post-mortem
Figure 1 Phosphorylation of regulatory myofilament proteins in
biopsy samples from OHT and CTRL dog hearts, by Pro-Q
Diamond/SYPRO Ruby staining. Top panels show representative
SDSPAGE gels. M, peppermint-stick marker. Graphs show mean
phospholevels of cMyBPC, cTnI, cTnT, and cMLC2, normalized to
their respective total protein level. Heart samples (n¼7 9) were
analysed in duplicate. *P,0.05.
N. Hamdani et al.466
by guest on January 12, 2016Downloaded from
tissues from 57.5 +6.1% in CTRL to 65.9 +5.1% in OHT (Supple-
mentary material online, Figure S8B). A ‘T2’ titin degradation band
was barely detectable in biopsy samples, but was more frequent
and more intense in post-mortem tissues.
3.5. Phosphorylation deficit of all-titin
in OHT and rescue by PKG
All-titin phosphorylation measured by Pro-Q Diamond/SYPRO Ruby
staining decreased by 30% in OHT vs.CTRL biopsies, and by ~60%
in post-mortem tissue (Figure 4C; Supplementary material online,
Figure S8C). Both N2BA and N2B titin isoforms were hypophosphory-
lated in OHT. Importantly, ex vivo phosphorylation by
cGMP-dependent PKG significantly increased all-titin phosphorylation
in OHT, up to the level measured in CTRL (Figure 4C; Supplementary
material online, Figure S8C). This increase was larger in N2B than in
N2BA titin.
3.6. Site-specific phosphorylation at
titin-N2Bus (S4010/S4099) and titin-PEVK
(S11878)
Alterations in all-titin phosphorylation reflect modifications at poten-
tially hundreds of amino acids within titin. Furthermore, the Pro-Q
Diamond stain reportedly fails to detect phosphosites within titin’s
PEVK domain.
18
Using custom-made phosphospecific titin antibodies,
we measured changes in phosphorylation at two conserved serines
within the N2Bus (S4010 and S4099 of full-length human titin) and
at a conserved serine of the PEVK segment (S11878) by western
blot (Figure 5; Supplementary material online, Figure S9). The mean
proportions of titin N2Bus phosphorylation at S4010 (Figure 5A; Sup-
plementary material online, Figure S9A) and S4099 (Figure 5B; Supple-
mentary material online, Figure S9B) were significantly lower in OHT
vs.CTRL. In contrast, the mean proportion of phospho-PEVK
(S11878) was significantly higher in OHT than in CTRL (Figure 5C;
Supplementary material online, Figure S9C). These phosphorylation
changes occurred in both titin isoforms.
Figure 2 Expression and phosphorylation of cMyBPC, cTnI, and
cMLC2, in CTRL and OHT biopsy samples by western blot. Left
panels show phosphorylated, right panels total protein levels; repre-
sentative immunoblots above graphs, which indicate mean phos-
phorylation/expression. (A), cMyBPC (S282) phosphorylation and
cMyBPC expression; (B), cTnI (S23/S24) phosphorylation and cTnI
expression; (C), cMLC2 (S19) phosphorylation and cMLC2 expres-
sion. Data in graphs are normalized to beta-actin signals. Heart
samples (n¼79) were analysed in duplicate. *P,0.05.
Figure 3 Myofilament calcium sensitivity of skinned isolated cardi-
omyocytes from biopsy samples. (A), Relative force vs. pCa relation-
ship at 2.2 mm SL of OHT compared with CTRL cardiomyocytes.
Inset, absolute values for actively developed tension vs.pCa. (B),
Relative force vs. pCa relationship for CTRL and OHT cardiomyo-
cytes at 2.2 mm SL, before (black symbols and curves; data taken
from panel A) and after incubation with a PKA catalytic subunit
(red symbols and curves). n¼10 16 myocytes per group, from
three different hearts per group. *P,0.05.
Myofilament alterations in HFpEF 467
by guest on January 12, 2016Downloaded from
3.7. Cardiomyocyte F
passive
is increased
in OHT but lowered by administration of
PKA or PKG
The passive SLtension relationship of isolated skinned cardiomyo-
cytes (n, 1016 per group, from 2 to 3 hearts per group) was gener-
ally steeper in OHT than in CTRL (Figure 6; Supplementary material
online, Figure S10). Administration of PKA significantly reduced F
passive
of OHT cells at a SL of 2.22.4 mm, sometimes already at shorter SLs
(Figure 6A; Supplementary material online, Figure S10A). Even in cardi-
omyocytes from beating-heart biopsies, PKA significantly lowered
F
passive
also in CTRL (Figure 6A). Additional administration of
cGMP-dependent PKG had no obvious mechanical effect in all
groups (Figure 6A; Supplementary material online, Figure S10A). If
PKG was administered first, F
passive
dropped as seen with PKA first
and additional administration of PKA caused no further F
passive
change in the myocytes (Figure 6B; Supplementary material online,
Figure S10B). We also tested whether administration of PKCaalters
F
passive
, but found no significant effect in both CTRL and OHT
(Figure 6C). These results demonstrate that F
passive
is increased in
OHT, but can be corrected by PKG- or PKA-mediated protein
phosphorylation.
4. Discussion
The number of patients hospitalized for HFpEF grows steadily, but
neither is the aetiology of the disease understood well, nor are effect-
ive treatment strategies available.
13
Mechanistic studies on human
Figure 4 Titin-isoform composition and all-titin phosphorylation
in biopsy samples, by Pro-Q Diamond/SYPRO Ruby staining. (A),
Representative titin gels comparing CTRL and OHT samples, and
OHT samples before and after incubation with cGMP-dependent
PKG. Indicated are the N2BA and N2B titin isoforms and titin pro-
teolytic fragment, T2. (B), Mean titin-isoform composition in OHT
and CTRL hearts, measured on SYPRO Ruby-stained gels. Combined
N2BA +N2B signal intensities are 100%. (C) Mean all-titin phos-
phorylation in CTRL and OHT, and in OHT samples incubated ex
vivo with cGMP-dependent PKG. Combined N2BA +N2B phos-
phorylation of CTRL set to 100%. n¼79 per group; heart
samples analysed in triplicate. *P,0.05.
Figure 5 Site-specific phosphorylation of titin in biopsy samples by
western blot. Images on left panels show representative immuno-
blots with OHT and CTRL heart tissue using phosphosite-specific
(top) and corresponding sequence-specific (bottom) anti-titin anti-
bodies. PVDF stains are included for comparison. Graphs on right
show mean phosphorylation levels for N2BA and N2B isoforms in
OHT and CTRL, at position S4010 (N2Bus) (A), S4099 (N2Bus)
(B), and S11878 (PEVK domain) (C). (A) and (B) combined
N2BA +N2B signal intensities of the CTRL set to 100%; (C) com-
bined N2BA +N2B signal intensities of OHT set to 100%. Heart
samples (n¼79) were analysed in triplicate. *P,0.05.
N. Hamdani et al.468
by guest on January 12, 2016Downloaded from
HFpEF are limited by the low availability of tissue samples from dis-
eased and healthy control hearts. Unlike hearts in end-stage failure,
which after transplantation can be used for research purposes,
HFpEF hearts usually do not get explanted. Biopsy samples are some-
times obtained from human HFpEF myocardium, but obviously not
from healthy control hearts. Non-transplanted non-failing donor
hearts occasionally become available for research, but their preserva-
tion is highly variable. These limitations with human cardiac tissue
warrant the study of well-defined animal models of HFpEF, which
can be controlled in terms of age, genetic background, or pharmaco-
logical treatment.
In this study, we used an old dog model of hypertrophy-associated
early HFpEF, which shows signs of diastolic dysfunction resembling
those frequently seen in elderly HFpEF patients: impaired LV relax-
ation, unaltered coefficient of LV diastolic stiffness but reduced dia-
stolic capacitance, along with elevated natriuretic peptides, normal
LV volume but increased LV mass and myocardial fibrosis.
28,30
We
detected profound alterations in cardiac myofilament phosphorylation
and function in dog HFpEF compared with normal dog hearts. Hypo-
phosphorylation of sarcomeric proteins in experimental HFpEF pre-
sumably resulted, at least in part, from the increased PP1 and PP2a
expression and the reduced PKG activity. The deranged
phosphorylation in dog HFpEF altered the calcium sensitivity of the
contractile apparatus and increased cardiomyocyte F
passive
. By focusing
on the elastic protein titin, we found that both isoform switching and
altered phosphorylation increased cardiomyocyte F
passive
in HFpEF.
Since hypophosphorylation of sarcomeric proteins persists in end-
stage human HF,
20
a deficit in phosphorylation of these proteins
could be a general property in the transition to HF. In vivo heart func-
tion was modestly impaired in the dog HFpEF model at rest, but
reserve function, including catecholamine responsiveness (not
studied here), could be more strongly affected. In summary, contrib-
uting factors that drive the HFpEF hearts into diastolic dysfunction
likely include biochemical changes at myofilament proteins leading
to increased calcium responsiveness of force generation and elevated
titin-based passive stiffness.
4.1 Hypophosphorylation of regulatory
myofilament proteins and increased pCa
50
in dog HFpEF
Myofilament Ca
2+
sensitivity is decreased after beta-adrenergic
stimulation of cardiac muscle, an effect largely mediated by increased
PKA-dependent phosphorylation of cTnI.
20,21,24,29,31
Because the
Figure 6 Passive tension of skinned cardiomyocytes isolated from biopsy samples, and effect of incubation with PKA, PKG, or PKCa.(A) and (B),
F
passive
at SL 1.8– 2.4 mm recorded on CTRL (red symbols and curves) or OHT myocytes (green symbols and curves) in non-activating buffer (solid
curves; ‘before’), following incubation with a PKA catalytic subunit (dotted curves; ‘after PKA’), and following incubation with PKG and activator cGMP
(dashed curves; ‘after PKG’). (A) PKA administered first, followed by PKG; (B) PKG administered first, followed by PKA. (C), F
passive
at SL 1.8 2.4 mm
recorded with CTRL or OHT myocytes in non-activating buffer (solid curves; ‘before’) and following incubation with PKCa(dotted curves; ‘after
PKCa’). n¼1016 cardiomyocytes per group, from 2 to 3 hearts per group. Curves are three-order regressions. *P,0.05 CTRL‘before’ vs. OHT‘-
before’;
#
P,0.05 OHT‘before’ vs. OHT‘after PKA’ (in A) and OHT‘before’ vs. OHT‘after PKG’ (in B);
P,0.05 CTRL‘before’ vs. CTRL‘after PKA’ in
(A) and CTRL‘before’ vs. CTRL‘after PKG’ in (B).
Myofilament alterations in HFpEF 469
by guest on January 12, 2016Downloaded from
beta-adrenoceptor density and adenylate cyclase activity are reduced in
HF,
32
hypophosphorylation of cTnI is a usual consequence, particularly
at the PKA
24
(and PKG
33
)-dependent phosphosites, S23/S24. In dog
HFpEF, we found decreased cTnI phosphorylation (including S23/S24
phosphorylation) compared with normal dog hearts, presumably
explaining the increased Ca
2+
sensitivity of skinned cardiomyocytes
from biopsied HFpEF hearts. In line with this interpretation, the
increased Ca
2+
sensitivity of HFpEF (biopsy) cardiomyocytes could
be normalized by ex-vivo PKA treatment. Thus, the dog HFpEF hearts
show alterations in cTnI phosphorylation and myofilament calcium sen-
sitivity resembling those frequently (but not consistently
23,24
) reported
in human HF vs.donor hearts and in animal models of HF.
20,21,29,31
Along this line, b-blockade in HFpEF patients has been associated
with increased cardiomyocyte pCa
50
compared with HFpEF patients
not treated with b-blockers.
29
Furthermore, myofilament pCa
50
changes are unlikely to be due only to cTnI phosphorylation, but also
due to phosphorylation of cMyBPC and cMLC2. We found both
these myofilament proteins, as well as cTnT, to be hypophosphorylated
in biopsy samples of HFpEF dogs. In summary, hypophosphorylation of
regulatory myofilament proteins and increased calcium sensitivity in this
model suggest that functional impairment at the sarcomere level may be
an early event in the development of HFpEF.
Surprisingly, reduced Ca
2+
sensitivity was found in skinned cardi-
omyocytes from HFpEF hearts procured post-mortem, although
phosphorylation of cTnI, cMyBPC, and cTnT was also lowered in
the post-mortem HFpEF group. PKA activity was similar in biopsy
and post-mortem samples, perhaps because all dogs were under
autonomic blockade. The different direction of Ca
2+
-sensitivity
shift in the post-mortem compared with the beating-heart biopsy
group likely originates in events associated with death, such as a cat-
echolamine surge, enzymatic dysfunction, or activation of proteases.
Unlike in biopsy samples, cMLC2 phosphorylation was unaltered in
post-mortem HFpEF vs.CTRL, which might contribute to the differ-
ences in Ca
2+
-sensitivity shift. Additionally, phosphosites in myofila-
ment proteins not tested by us could be modified differently in the
post-mortem and beating-heart groups. Cardiac TnI contains sites
other than S23/S24 which can be phosphorylated and possibly be
important for the Ca
2+
sensitivity,
26
and some functionally relevant
cTnI phosphosites may still be unknown. In conclusion, since we
consider the beating-heart biopsies as the gold standard, the dog
HFpEF hearts have increased Ca
2+
sensitivity. Our results confirm
that degradation processes or other modifications at the time of
death can impact protein phosphorylation and function, which has
implications for the interpretation of data from the samples obtained
post-mortem.
4.2. Titin-isoform switch in HFpEF
vs. HFrEF
The pattern of titin-isoform expression correlates with systolic and
diastolic functional parameters in patients, including LV end-diastolic
wall stiffness,
6
EF, EDV, and ESV.
10
Titin-isoform shift towards the
more compliant N2BA variants occurs in end-stage failing hearts of
patients with ischaemic cardiomyopathy
9
or non-ischaemic dilated
cardiomyopathy (DCM).
10,11
In contrast, we found a modest decrease
in the proportion of N2BA titin in HFpEF dogs. Similarly, a decreased
N2BA proportion has been reported in rapid pacing canine models of
DCM.
34,35
Possibly, then, dog hearts are unique in their remodelling
response to mechanical stress. However, in aortic stenosis patients,
the N2BA proportion was also lowered compared with donor
hearts
36
—although the opposite result was found elsewhere.
7
In
human hypertrophic cardiomyopathy, the cardiac titin-isoform
pattern did not change compared with donor hearts,
37
whereas spon-
taneously hypertensive rats expressed slightly less N2BA proportions
than normotensive rat hearts.
38
In summary, a decreased N2BA:N2B
titin expression ratio may be a frequent (albeit not general) feature of
hypertrophied hearts, including those developing HFpEF. Titin switch-
ing towards the N2B isoform increases cardiomyocyte F
passive
in
HFpEF (this study), switching towards the N2BA isoform decreases
F
passive
in HFrEF.
911
4.3. Titin phosphorylation and F
passive
in HFpEF
Like human end-stage failing hearts,
7,14
dog HFpEF hearts showed a
deficit in phosphorylation of all-titin and at S4010 and S4099 within
titin’s cardiac-only N2Bus. Importantly, ex-vivo administration of
cGMP-dependent PKG corrected the all-titin phosphorylation
deficit in HFpEF heart tissue and administration of PKA or
cGMP-dependent PKG reduced the pathologically increased F
passive
of skinned OHT cardiomyocytes to CTRL levels. Since PKA activity
was unaltered among the dog groups, the deficit in all-titin phosphor-
ylation in HFpEF hearts may, at least partly, be a deficit in
PKG-mediated phosphorylation, causing the higher-than-normal
F
passive
. Lowering F
passive
via increased PKG-mediated phosphorylation
at the titin N2Bus, which improves diastolic function in these dog
hearts,
15
could thus be a useful therapeutic approach in HFpEF.
Against the reduced phosphorylation of all-titin, the PKCa-
dependent phosphosite at S11878 within the PEVK-titin segment
was hyperphosphorylated. Active PKCacan be increased in HF
17
and an elevated PKCaactivity was also apparent in the HFpEF
dog hearts. Because PKCa-dependent phosphorylation at titin-
S11878 (PEVK) increases cardiomyocyte F
passive
,
16
hyperphosphory-
lation at this site presumably added to the higher-than-normal F
passive
in HFpEF.
The increased cardiomyocyte F
passive
in dog HFpEF is consistent
with previous reports of elevated F
passive
in HFpEF patients
39
or
those with diabetes
40
or under b-blockade.
29
Regarding HFrEF,
either reduced
9,11
or elevated
7
F
passive
has been observed compared
with donor hearts. The decreased F
passive
in human HFrEF was
explained by a titin-isoform shift towards the compliant N2BA var-
iants,
9,11
the increased F
passive
by depressed titin phosphorylation,
because administration of PKA lowered F
passive
to control levels.
7
These findings underscore the importance of both titin-isoform tran-
sitions and titin phosphorylation changes for cardiomyocyte F
passive
in
chronic HF. In dog HFpEF, the changes in titin phosphorylation may be
more important for altering LV passive stiffness than the relatively
small transitions in titin isoforms. In any case, we conclude that titin-
based F
passive
is increased in dog experimental HFpEF and contributes
to elevated LV passive stiffness, the hallmark of HFpEF.
4.4. Conclusions
This clinically relevant large-animal model of HFpEF is characterized
by cardiac titin-isoform switch towards the stiffer N2B variant, a
deficit in phosphorylation of all-titin and at specific serines within
the N2Bus-titin domain, but hyperphosphorylation at titin’s PEVK
domain. These alterations act synergistically to elevate cardiomyocyte
F
passive
. A stiffer titin may be a key determinant of diastolic dysfunction
N. Hamdani et al.470
by guest on January 12, 2016Downloaded from
resulting from increased LV passive stiffness in HFpEF. Regulatory
myofilament proteins are hypophosphorylated in dog HFpEF,
causing increased Ca
2+
sensitivity. A phosphorylation deficit for myo-
filament proteins could be an early and general event in the transition
to HF, thus unbalancing cardiac mechanical function. Reversing the
phosphorylation deficit by pharmacological manipulation of PK or
phosphatase (PP) signalling pathways may be a useful therapeutic
strategy in HFpEF.
Supplementary material
Supplementary material is available at Cardiovascular Research online.
Acknowledgement
We thank Dr Martina Kru¨ger and Dr Tobias Voelkel for their help
with titin antibody design.
Conflict of interest: none declared.
Funding
This work was supported by a European Union FP7 grant (MEDIA) as well
as a German Research Foundation grant (SFB 1002, TPB3) to W.A.L. and
aEuropean Society of Cardiology grant to N.H.
References
1. Bursi F, Weston SA, Redfield MM, Jacobsen SJ, Pakhomov S, Nkomo VT et al. Systolic
and diastolic heart failure in the community. JAMA 2006;296:2209 2216.
2. Paulus WJ, Tschope C, Sanderson JE, Rusconi C, Flachskampf FA, Rademakers FE
et al. How to diagnose diastolic heart failure: a consensus statement on the diagnosis
of heart failure with normal left ventricular ejection fraction by the Heart Failure and
Echocardiography Associations of the European Society of Cardiology. Eur Heart J
2007;28:2539–2550.
3. Zile MR, Baicu CF, Gaasch WH. Diastolic heart failure-abnormalities in active relax-
ation and passive stiffness of the left ventricle. N Engl J Med 2004;350:1953 1959.
4. Martos R, Baugh J, Ledwidge M, O’Loughlin C, Conlon C, Patle A et al. Diastolic heart
failure: evidence of increased myocardial collagen turnover linked to diastolic dysfunc-
tion. Circulation 2007;115:888–895.
5. Linke WA. Sense and stretchability: The role of titin and titin-associated proteins in
myocardial stress-sensing and mechanical dysfunction. Cardiovasc Res 2008;77:
637– 648.
6. van Heerebeek L, Borbely A, Niessen HW, Bronzwaer JG, Van der Velden J,
Stienen GJ et al. Myocardial structure and function differ in systolic and diastolic
heart failure. Circulation 2006;113:1966– 1973.
7. Borbely A, Falcao-Pires I, van Heerebeek L, Hamdani N, Edes I, Gavina C et al. Hypo-
phosphorylation of the stiff N2B titin isoform raises cardiomyocyte resting tension in
failing human myocardium. Circ Res 2009;104:780–786.
8. Freiburg A, Trombitas K, Hell W, Cazorla O, Fougerousse F, Centner T et al. Series of
exon-skipping events in the elastic spring region of titin as the structural basis for
myofibrillar elastic diversity. Circ Res 2000;86:1114 1121.
9. 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.
10. Nagueh SF, Shah G, Wu Y, Torre-Amione G, King NM, Lahmers S et al. Altered titin
expression, myocardial stiffness, and left ventricular function in patients with dilated
cardiomyopathy. Circulation 2004;110:155– 162.
11. Makarenko I, Opitz CA, Leake MC, Neagoe C, Kulke M, Gwathmey JK et al. Passive
stiffness changes caused by upregulation of compliant titin isoforms in human dilated
cardiomyopathy hearts. Circ Res 2004;95:708 –716.
12. 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.
13. 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.
14. Kruger M, Kotter S, Grutzner A, Lang P, Andresen C, Redfield MM et al. Protein
kinase G modulates human myocardial passive stiffness by phosphorylation of the
titin springs. Circ Res 2009;104:87 94.
15. Bishu K, Hamdani N, Mohammed SF, Kruger M, Ohtani T, Ogut O et al. Sildenafil and
B-type natriuretic peptide acutely phosphorylate titin and improve diastolic distensi-
bility in vivo. Circulation 2011;124:2882 2891.
16. Hidalgo C, Hudson B, Bogomolovas J, Zhu Y, Anderson B, Greaser M et al. PKC phos-
phorylation of titin’s PEVK element: a novel and conserved pathway for modulating
myocardial stiffness. Circ Res 2009;105:631 638.
17. Belin RJ, Sumandea MP, Allen EJ, Schoenfelt K, Wang H, Solaro RJ et al. Augmented
protein kinase C-alpha-induced myofilament protein phosphorylation contributes to
myofilament dysfunction in experimental congestive heart failure. Circ Res 2007;101:
195– 204.
18. Hudson B, Hidalgo C, Saripalli C, Granzier H. Hyperphosphorylation of mouse
cardiac titin contributes to transverse aortic constriction-induced diastolic dysfunc-
tion. Circ Res 2011;109:858–866.
19. Kobayashi T, Jin L, de Tombe PP. Cardiac thin filament regulation. Pflugers Arch 2008;
457:37– 46.
20. Hamdani N, Kooij V, van Dijk S, Merkus D, Paulus WJ, Remedios CD et al. Sarcomeric
dysfunction in heart failure. Cardiovasc Res 2008;77:649 658.
21. Solaro RJ, Kobayashi T. Protein phosphorylation and signal transduction in cardiac thin
filaments. J Biol Chem 2011;286:9935 9940.
22. Burkart EM, Sumandea MP, Kobayashi T, Nili M, Martin AF, Homsher E et al. Phos-
phorylation 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.
23. 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.
24. Marston SB, de Tombe PP. Troponin phosphorylation and myofilament Ca
2+
-
sensitivity in heart failure: increased or decreased? J Mol Cell Cardiol 2008;45:603 607.
25. Hamdani N, de Waard M, Messer AE, Boontje NM, Kooij V, van Dijk S et al. Myofila-
ment dysfunction in cardiac disease from mice to men. J Muscle Res Cell Motil 2008;29:
189– 201.
26. Solaro RJ, van der Velden J. Why does troponin I have so many phosphorylation sites?
Fact and fancy. J Mol Cell Cardiol 2010;48:810 816.
27. Dong X, Sumandea CA, Chen YC, Garcia-Cazarin ML, Zhang J, Balke CW et al. Aug-
mented phosphorylation of cardiac troponin I in hypertensive heart failure. J Biol Chem
2012;287:848–857.
28. Munagala VK, Hart CY, Burnett JC Jr, Meyer DM, Redfield MM. Ventricular structure
and function in aged dogs with renal hypertension: a model of experimental diastolic
heart failure. Circulation 2005;111:1128–1135.
29. Hamdani N, Paulus WJ, van Heerebeek L, Borbely A, Boontje NM, Zuidwijk MJ et al.
Distinct myocardial effects of beta-blocker therapy in heart failure with normal and
reduced left ventricular ejection fraction. Eur Heart J 2009;30:1863 1872.
30. Shapiro BP, Lam CS, Patel JB, Mohammed SF, Kruger M, Meyer DM et al. Acute and
chronic ventricular– arterial coupling in systole and diastole: insights from an elderly
hypertensive model. Hypertension 2007;50:503 –511.
31. Strang KT, Sweitzer NK, Greaser ML, Moss RL. Beta-adrenergic receptor stimulation
increases unloaded shortening velocity of skinned single ventricular myocytes from
rats. Circ Res 1994;74:542– 549.
32. Bristow MR, Ginsburg R, Umans V, Fowler M, Minobe W, Rasmussen R et al. Beta 1-
and beta 2-adrenergic-receptor subpopulations in nonfailing and failing human ven-
tricular myocardium: coupling of both receptor subtypes to muscle contraction
and selective beta 1-receptor down-regulation in heart failure. Circ Res 1986;59:
297– 309.
33. Lee DI, Vahebi S, Tocchetti CG, Barouch LA, Solaro RJ, Takimoto E et al. PDE5A sup-
pression of acute beta-adrenergic activation requires modulation of myocyte beta-3
signaling coupled to PKG-mediated troponin I phosphorylation. Basic Res Cardiol
2010;105:337–347.
34. Wu Y, Bell SP, Trombitas K, Witt CC, Labeit S, LeWinter MM et al. Changes in titin
isoform expression in pacing-induced cardiac failure give rise to increased passive
muscle stiffness. Circulation 2002;106:13841389.
35. Jaber WA, Maniu C, Krysiak J, Shapiro BP, Meyer DM, Linke WA et al. Titin isoforms,
extracellular matrix, and global chamber remodeling in experimental dilated cardio-
myopathy: functional implications and mechanistic insight. Circ Heart Fail 2008;1:
192– 199.
36. Williams L, Howell N, Pagano D, Andreka P, Vertesaljai M, Pecor T et al. Titin isoform
expression in aortic stenosis. Clin Sci (Lond) 2009;117:237 242.
37. Hoskins AC, Jacques A, Bardswell SC, McKenna WJ, Tsang V, dos Remedios CG et al.
Normal passive viscoelasticity but abnormal myofibrillar force generation in human
hypertrophic cardiomyopathy. J Mol Cell Cardiol 2010;49:737–745.
38. Warren CM, Jordan MC, Roos KP, Krzesinski PR, Greaser ML. Titin isoform expres-
sion in normal and hypertensive myocardium. Cardiovasc Res 2003;59:86 94.
39. Borbely A, van der Velden J, Papp Z, Bronzwaer JG, Edes I, Stienen GJ et al. Cardio-
myocyte stiffness in diastolic heart failure. Circulation 2005;111:774 781.
40. van Heerebeek L, Hamdani N, Handoko ML, Falcao-Pires I, Musters RJ, Kupreishvili K
et al. Diastolic stiffness of the failing diabetic heart: importance of fibrosis, advanced
glycation end products, and myocyte resting tension. Circulation 2008;117:43 51.
Myofilament alterations in HFpEF 471
by guest on January 12, 2016Downloaded from
... In a healthy adult human heart, the N2BA isoform comprises 30%-50% of titin proteins, while the N2B isoform makes up 50%-70% of titin proteins Neagoe et al., 2002). The expression of cardiac titin isoforms has been shown to change during development and in pathological conditions (Bell et al., 2000;Neagoe et al., 2002;Wu et al., 2002;Warren et al., 2003;Makarenko et al., 2004;Nagueh et al., 2004;Opitz et al., 2004;Warren et al., 2004;Shapiro et al., 2007;Borbély et al., 2009;Hudson et al., 2011;Hamdani et al., 2013b;Zhu et al., 2017). ...
... Diastolic dysfunction is a key element of heart diseases such as HFpEF and DbCM (Borlaug, 2014;Jia et al., 2016). Titin size changes resulting from alternative splicing plays a major role in myocardial stiffness, while PTMs in titin's spring-like domain fine-tune myocardial stiffness, as discussed above (Forbes et al., 2005;Krüger et al., 2009;Raskin et al., 2012;Hamdani et al., 2013a;Hamdani et al., 2013b;Hidalgo et al., 2013;Kötter et al., 2013;Perkin et al., 2015;Krysiak et al., 2018;Herwig et al., 2020;Michel et al., 2020;Loescher et al., 2022). Thus, either manipulating titin size switching or altering PTMs in titin could reduce diastolic stiffness and, therefore, dysfunction. ...
Article
Full-text available
The giant protein titin is an essential component of muscle sarcomeres. A single titin molecule spans half a sarcomere and mediates diverse functions along its length by virtue of its unique domains. The A-band of titin functions as a molecular blueprint that defines the length of the thick filaments, the I-band constitutes a molecular spring that determines cell-based passive stiffness, and various domains, including the Z-disk, I-band, and M-line, serve as scaffolds for stretch-sensing signaling pathways that mediate mechanotransduction. This review aims to discuss recent insights into titin’s functional roles and their relationship to cardiac function. The role of titin in heart diseases, such as dilated cardiomyopathy and heart failure with preserved ejection fraction, as well as its potential as a therapeutic target, is also discussed.
... Complementing these findings was evidence pointing to myofilament proteins being less phosphorylated, coupled with an increased Ca 2+ sensitivity. This hints at malfunctions at the sarcomere level as potential early indicators in HFpEF onset [121]. ...
... The relaxation of these filaments is governed by diastolic [Ca 2+ ]i levels and their responsiveness to Ca 2+ . Elevated sensitivity to Ca 2+ in the myofilament, often a result of cardiac TnI's hypophosphorylation, has been observed in cases of HFpEF [121]. Additionally, this heightened Ca 2+ sensitivity in the myofilament has been linked to the diastolic dysfunction seen in hypertrophic cardiomyopathy, a condition often triggered by mutations in sarcomeric genes [124,125]. ...
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.
... Renal wrapping-induced hypertension has been extensively employed in canine models [11,35,36]. The first study specifically designed to simulate HFpEF pathology by inducing renal hypertension on large animal models was conducted by Munagala et al. in 2005 and adopted a bilateral canine renal wrapping model using older dogs and compared their diastolic LV function with young control dogs and old control dogs [35]. ...
... However, further data is required to support the reliability of these findings [13]. A more recent canine renal wrapping model by Hamdani et al. employed old dogs and demonstrated induced chronic hypertension with secondary LVH and fibrosis and impaired diastolic dysfunction, which is consistent with HFpEF pathophysiology [36]. Although this model is established as a large animal HFpEF model and recapitulates several aspects of HFpEF, the requirement for old animals makes this model relatively impractical and expensive. ...
Article
Full-text available
Our understanding of the complex pathophysiology of Heart failure with preserved ejection fraction (HFpEF) is limited by the lack of a robust in vivo model. Existing in-vivo models attempt to reproduce the four main phenotypes of HFpEF; ageing, obesity, diabetes mellitus and hypertension. To date, there is no in vivo model that represents all the haemodynamic characteristics of HFpEF, and only a few have proven to be reliable for the preclinical evaluation of potentially new therapeutic targets. HFpEF accounts for 50% of all the heart failure cases and its incidence is on the rise, posing a huge economic burden on the health system. Patients with HFpEF have limited therapeutic options available. The inadequate effectiveness of current pharmaceutical therapeutics for HFpEF has prompted the development of device-based treatments that target the hemodynamic changes to reduce the symptoms of HFpEF. However, despite the potential of device-based solutions to treat HFpEF, most of these therapies are still in the developmental stage and a relevant HFpEF in vivo model will surely expedite their development process. This review article outlines the major limitations of the current large in-vivo models in use while discussing how these designs have helped in the development of therapy devices for the treatment of HFpEF. Graphical abstract
... At low PKG activity, molecules of the N2B titin isoform are hypophosphorylated, that leads to an increase in Fpassive and, accordingly, a steeper rise in LV filling pressure [96]. It has been shown that titin hypophosphorylation is more pronounced in HFpEF than in HFrEF [97]. High stiffness of cardiomyocytes together with myocardial fibrosis plays the most important role in the myocardial stiffening and increased LV filling pressure [98]. ...
Preprint
Full-text available
Systemic inflammation and coronary microvascular endothelial dysfunction are essential pathophysiological factors in heart failure with preserved ejection fraction (HFpEF) that support the use of statins. The pleiotropic properties of statins, such as anti-inflammatory, antihypertrophic, antifibrotic, and antioxidant effects, are generally accepted and may be beneficial in HF, especially in HFpEF. Numerous observational clinical trials have consistently shown a beneficial prognostic effect of statins in patients with HFpEF, while the results of two larger trials in patients with HFrEF have been controversial. Such differences may be related to a more pronounced impact of the pleiotropic properties of statins on the pathophysiology of HFpEF and pro-inflammatory comorbidities (arterial hypertension, diabetes mellitus, obesity, chronic kidney disease) that are more common in HFpEF. This review discusses the potential mechanisms of statin action that may be beneficial for patients with HFpEF, as well as clinical trials that have evaluated the statin effects on left ventricular diastolic function and clinical outcomes in patients with HFpEF.
... This pathway is often impaired in HF, leading to the contractile dysfunction. HF with preserved ejection fraction is characterized by hypophosphorylation of myofilament proteins and increased calcium sensitivity, altered isoform composition and phosphorylation, contributing to increased left ventricular stiffness (Hamdani et al., 2013). ...
Preprint
Sodium-glucose cotransporter 2 inhibitors (SGLT2i), a new class of glucose-lowering drugs traditionally used to control blood glucose levels in patients with type 2 diabetes mellitus, have been proven to reduce major adverse cardiovascular events, including cardiovascular death, in patients with heart failure irrespective of ejection fraction and independently of the hypoglycemic effect. Because of their favorable effects on the kidney and cardiovascular outcomes, their use has been expanded in all patients with any combination of diabetes mellitus type 2, chronic kidney disease and heart failure. Although mechanisms explaining the effects of these drugs on the cardiovascular system are not well understood, their effectiveness in all these conditions suggests that they act at the intersection of the metabolic, renal and cardiac axes, thus disrupting maladaptive vicious cycles while contrasting direct organ damage. In this systematic review we provide a state of the art of the randomized controlled trials investigating the effect of SGLT2i on cardiovascular outcomes in patients with chronic kidney disease and/or heart failure irrespective of ejection fraction and diabetes. We also discuss the molecular targets and signaling pathways potentially explaining the cardiac effects of these pharmacological agents, from a clinical and experimental perspective.
... Force measurements were performed on single de-membranated cardiomyocytes (n = 20-26/4 hearts/group) as described before [66]. Briefly, LV samples were de-frozen in relaxing solution (containing in mM: 1.0 free Mg 2+ ; 100 KCl; 2.0 EGTA; 4.0 Mg-ATP; 10 imidazole; pH 7.0), mechanically disrupted and incubated for 5 min in relaxing solution supplemented with 0.5% Triton X−100 (all from Sigma-Aldrich, Saint Louis, MO, USA). ...
Article
Full-text available
Protein kinase D (PKD) enzymes play important roles in regulating myocardial contraction, hypertrophy, and remodeling. One of the proteins phosphorylated by PKD is titin, which is involved in myofilament function. In this study, we aimed to investigate the role of PKD in cardiomyocyte function under conditions of oxidative stress. To do this, we used mice with a cardiomyocyte-specific knock-out of Prkd1, which encodes PKD1 (Prkd1loxP/loxP; αMHC-Cre; PKD1 cKO), as well as wild type littermate controls (Prkd1loxP/loxP; WT). We isolated permeabilized cardiomyocytes from PKD1 cKO mice and found that they exhibited increased passive stiffness (Fpassive), which was associated with increased oxidation of titin, but showed no change in titin ubiquitination. Additionally, the PKD1 cKO mice showed increased myofilament calcium (Ca2+) sensitivity (pCa50) and reduced maximum Ca2+-activated tension. These changes were accompanied by increased oxidation and reduced phosphorylation of the small myofilament protein cardiac myosin binding protein C (cMyBPC), as well as altered phosphorylation levels at different phosphosites in troponin I (TnI). The increased Fpassive and pCa50, and the reduced maximum Ca2+-activated tension were reversed when we treated the isolated permeabilized cardiomyocytes with reduced glutathione (GSH). This indicated that myofilament protein oxidation contributes to cardiomyocyte dysfunction. Furthermore, the PKD1 cKO mice exhibited increased oxidative stress and increased expression of pro-inflammatory markers interleukin (IL)-6, IL-18, and tumor necrosis factor alpha (TNF-α). Both oxidative stress and inflammation contributed to an increase in microtubule-associated protein 1 light chain 3 (LC3)-II levels and heat shock response by inhibiting the mammalian target of rapamycin (mTOR) in the PKD1 cKO mouse myocytes. These findings revealed a previously unknown role for PKD1 in regulating diastolic passive properties, myofilament Ca2+ sensitivity, and maximum Ca2+-activated tension under conditions of oxidative stress. Finally, we emphasized the importance of PKD1 in maintaining the balance of oxidative stress and inflammation in the context of autophagy, as well as cardiomyocyte function.
Article
Full-text available
Systemic inflammation and coronary microvascular endothelial dysfunction are essential pathophysiological factors in heart failure (HF) with preserved ejection fraction (HFpEF) that support the use of statins. The pleiotropic properties of statins, such as anti-inflammatory, antihypertrophic, antifibrotic, and antioxidant effects, are generally accepted and may be beneficial in HF, especially in HFpEF. Numerous observational clinical trials have consistently shown a beneficial prognostic effect of statins in patients with HFpEF, while the results of two larger trials in patients with HFrEF have been controversial. Such differences may be related to a more pronounced impact of the pleiotropic properties of statins on the pathophysiology of HFpEF and pro-inflammatory comorbidities (arterial hypertension, diabetes mellitus, obesity, chronic kidney disease) that are more common in HFpEF. This review discusses the potential mechanisms of statin action that may be beneficial for patients with HFpEF, as well as clinical trials that have evaluated the statin effects on left ventricular diastolic function and clinical outcomes in patients with HFpEF.
Article
BACKGROUND A healthy heart is able to modify its function and increase relaxation through posttranslational modifications of myofilament proteins. While there are known examples of serine/threonine kinases directly phosphorylating myofilament proteins to modify heart function, the roles of tyrosine (Y) phosphorylation to directly modify heart function have not been demonstrated. The myofilament protein TnI (troponin I) is the inhibitory subunit of the troponin complex and is a key regulator of cardiac contraction and relaxation. We previously demonstrated that TnI-Y26 phosphorylation decreases calcium-sensitive force development and accelerates calcium dissociation, suggesting a novel role for tyrosine kinase–mediated TnI-Y26 phosphorylation to regulate cardiac relaxation. Therefore, we hypothesize that increasing TnI-Y26 phosphorylation will increase cardiac relaxation in vivo and be beneficial during pathological diastolic dysfunction. METHODS The signaling pathway involved in TnI-Y26 phosphorylation was predicted in silico and validated by tyrosine kinase activation and inhibition in primary adult murine cardiomyocytes. To investigate how TnI-Y26 phosphorylation affects cardiac muscle, structure, and function in vivo, we developed a novel TnI-Y26 phosphorylation-mimetic mouse that was subjected to echocardiography, pressure-volume loop hemodynamics, and myofibril mechanical studies. TnI-Y26 phosphorylation-mimetic mice were further subjected to the nephrectomy/deoxycorticosterone acetate model of diastolic dysfunction to investigate the effects of increased TnI-Y26 phosphorylation in disease. RESULTS Src tyrosine kinase is sufficient to phosphorylate TnI-Y26 in cardiomyocytes. TnI-Y26 phosphorylation accelerates in vivo relaxation without detrimental structural or systolic impairment. In a mouse model of diastolic dysfunction, TnI-Y26 phosphorylation is beneficial and protects against the development of disease. CONCLUSIONS We have demonstrated that tyrosine kinase phosphorylation of TnI is a novel mechanism to directly and beneficially accelerate myocardial relaxation in vivo.
Article
Full-text available
An altered cardiac myofilament response to activating Ca2+ is a hallmark of human heart failure. Phosphorylation of cardiac troponin I (cTnI) is critical in modulating contractility and Ca2+ sensitivity of cardiac muscle. cTnI can be phosphorylated by protein kinase A (PKA) at Ser22/23 and protein kinase C (PKC) at Ser22/23, Ser42/44, and Thr143. Whereas the functional significance of Ser22/23 phosphorylation is well understood, the role of other cTnI phosphorylation sites in the regulation of cardiac contractility remains a topic of intense debate, in part, due to the lack of evidence of in vivo phosphorylation. In this study, we utilized top-down high resolution mass spectrometry (MS) combined with immunoaffinity chromatography to determine quantitatively the cTnI phosphorylation changes in spontaneously hypertensive rat (SHR) model of hypertensive heart disease and failure. Our data indicate that cTnI is hyperphosphorylated in the failing SHR myocardium compared with age-matched normotensive Wistar-Kyoto rats. The top-down electron capture dissociation MS unambiguously localized augmented phosphorylation sites to Ser22/23 and Ser42/44 in SHR. Enhanced Ser22/23 phosphorylation was verified by immunoblotting with phospho-specific antibodies. Immunoblot analysis also revealed up-regulation of PKC-α and -δ, decreased PKCϵ, but no changes in PKA or PKC-β levels in the SHR myocardium. This provides direct evidence of in vivo phosphorylation of cTnI-Ser42/44 (PKC-specific) sites in an animal model of hypertensive heart failure, supporting the hypothesis that PKC phosphorylation of cTnI may be maladaptive and potentially associated with cardiac dysfunction.
Article
Full-text available
Homeostasis of cardiac function requires significant adjustments in sarcomeric protein phosphorylation. The existence of unique peptides in cardiac sarcomeres, which are substrates for a multitude of kinases strongly supports this concept (1) We focus here on the troponin complex of the thin filaments, which contain two major proteins that participate in these phosphoryl group transfer reactions: the inhibitory protein (cTnI2), and the Tm-binding protein (cTnT). We describe relatively new understanding of the molecular mechanisms of thin filament based control of the heart beat and how these mechanisms are altered by phosphorylation. We also discuss new concepts regarding the relation between the beat of the heart and the location of thin filament proteins, and their long and short range interactions. One of the most intriguing ideas is that the troponin complex is not only active in driving a relaxed state, but also significantly involved in driving the active state of the thin filaments. This idea is quite different from the text book view of the role of Tn in switching thin filaments on and off. These new concepts affect our understanding of how phosphorylation may modify the intensity and the dynamics of the heart beat. We also discuss elucidation of mechanisms by with these phosphorylations exacerbate or ameliorate effects of mutations in the myofilament proteins that are linked to familial cardiomyopathies.
Article
Full-text available
Hypertrophic cardiomyopathy (HCM) is characterized by left ventricular hypertrophy, increased ventricular stiffness and impaired diastolic filling. We investigated to what extent myocardial functional defects can be explained by alterations in the passive and active properties of human cardiac myofibrils. Skinned ventricular myocytes were prepared from patients with obstructive HCM (two patients with MYBPC3 mutations, one with a MYH7 mutation, and three with no mutation in either gene) and from four donors. Passive stiffness, viscous properties, and titin isoform expression were similar in HCM myocytes and donor myocytes. Maximal Ca(2+)-activated force was much lower in HCM myocytes (14 ± 1 kN/m(2)) than in donor myocytes (23 ± 3 kN/m(2); P<0.01), though cross-bridge kinetics (k(tr)) during maximal Ca(2)(+) activation were 10% faster in HCM myocytes. Myofibrillar Ca(2)(+) sensitivity in HCM myocytes (pCa(50)=6.40 ± 0.05) was higher than for donor myocytes (pCa(50)=6.09 ± 0.02; P<0.001) and was associated with reduced phosphorylation of troponin-I (ser-23/24) and MyBP-C (ser-282) in HCM myocytes. These characteristics were common to all six HCM patients and may therefore represent a secondary consequence of the known and unknown underlying genetic variants. Some HCM patients did however exhibit an altered relationship between force and cross-bridge kinetics at submaximal Ca(2+) concentrations, which may reflect the primary mutation. We conclude that the passive viscoelastic properties of the myocytes are unlikely to account for the increased stiffness of the HCM ventricle. However, the low maximum Ca(2+)-activated force and high Ca(2+) sensitivity of the myofilaments are likely to contribute substantially to any systolic and diastolic dysfunction, respectively, in hearts of HCM patients.
Article
Full-text available
Phosphodiesterase type 5A (PDE5A) inhibitors acutely suppress beta-adrenergic receptor (beta-AR) stimulation in left ventricular myocytes and hearts. This modulation requires cyclic GMP synthesis via nitric oxide synthase (NOS)-NO stimulation, but upstream and downstream mechanisms remain un-defined. To determine this, adult cardiac myocytes from genetically engineered mice and controls were studied by video microscopy to assess sarcomere shortening (SS) and fura2-AM fluorescence to measure calcium transients (CaT). Enhanced SS from isoproterenol (ISO, 10 nM) was suppressed >or=50% by the PDE5A inhibitor sildenafil (SIL, 1 microM), without altering CaT. This regulation was unaltered despite co-inhibition of either the cGMP-stimulated cAMP-esterase PDE2 (Bay 60-7550), or cGMP-inhibited cAMP-esterase PDE3 (cilostamide). Thus, the SIL response could not be ascribed to cGMP interaction with alternative PDEs. However, genetic deletion (or pharmacologic blockade) of beta3-ARs, which couple to NOS signaling, fully prevented SIL modulation of ISO-stimulated SS. Importantly, both PDE5A protein expression and activity were similar in beta3-AR knockout (beta3-AR(-/-)) myocytes as in controls. Downstream, cGMP stimulates protein kinase G (PKG), and we found contractile modulation by SIL required PKG activation and enhanced TnI phosphorylation at S23, S24. Myocytes expressing the slow skeletal TnI isoform which lacks these sites displayed no modulation of ISO responses by SIL. Non-equilibrium isoelectric focusing gel electrophoresis showed SIL increased TnI phosphorylation above that from concomitant ISO in control but not beta3-AR(-/-) myocytes. These data support a cascade involving beta3-AR stimulation, and subsequent PKG-dependent TnI S23, S24 phosphorylation as primary factors underlying the capacity of acute PDE5A inhibition to blunt myocardial beta-adrenergic stimulation.
Article
Mechanisms underlying diastolic dysfunction need to be better understood. To study the role of titin in diastolic dysfunction using a mouse model of experimental heart failure induced by transverse aortic constriction. Eight weeks after transverse aortic constriction surgery, mice were divided into heart failure (HF) and congestive heart failure (CHF) groups. Mechanical studies on skinned left ventricle myocardium measured total and titin-based and extracellular matrix-based passive stiffness. Total passive stiffness was increased in both HF and CHF mice, and this was attributable to increases in both extracellular matrix-based and titin-based passive stiffness, with titin being dominant. Protein expression and titin exon microarray analysis revealed increased expression of the more compliant N2BA isoform at the expense of the stiff N2B isoform in HF and CHF mice. These changes are predicted to lower titin-based stiffness. Because the stiffness of titin is also sensitive to titin phosphorylation by protein kinase A and protein kinase C, back phosphorylation and Western blot assays with novel phospho-specific antibodies were performed. HF and CHF mice showed hyperphosphorylation of protein kinase A sites and the proline glutamate valine lysine (PEVK) S26 protein kinase C sites, but hypophosphorylation of the PEVK S170 protein kinase C site. Protein phosphatase I abolished differences in phosphorylation levels and normalized titin-based passive stiffness levels between control and HF myocardium. Transverse aortic constriction-induced HF results in increased extracellular matrix-based and titin-based passive stiffness. Changes in titin splicing occur, which lower passive stiffness, but this effect is offset by hyperphosphorylation of residues in titin spring elements, particularly of PEVK S26. Thus, complex changes in titin occur that combined are a major factor in the increased passive myocardial stiffness in HF.
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
Background— Titin contains a molecular spring segment that underlies passive myocardial stiffness. Myocardium coexpresses titin isoforms with molecular spring length variants and, consequently, distinct stiffness characteristics: the stiff N2B isoform (short spring) and more compliant N2BA isoform (long spring). We tested whether changes in titin isoform expression occur in the diastolic dysfunction that accompanies heart failure. Methods and Results— We used the tachycardia-induced dilated cardiomyopathy canine model (4-week pacing) and found that control myocardium coexpresses the N2B and N2BA isoforms at similar levels, whereas in dilated cardiomyopathy the expression ratio had shifted, without affecting the amount of total titin, toward more prominent N2B expression. This shift was accompanied by elevated titin-based passive muscle stiffness. Pacing also resulted in significant upregulation of obscurin, an ≈800-kDa elastic protein with several signaling domains. Conclusions— Coexpression of titin isoforms with distinct mechanical properties allows modulation of passive stiffness via adjustment of the isoform expression ratio. The canine pacing-induced heart failure model uses this mechanism to increase myocardial stiffness. Thus, changes in titin isoform expression may play a role in diastolic dysfunction in heart failure.
Article
In vitro studies suggest that phosphorylation of titin reduces myocyte/myofiber stiffness. Titin can be phosphorylated by cGMP-activated protein kinase. Intracellular cGMP production is stimulated by B-type natriuretic peptide (BNP) and degraded by phosphodiesterases, including phosphodiesterase-5A. We hypothesized that a phosphodiesterase-5A inhibitor (sildenafil) alone or in combination with BNP would increase left ventricular diastolic distensibility by phosphorylating titin. Eight elderly dogs with experimental hypertension and 4 young normal dogs underwent measurement of the end-diastolic pressure-volume relationship during caval occlusion at baseline, after sildenafil, and BNP infusion. To assess diastolic distensibility independently of load/extrinsic forces, the end-diastolic volume at a common end-diastolic pressure on the sequential end-diastolic pressure-volume relationships was measured (left ventricular capacitance). In a separate group of dogs (n=7 old hypertensive and 7 young normal), serial full-thickness left ventricular biopsies were harvested from the beating heart during identical infusions to measure myofilament protein phosphorylation. Plasma cGMP increased with sildenafil and further with BNP (7.31±2.37 to 26.9±10.3 to 70.3±8.1 pmol/mL; P<0.001). Left ventricular diastolic capacitance increased with sildenafil and further with BNP (51.4±16.9 to 53.7±16.8 to 60.0±19.4 mL; P<0.001). Changes were similar in old hypertensive and young normal dogs. There were no effects on phosphorylation of troponin I, troponin T, phospholamban, or myosin light chain-1 or -2. Titin phosphorylation increased with sildenafil and BNP, whereas titin-based cardiomyocyte stiffness decreased. Short-term cGMP-enhancing treatment with sildenafil and BNP improves left ventricular diastolic distensibility in vivo, in part by phosphorylating titin.
Article
We discuss a current controversy regarding the relative role of phosphorylation sites on cardiac troponin I (cTnI) (Fig. 1) in physiological and patho-physiological cardiac function. Studies with mouse models and in vitro studies indicate that multi-site phosphorylations are involved in both control of maximum tension and sarcomeric responsiveness to Ca(2+). Thus one hypothesis is that cardiac function reflects a balance of cTnI phosphorylations and a tilt in this balance may be maladaptive in acquired and genetic disorders of the heart. Studies on human heart samples taken mainly at end-stage heart failure, and in depth proteomic analysis of human and rat heart samples demonstrate that Ser23/Ser24 are the major and perhaps the only sites likely to be relevant to control cardiac function. Thus functional significance of Ser23/Ser24 phosphorylation is taken as fact, whereas the function of some other sites is treated as fancy. Maybe the extremes will meet: in any case we both agree that further work needs to be carried out with relatively large mammals and with determination of the time course of changes in phosphorylation to identify transient modifications that may be relevant at a beat-to-beat basis. Moreover, we agree that the changes and effects of cTnI phosphorylation need to be fully integrated into the effects of other phosphorylations in the cardiac myocyte.