Download 10 Chapter General Discussion

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Electrocardiography wikipedia , lookup

Hypertrophic cardiomyopathy wikipedia , lookup

Remote ischemic conditioning wikipedia , lookup

Heart failure wikipedia , lookup

Antihypertensive drug wikipedia , lookup

Cardiac surgery wikipedia , lookup

Cardiac contractility modulation wikipedia , lookup

Coronary artery disease wikipedia , lookup

Management of acute coronary syndrome wikipedia , lookup

Quantium Medical Cardiac Output wikipedia , lookup

Transcript
Chapter
10
General Discussion
Summary
Future Perspectives
General discussion
Heart failure with preserved ejection fraction (HFpEF) is widely prevalent and has a
cumbersome prognosis, while its pathophysiological mechanisms are incompletely
understood and specific therapeutic strategies remain uncertain1-3. It is still debated whether
HFpEF and HF with reduced EF (HFrEF) represent distinct HF phenotypes4. However,
prominent differences in patient demographics, risk factor profile, hemodynamic
characteristics and macroscopic and myocellular remodeling coupled to disparate responses to
similar HF pharmacotherapy would all seem to justify that HFpEF and HFrEF indeed
represent distinct HF phenotypes2,5-7. Elevated diastolic left ventricular (LV) stiffness is an
important feature in HFpEF8-10. Numerous studies, of which several are included in the
present thesis, demonstrated that raised intrinsic cardiomyocyte stiffness (Fpassive) importantly
contributes to high diastolic LV stiffness in HFpEF5,11-14. Cardiomyocyte Fpassive is mainly
determined by the giant elastic sarcomeric protein titin, which regulates myocardial passive
tension, stiffness and biomechanical stress/stretch signaling15-17. Titin spans a half sarcomere,
running from the Z-disk to the M-band and functions as a bidirectional spring responsible for
early diastolic recoil and late diastolic resistance to stretch15-17. Cardiomyocyte titin-based
elasticity can be adjusted through transcriptional and posttranslational modifications. At the
transcriptional level, titin modulates stiffness through shifts in expression of its compliant
N2BA (3.2-3.7 MDa) and stiff N2B (3.0MDa) isoforms, which are co-expressed in the
sarcomere at a N2BA:N2B ratio of approximately 35:65 in the normal heart15-17. In
eccentrically remodeled explanted hearts from dilated18,19 or ischemic cardiomyopathy20
patients, as well as in LV endomyocardial biopsies from HFrEF patients5,13, the N2BA:N2B
expression ratio was increased, resulting in reduced myocardial stiffness. In contrast, the
N2BA:N2B expression ratio was decreased in LV endomyocardial biopsies from HFpEF
patients compared to HFrEF5,13, whereas mixed results were reported in LV biopsies from
patients with severe AS, ranging from either reduced21 or similar22 N2BA:N2B expression
ratios compared to donor hearts. While titin-based stiffness may be altered by titin isoform
switching within days or weeks, titin-based stiffness can also be modulated acutely through
posttranslational modifications, such as titin isoform phosphorylation16.
Both protein kinases A and G (PKA and PKG) phosphorylate the stiff titin N2B isoform
thereby acutely lowering titin-based cardiomyocyte stiffness, which has been demonstrated in
skinned human LV muscle strips23,24, in isolated human LV myofibrils24 and in isolated
cardiomyocytes from HFpEF and HFrEF patients2,11,13,14. Two additional posttranslational
modifications of titin, both increasing titin-based stiffness, include protein kinase C α (PKCα) mediated phosphorylation of titin’s PEVK segment (rich in Proline, Glutamic acid, Valine
and Lysine)25 and oxidative stress-induced formation of disulfide bridges in the stiff N2B
segment of titin26. Contribution by other myofilamentary proteins, such as myosin heavy
chain, desmin, actin, troponin T, troponin I (TnI), myosin light chain 1 and -2 was recently
ruled out11,13, suggesting that phosphorylation-mediated posttranslational modification of titin
indeed represents the most likely mechanism for acute modulation of cardiomyocyte Fpassive.
Intrinsic cardiomyocyte stiffness is significantly higher in patients with HFpEF than in
controls, in patients with HFrEF and in patients operated for severe aortic stenosis (AS)5,11-14.
Because the fall in cardiomyocyte resting tension following in-vitro administration of PKA
and PKG is significantly larger in HFpEF than in HFrEF and AS5,12-14, a larger
phosphorylation deficit of presumably titin could be present in HFpEF as recently
suggested13. Indeed, stimulation of PKG activity through inhibited breakdown of cyclic
guanosine monophosphate (cGMP) by phosphodiesterase type 5A (PDE5A)27 by the PDE5A
inhibitor sildenafil, was shown to ameliorate diastolic dysfunction in various experimental
and clinical studies. Sildenafil restored LV relaxation kinetics in mice exposed to transverse
aortic constriction (TAC)28 and reduced diastolic LV stiffness in an old hypertensive dog
model29, in patients with HFrEF30 and in HFpEF patients with pulmonary hypertension31.
Administration of sildenafil to old hypertensive dogs lowered diastolic LV stiffness through
restored phosphorylation of the titin N2B segment29. Moreover, PDE5A inhibitors have also
been shown to attenuate adrenergic stimulation32, reduce ventricular-vascular stiffening33,
improve endothelial function34, reduce pulmonary vascular resistance35 and enhance exercise
tolerance35,36 in HFrEF. Recently, we measured PKG activity, upstream control of PKG
activity and downstream effects of PKG activity in LV myocardial biopsies of HFpEF
patients14. To discern altered control by PKG of the myocardial remodeling process in
HFpEF, measurements were compared to measurements obtained from LV myocardium
remodeled concentrically by severe AS or eccentrically by nonischemic HFrEF. Our study
showed that relative to both AS and HFrEF, HFpEF patients had reduced myocardial PKG
activity and lower cGMP concentration, which related to higher cardiomyocyte Fpassive and
increased myocardial nitrotyrosine levels, indicative of raised nitrosative/oxidative stress. In
all groups, cardiomyocyte Fpassive was acutely lowered by in vitro administration of PKG,
whereas cardiomyocytes from HFpEF patients demonstrated the largest PKG-induced fall in
Fpassive14. Reduced PKG activity and lower myocardial cGMP concentration in HFpEF did not
result from altered myocardial soluble guanylate cyclase (sGC) or PDE5A expression, which
were similar in all groups, or from unequal brain type natriuretic peptide (BNP) expression,
which was comparable in HFpEF and AS. Because of comparable proBNP-108 expression in
HFpEF and AS, BNP is unlikely to account for the widely different PKG activities and cGMP
concentrations observed in both conditions14. Lower proBNP-108 expression in HFpEF than
in HFrEF also explains the low positive predictive value of BNP for the diagnosis of
HFpEF37. Downregulation of cGMP-PKG signaling in HFpEF was therefore most likely
related to low myocardial NO bioavailability because of high nitrosative/oxidative stress,
which was almost fourfold higher in HFpEF than in both HFrEF and AS14. Oxidative stress is
known to lower NO bioavailability and downregulate cGMP-PKG signaling38-41.
Importance of NO for diastolic function
LV distensibility
Endothelial dysfunction plays a prominent role in the pathophysiology and prognosis of HF42.
Impaired endothelium-dependent vasodilation in HF patients has been attributed to reduced
activity of the L-arginine-NO synthetic pathway, increased degradation of NO by reactive
oxygen species (ROS) and hyporesponsiveness in vascular smooth muscle43,44.
NO is crucially important for diastolic function, since it potently enhances LV relaxation and
distensibility through cGMP-PKG dependent and independent mechanisms, including
reduction of myofilament calcium (Ca2+) sensitivity by TnI phosphorylation and enhancing
phospolamban-mediated sarcoplasmic reticular (SR) Ca2+ reuptake, respectively45-47. NO is an
ubiquitous intra- and intercellular signaling molecule generated in a temporally and spatially
restricted manner by a family of NO synthases (NOSs), including endothelial NOS (eNOS),
neuronal NOS (nNOS) and inducible NOS (iNOS), which vary in their subcellular
localization46,47. The myocardial autocrine and paracrine effects of NO depend on where and
by which NOS isoform NO is produced46. Myocardial eNOS-derived NO was demonstrated
to enhance LV relaxation and distensibility48-50 and to modulate cardiac β-adrenergic
responsiveness51. Stimulation of eNOS activity by intracoronary administration of NO donors
instantaneously hastened LV relaxation, accelerated LV pressure decline and increased
diastolic LV distensibility in the normal human heart48 as well as in patients with dilated
cardiomyopathy49 and severe AS50. The importance of myocardial NO bioavailability for
diastolic LV function was recently reappraised because of close correlations between
asymmetric dimethylarginine and diastolic LV dysfunction in patients with ischemic and nonischemic dilated cardiomyopathy52,53.
In addition, myocardial endothelial eNOS-derived NO regulates cardiac metabolism through
modulation of myocardial substrate utilization and by reduction of myocardial oxygen
consumption through inhibition of complexes I and II of the mitochondrial electron transport
chain46,54. Interestingly, mitochondrial biogenesis is stimulated in response to increased
myocardial NO bioavailability and cGMP levels55. Because cross-bridge detachment is an
energy consuming process, slow LV relaxation in HFpEF could therefore also result from a
myocardial energy deficit because of reduced myocardial NO bioavailability due to increased
nitrosative/oxidative stress burden. Recent 31P-magnetic resonance spectroscopy studies
indeed demonstrated reduced myocardial energy reserve in patients with HFpEF56-58, HFrEF59
and in both diabetic60 and obese61 patients, in whom impaired myocardial bioenergetics
correlated with diastolic LV dysfunction60,61.
NO-mediated activation of cGMP-PKG signaling
NO activates sGC, which is present predominantly in the cytosol and in smaller amounts at
the plasma membrane, via a complex interplay between binding of NO to both heme and nonheme sites of sGC47,62,63. Stimulated sGC subsequently enhances cGMP generation, which
activates its primary effector kinase PKG27,47. In turn, PKG phosphorylates a vast number of
target proteins, exerting a wide range of downstream effects like enhanced SR Ca2+ reuptake,
inhibition of Ca2+ influx and suppression of pro-hypertrophic and fibrotic signaling as well as
stimulation of relaxation and LV distensiblity by phosphorylation of TnI and the stiff titin
N2B segment16,24,27,64. Because NO diffusion distances in cardiomyocytes are limited, NOsGC-cGMP signaling is compartmentalized into functional signalosomes, coupling NO
synthesis to downstream sGC-cGMP signals47. For instance, in cardiomyocytes and
endothelial cells, eNOS, sGC and PKG colocalize with caveolin at plasma membrane
invaginations, called caveolae, which may serve as microdomains for optimized NO-sGCcGMP signaling47,63. Furthermore, besides being subject to intricate intracellular
compartmentalization, signaling through the eNOS-NO-cGMP-PKG pathway is also
characterized by considerable crosstalk with β-adrenergic mediated cAMP signaling,
providing the heart with a complex and highly differentially regulated machinery to modulate
and integrate divergent cardiovascular (patho)physiological conditions47,65-67.
Importance of cGMP-PKG signaling in myocardial remodelling
Cardiac remodeling in HFpEF is characterized by concentric LV and cardiomyocyte
hypertrophy and interstitial fibrosis5,9,68-70. The Framingham Heart Study determined that LV
hypertrophy follows aging as independent risk factor for cardiovascular morbidity and
mortality71. Stimuli for LV hypertrophy activate an array of membrane-bound receptors
coupled to multiple intracellular signaling cascades, which ultimately stimulate prohypertrophic gene expression72,73. Important upstream triggers for cardiac hypertrophy
include mechanical stretch, pressure overload, cytokines, activation of RAAS, endothelin and
catecholamines, whereas cGMP-PKG signaling exerts prominent anti-hypertrophic and antifibrotic effects27,72,73. Indeed, modulation of the sGC-PKG-PDE5A axis affected myocardial
remodeling with less cardiomyocyte hypertrophy and interstitial fibrosis in TAC mice
exposed to sildenafil28. Despite similar levels of LV and cardiomyocyte hypertrophy in
patients with HFpEF and AS, HFpEF patients demonstrated higher cardiomyocyte Fpassive and
lower myocardial PKG activity than AS patients14. The finding in AS, of higher PKG activity
corresponding with low Fpassive but not with cardiomyocyte diameter, suggests cardiomyocyte
diastolic stiffness to evolve independently from cardiomyocyte hypertrophy. This concept
also emerged from the VALIDD trial, in which only 3% of hypertensives had significant LV
hypertrophy despite all having diastolic LV dysfunction74. External myocardial force signals
imposed during hemodynamic load are transmitted to ECM components and the
cardiomyocytes15. Mechanical stimuli sensed at the outer cardiomyocyte cell membrane can
activate cytoskeletal and sarcomeric signaling cascades, which subsequently trigger prohypertrophic factors15. In addition to its prominent role in regulating cardiomyocyte stiffness,
titin also acts as a dominant signaling protein, which integrates both internally generated
sarcomeric forces and externally sensed biomechanical stretch/stress signals to hypertrophy
regulation75. Thus, it could be speculated that increased cardiomyocyte stiffness sensed by
titin precedes titin-mediated stimulation of pro-hypertrophic signaling.
Nitrosative/oxidative stress lowers NO bioavailability and NO signalling in
HFpEF
Reduced NO bioavailability, dysregulation of NO-mediated signaling and increased
nitrosative/oxidative stress are firmly implicated in the pathogenesis of HF40,41,76-78. Although
generation of ROS (i.e. superoxide, hydroxyl radical and non-radical species such as
hydrogen peroxide) is a normal component of cellular redox-based signaling, excessive ROS
production causes nitrosative/oxidative stress40,41,76-79.
Nitrosative/oxidative stress impairs NO bioavailability and NO-mediated signaling through a
number of mechanisms. First, superoxide rapidly reacts with NO to form peroxynitrite, which
triggers eNOS uncoupling with subsequent generation of superoxide instead of NO, thereby
augmenting oxidative stress. Second, by scavenging NO, superoxide reduces NO
bioavailability and prevents NO from stimulating sGC-mediated activation of cGMP-PKG
signaling37,40,41. Finally, sGC activity is adversely affected by nitrosative/oxidative stress
through several mechanisms, including superoxide and peroxynitrite mediated decrease in
sGC activity and sGC heme group oxidation converting sGC to its NO-insensitive state40,63.
The lower myocardial PKG activity and reduced cGMP concentration in HFpEF patients,
which was associated with increased myocardial nitrotyrosine levels14, could therefore have
resulted from upstream nitrosative/oxidative stress-induced impairments of NO bioavailability
and reduced NO-mediated activation of sGC-induced cGMP/PKG signaling. Accordingly, a
number of recent studies also suggested oxidative stress to be involved in the pathogenesis of
diastolic LV dysfunction in HFpEF. In HFpEF rodent models, oxidative stress uncoupled
cardiac NOS and induced diastolic LV dysfunction, whereas stimulation of eNOS activity
reversed oxidative stress and improved diastolic LV function80,81. Furthermore, high plasma
levels of methylated L-arginine metabolites, indicative of increased oxidative stress, were
strongly related to diastolic LV dysfunction in patients with HFrEF52. Moreover, in patients
with HFpEF, myocardial fibrosis and inflammation were associated with diastolic LV
dysfunction and with increased oxidative stress in endomyocardial biopsies compared to
controls82. Abundant evidence demonstrates that ROS potently activate pro-hypertrophic and
pro-fibrotic signaling, and by interfering with NO-sGC-cGMP-PKG signalling, ROS inhibit
anti-hypertrophic and anti-fibrotic signalling cascades41,72.
In addition, nitrosative/oxidative stress induces a state of NO/redox disequilibrium, which
impairs S-nitrosylation signaling (a posttranslational protein modification process through
coupling of an NO moiety to a reactive cysteine thiol). S-nitrosylation serves as a major
effector of NO bioactivity and an important mode of cellular signal transduction, regulating
the activity of numerous metabolic enzymes, kinases, phosphatases and respiratory proteins as
well as cytoskeletal and structural components and transcription factors41,76,83.
Possible mechanisms for enhanced nitrosative/oxidative stress in HFpEF
It was recently suggested that the particularly high prevalence of metabolic risk factors in
HFpEF could be associated with increased inflammation and nitrosative/oxidative stress in
this condition14. HFpEF patients demonstrate a high prevalence of obesity and DMII84-86.
Obesity and DMII are strongly related to insulin resistance (IR) and the metabolic syndrome
(a constellation of cardiovascular risk factors, including obesity, hypertension, IR,
hyperglycemia, dyslipidemia, microalbuminuria and hypercoagulability)87 and the frequent
clustering of these metabolic risk factors causes synergistic adverse effects on myocardial
structure and function88. The prevalence of obesity, DMII, IR and metabolic syndrome
increases rapidly and is expected to reach pandemic proportions in the next few decades88-92,
while these metabolic risk factors have all been prospectively identified as precursors of
incident HF93-95 and are independently associated with early development of diastolic LV
dysfunction96-99. Diastolic LV dysfunction is recognized as the earliest manifestation of
DMII-induced LV dysfunction96,100-104, while the excessive diastolic LV stiffness of the
diabetic heart has been related to myocardial fibrosis105 and to circulating advanced glycation
endproducts (AGEs)106. In a recent study, DM increased diastolic LV stiffness in both HFpEF
and HFrEF patients through distinct mechanisms12. In diabetic HFrEF, raised diastolic
stiffness was associated with both increased CVF and myocardial deposition of AGEs.
Conversely, in diabetic HFpEF, myocardial CVF was unchanged and elevated diastolic
stiffness related to increased cardiomyocyte Fpassive12. Diabetes also increases LV diastolic
and cardiomyocyte stiffness in patients with severe AS102 and elevated cardiomyocyte
stiffness in diabetic AS patients related to hypophosphorylation of the titin N2B segment,
which could be corrected by phosphorylation with PKA22.
Obesity, DMII and IR can have direct adverse effects on myocardial structure and function
independently from common confounders as hypertension or CAD, which has been referred
to as “obesity”107, “diabetic”102, or “insulin-resistant”108 cardiomyopathy. Moreover,
metabolic risk factors are strongly associated with endothelial dysfunction, inflammation,
oxidative stress, impaired myocardial energetics, abnormal cardiomyocyte Ca2+ handling,
reduced NO bioavailability and maladaptive cardiac remodelling102,107,108.
Metabolic risk factors interfere with NO-cGMP-PKG signaling
Oxidative stress-mediated downregulation of NO-cGMP-PKG signaling has been
demonstrated in various experimental models of obesity, diabetes, IR and metabolic
syndrome109,110. Cultured rodent aortic smooth muscle cells subjected to hyperglycaemia
demonstrated significantly reduced PKG expression and activity, which occurred through a
PKC-dependent activation of NADPH oxidase-derived superoxide production109. In this
model, high glucose-mediated lowering of PKG levels was inhibited by a superoxide
scavenger or NADPH oxidase inhibitors109. In addition, in human diabetic cardiomyopathy,
sildenafil improved measurements of LV geometry, strain and torsion determined by
magnetic resonance imaging111. Furthermore, in a rodent model of diet-induced obesity and
IR, signaling through the NO-cGMP pathway was reduced, while vascular inflammation and
IR were completely prevented by administration of sildenafil110.
Physiologically, insulin phosphorylates titin through activation of the phosphoinositol-3kinase (PI3K)/Akt/eNOS pathway, while IR, FFAs and adipokines inhibit insulin-dependent
PI3K/Akt signaling, resulting in reduced eNOS activity and impaired NO generation112,113.
Inflammation and oxidative stress associated with metabolic risk factors can reduce eNOS
activity and NO bioavailability through a number of ways, including RAAS-mediated
activation of ROS producing enzymes, increased flux through the hexosamine biosynthesis
pathway, mitochondrial uncoupling, activation of the polyol pathway, lipotoxicity, glucose
auto-oxidation and formation of AGEs113. Moreover, obesity, DM and IR induce activation of
multiple PKC isoforms, with PKCs being prominently involved in numerous cellular
functions and signal transduction pathways114. PKC can hamper myocardial eNOS activity
and NO generation by inhibition of PI3K/Akt signaling and stimulation of NADPH oxidase
activity114, whereas PKCα was recently shown to increase titin-based cardiomyocyte stiffness
through phosphorylation of the PEVK segment25. Interestingly, in a rodent diabetic HFpEF
model, PKCβ inhibition improved diastolic distensiblity, contractility and maladaptive cardiac
remodelling115. Recent evidence demonstrates that in addition to downregulation of NOcGMP-PKG signaling, metabolic conditions such as IR, metabolic syndrome and DM also
induce a state of myocardial energy deficiency with decreased mitochondrial oxidative
phosphorylation and diminished mitochondrial biogenesis, which predisposes to a metabolic
cardiomyopathy characterized by early development of diastolic LV dysfunction116,117.
In summary, metabolic risk factors could contribute to diastolic LV dysfunction and elevated
myocardial stiffness in HFpEF through provoking myocardial inflammation and
nitrosative/oxidative stress, which on its turn results in decreased NO bioavailability,
downregulation of cGMP-PKG signaling, impaired myocardial bioenergetics and maladaptive
remodeling. Furthermore, downregulation of cGMP-PKG signaling could subsequently result
in hypophosphorylation of titin, which elevates cardiomyocyte stiffness and possibly triggers
cardiomyocyte hypertrophy.
Novel treatment strategies for HFpEF
In HFpEF, the reduced myocardial PKG activity, increased nitrosative/oxidative stress and the
presence of myocardial fibrosis, impaired myocardial energetics and the high prevalence of
metabolic risk factors supports use of NO-donors, PDE5A inhibitors, statins, anti-fibrotic
agents, such as aldosterone antagonists and optimized treatment of metabolic risk.
Stimulation of NO-cGMP-PKG signaling
NOS uncoupling has emerged as an important contributor to pathologic chamber remodeling,
endothelial dysfunction and diastolic dysfunction in animal models of HF80,81. Oxidative
depletion of the essential NOS cofactor tetrahydrobiopterin (BH4), results in NOS
destabilization and uncoupling subsequently causing NOS to generate superoxide instead of
NO118. Administration of BH4 in animal models reduces pressure-overload hypertrophy,
fibrosis, NOS uncoupling and oxidative stress, while at the same time systolic and diastolic
function are improved80,81,119, suggesting that targeting NOS uncoupling could improve
diastolic LV dysfunction in HFpEF. Exogenous administration of BH4 has also been shown
to improve endothelial dysfunction in patients with hypercholesterolemia120, DM121 and
hypertension122 through improvement of NOS function and enhanced NO bioactivity coupled
with BH4 treatment.
Acute administration of NO donors is known to improve diastolic LV function with an earlier
onset of LV relaxation, lower LV peak systolic, end-systolic and end-diastolic pressures, with
rightward displacement of the diastolic LV pressure-volume relation45,48.
Current ESC HF guidelines accord a class IIa indication for administration of NO donors in
patients admitted for acute HF with pulmonary edema without concomitant cardiogenic
shock123. Unfortunately, longterm use of NO donors is frequently hampered by development
of NO resistance124. NO resistance largely results from a combination of scavenging of NO by
superoxide and of (reversible) inactivation of sGC125. Conversely, chronic use of isosorbide
dinitrate combined with the antioxidant hydralazine improved outcome in V-HeFT I and AHeFT trials126,127. Hydralazine reduces superoxide generation by xanthine oxidase and
NADPH oxidase128. The clinical characteristics of the A-HeFT HFrEF patients revealed a
high prevalence of obesity and DM129, conditions which are also highly prevalent in HFpEF.
Combined use of isosorbide dinitrate and the antioxidant hydralazine could therefore be
potentially favourable in HFpEF.
An exciting promising new inroad for HFpEF treatment is represented by agents that enhance
cellular cGMP signaling27. Upstream activation of sGC by NO stimulates cGMP-PKG
activation in the cytosolic and subsarcolemmal compartments, while upstream activation of
particulate GC (pGC) by natriuretic peptides stimulates cGMP-PKG signaling at the plasma
membrane63,65. Several key transcription factors and sarcomeric proteins involved in
hypertrophy signaling, diastolic relaxation and stiffness and vasorelaxation are favourably
modified by PKG-dependent phosphorylation, suggesting that these agents may be beneficial
in HFpEF13,24,27,28,48. PDE5A inhibitors, such as sildenafil, increase cGMP levels by blocking
their catabolism. PDE5A inhibitors attenuate adrenergic stimulation32, reduce ventricularvascular stiffening32, antagonize maladaptive chamber remodeling28,30, improve endothelial
function34, reduce pulmonary vascular resistance30,31,35 and lower diastolic LV stiffness in
patients with HFrEF30 and in HFpEF patients with pulmonary hypertension31.
An alternative approach to enhance cGMP-PKG signaling is through direct stimulation of
sGC activity. Recently, two classes of drugs have been discovered, the sGC activators and
sGC stimulators, which target two different redox states of sGC: the NO-sensitive reduced
(ferrous) sGC and NO-insensitive oxidized (ferric) sGC respectively130. sGC consists of an
α/β-heterodimeric protein with a prosthetic ferrous heme group. The presence of a reduced
Fe2+ (ferrous) heme group is crucial for NO-sensing and NO-dependent sGC stimulation130.
The heme group can exist in different redox states, which may enable sGC to modulate
intracellular redox homeostasis in addition to its NO-sensing capability130. Oxidative stress
favours heme-free sGC, which is unresponsive to NO. Hence, oxidative stress synergistically
hampers NO-cGMP signaling through sGC oxidation and through ROS-mediated scavenging
of NO38,40 thereby compromising NO-sGC-cGMP mediated signaling130-132. The sGC
stimulators have a dual mode of action; they sensitize sGC to low levels of NO and can
stimulate sGC directly in the absence of any endogenous NO. Conversely, sGC activators
specifically activate the NO-unresponsive, heme-free form of the enzyme irrespective of NO
bioavailability130-133.
In a nonrandomized proof of concept study, the direct NO- and heme-independent sGC
activator cinaciguat was shown to acutely reduce pulmonary capillary and artery pressures,
lower pulmonary and systemic vascular resistance, while improving cardiac output in patients
admitted for acute decompensated HF134. A number of phase IIb studies (COMPOSE
program) evaluated the safety and efficacy of fixed low doses of intravenous cinaciguat as
add-on therapy in patients hospitalized for acute HF and demonstrated cinaciguat to be
associated with an excess of non-fatal hypotensive episodes without improvements in dyspnea
or cardiac index135. Recently, the sustained sGC stimulator (BAY 41-8543) was found to
offset NOS inhibition and to restore acute cardiac modulation by sildenafil in adult mice,
which suggests that direct sGC stimulators could be used to enhance the action of PDE5A
inhibitors in settings where critical sGC-generated cGMP is inadequate136.
Stimulation of cGMP-PKG signaling can also be achieved through NP-mediated activation of
pGC. The synthetic NP nesiritide was shown to acutely reduce pulmonary capillary wedge
pressure and systemic vascular resistance, while increasing cardiac index and stroke volume
index in HFrEF patients137. Acute NP administration was recently reported to lower diastolic
LV stiffness and to increase myocardial titin phosphorylation in an old hypertensive HFpEF
dog model29 but failed to improve clinical endpoints in a large, randomized clinical trial of
acutely decompensated HF patients with either LVEF <40% and LVEF ≥40%138. Conversely,
the ubiquitous enzyme dipeptidyl peptidase IV (DPP IV) cleaves BNP1-32 into BNP3-32, which
has reduced biological activity compared to BNP1-32139,140 and was found increased in patients
with chronic HF141. Interestingly, sitagliptin, a specific DPP IV inhibitor, preserved renal
function, modulated stroke volume and heart rate and potentiated the positive inotropic effect
of exogenous BNP in a pacing-induced pig HF model142.
Modulation of myocardial remodeling and nitrosative/oxidative stress
Prominent features of HFpEF, DM, IR and obesity are cardiac hypertrophy and fibrosis.
Hypertrophic stimuli, such as mechanical stretch, neurohumoral activation and inflammatory
markers activate various downstream pro-hypertrophic signaling pathways, whereas NO-PKG
mediated signaling exerts anti-hypertrophic and anti-fibrotic effects72,143. Furthermore,
abundant evidence demonstrates that ROS potently activate pro-hypertrophic and pro-fibrotic
pathways, and by interfering with both NO-sGC-cGMP-PKG signalling and NO-mediated Snitrosylation, ROS inhibit anti-hypertrophic and anti-fibrotic signalling cascades72,76,78,79,143.
Hence, in HFpEF, the combination of low myocardial PKG activity and nitrosative/oxidative
stress could accelerate myocardial maladaptive remodeling, because of unopposed activation
of pro-hypertrophic and pro-fibrotic signaling.
Angiotensin converting enzyme inhibitors and angiotensin receptor blockers
Angiotensin converting enzyme inhibitors (ACE-I) and angiotensin receptor blockers (ARBs)
prevent new onset DMII, ameliorate IR and inhibit formation of AGEs144-148. Moreover,
ACE-I and ARBs improve maladaptive remodeling in HFrEF149,150, reduce myocardial
fibrosis and LV mass in patients with hypertensive heart disease151,152, inhibit inflammation
and NADPH oxidase-mediated ROS production153,154 and stimulate NO production40,155,156.
Although ACE-I and ARBs improved diastolic LV dysfunction in hypertensive heart
disease157-159, their importance for HFpEF treatment has been seriously challenged, because of
the neutral outcomes of large HFpEF trials investigating ACE-I160 and ARBs161,162. A recent
large meta-analysis demonstrated divergent stimulatory effects of ACE-I on endothelial
function163, with failure of ACE-I to improve brachial flow mediated dilation in diabetic164
and obese patients165. Furthermore, in the ALLHAT study (Antihypertensive and LipidLowering Treatment to Prevent Heart Attack Trial), lisinopril was inferior to chlorthalidone in
preventing new-onset HFpEF166, while enalapril failed to improve exercise capacity or aortic
distensibility in HFpEF patients167. The reason why ACE-I and ARBs fail to improve
outcome in HFpEF patients, despite their ability to improve endothelial function remains
elusive. However, other mechanisms than RAAS activation can impair NO bioavailability and
PKG signaling in the setting of metabolic risk factors. Insulin resistance, FFA and adipokines
inhibit insulin-dependent PI3K/Akt signaling, resulting in reduced eNOS activity and
impaired NO generation113. In addition, hyperglycemia and adipokines can hamper eNOS
activity through various mechanisms, including increased flux through the hexosamine
biosynthesis pathway113. Oxidative stress in DMII, obesity and IR can be generated from
other sources than RAAS-associated ROS-producing enzymes, including mitochondrial
uncoupling, activation of the polyol pathway, lipotoxicity, glucose auto-oxidation and
formation of AGEs113,168. Furthermore, DM, obesity and IR induce activation of multiple PKC
isoforms, with PKCs being prominently involved in numerous cellular functions and signal
transduction pathways169. PKC can hamper myocardial eNOS activity and NO generation by
inhibition of PI3K/Akt signaling and stimulation of NADPH oxidase activity113,169, whereas
PKCα was recently shown to increase titin-based cardiomyocyte stiffness through
phosphorylation of the PEVK segment25. Interestingly, in a rodent diabetic HFpEF model,
PKCβ inhibition improved diastolic distensiblity, contractility and maladaptive cardiac
remodeling170. Thus, in HFpEF, increased prevalence of (frequently co-existing) metabolic
risk factors will exert complex and profound adverse effects on endothelial function and NO
bioavailability, which are mediated by synergistic contributions of inflammation, oxidative
stress, metabolic disturbances and deranged intracellular endothelial and cardiomyocyte
signaling and which could explain why isolated treatment of ACE-I and ARBs resulted in
neutral clinical outcome in the large HFpEF trials.
Aldosterone antagonists
Aldosterone plays an important role in the pathogenesis of vascular stiffening and endothelial
dysfunction, and increased plasma aldosterone levels are associated with inflammation,
oxidative stress, IR and the metabolic syndrome171-173. In addition, elevated aldosterone levels
are also related to cardiac hypertrophy, fibrosis and diastolic LV dysfunction171-173. In HFpEF
clinical trials, aldosterone antagonists improved exercise capacity and diastolic LV
stiffness174, as well as LV long axis function and cardiac geometry175. In a recent multicenter,
prospective, randomized, double-blind, placebo controlled trial, spironolactone improved
diastolic LV stiffness, but failed to improve exercise capacity176.
AGE breaking compounds
In small scale clinical HFpEF177 and HFrEF178 trials, the AGE breaking compound
alagebrium was shown to improve diastolic function and LV remodeling, whereas it enhanced
arterial compliance in aged hypertensives with vascular stiffening179. In contrast, in a recent
randomized study, alagebrium did not improve exercise tolerance, nor systolic or diastolic
function in HFrEF patients180.
Statins
Statins have been shown to improve endothelial function by stimulating activity and
expression of eNOS and by enhancing NO production through both cholesterol-dependent and
cholesterol-independent mechanisms181-183. The “pleiotropic effects” of statins also include
suppression of inflammation and oxidative stress, inhibition of neurohormonal activation,
inhibition of small GTP-binding protein (Rho, Rac and Ras) signaling and attenuation of
cardiac hypertrophy and fibrosis181-183. In a prospective, observational study, statin treatment
substantially improved survival in HFpEF, in contrast to ACE-I, ARBs, beta blockers or
calcium channel blockers which had no effect on survival184. More recently, two additional
prospective clinical studies demonstrated statins to be associated with improved survival in
HFpEF patients followed over a 5 year period185,186.
Life style changes and exercise training
Standard lifestyle recommendations (exercise, smoking cessation, weight loss) improve
insulin sensitivity and protect against DM development in obese, non-diabetic
individuals108,187. Obesity induces cardiac structural and functional changes characterized by
concentric LV remodeling188, altered myocardial substrate metabolism189,190 and diastolic LV
dysfunction, which is independently related to the extent of visceral adiposity191. Conversely,
weight loss in obesity ameliorates cardiac hypertrophy and diastolic dysfunction and improves
myocardial bioenergetics and vascular stiffness independent of changes in blood pressure and
other obesity-related co-morbidities192-199.
Physical exercise was shown to improve both clinical outcome and insulin sensitivity in nonischemic HFrEF patients200. The ESC HF guidelines firmly recommend regular physical
activity and structured exercise training. This recommendation is based on the fact that
exercise training improves exercise capacity and quality of life, does not adversely affect LV
remodeling and may reduce mortality and hospitalization in patients with mild-to-moderate
chronic HFrEF123,201-203. Exercise training could also be beneficial for improving outcome in
patients with HFpEF as exercise training was shown to improve endothelial dysfunction,
systemic inflammation and metabolic syndrome204-206. Moreover, exercise training was
suggested to attenuate the age-dependent decline in diastolic function207. Recently, the
randomized Exercise training in Diastolic Heart Failure Pilot Study (Ex-DHF) indeed
demonstrated in HFpEF patients that exercise training improved exercise capacity and quality
of life, which was associated with reverse atrial remodeling and enhanced diastolic LV
function208. Conversely, in a more recent study, HFpEF patients were allocated to a 16 weeks
exercise training program and although exercise training improved peak VO2 max, no
significant changes were observed in systolic and diastolic function209.
Improved metabolic control
Under physiological conditions, myocardial energy production is derived from a balanced
metabolism of glucose utilization and FFAs. Due to a decrease in glucose transport through
the sarcolemmal membrane of cardiomyocytes in diabetic patients with depletion of glucose
transporters 1 and 4, energy production is shifted to the beta-oxidation of FFAs210,211. The
oxidation of FFAs is less favourable in the energy balance due to the higher oxygen
demand212, while the increase in cardiac triglyceride accumulation is associated with diastolic
dysfunction213. Partial inhibitors of myocardial fatty acid oxidation, such as trimetazidine,
improved myocardial ischemia and exercise capacity in diabetic patients214. Trimetazidine,
increases myocardial efficiency by enhancing glucose metabolism and decreasing FFA
metabolism through inhibiting FFA beta-oxidation108,215,216. Trimetazidine exerts
cardioprotective effects by reducing oxidative damage, inhibiting inflammation and apoptosis
and improving endothelial function217-219. Indeed, in HFrEF, trimetazidine improved
myocardial function, remodeling, bioenergetics and functional class, while trimetazidine
decreased HF hospitalization215,220.
Metformin is a widely prescribed drug in DMII that improves insulin sensitivity and clinical
outcome in diabetic HF patients221, while metformin also exhibits anti-oxidant effects222.
Concerns about metformin being associated with lactic acidosis restricted its use in HF,
although clinical experience suggests that this risk is low223. Indeed, observational data
suggest that metformin may actually be beneficial for congestive HF224,225 and metformin
improved insulin sensitivity and minute ventilation, while it induced weight loss in nondiabetic IR HFrEF patients226. Since metformin is not a hypoglycemic agent, metformin could
represent a suitable metabolic therapy for nondiabetic HF patients.
Previously, it was shown that each 1% increase in HbA1c was associated with an 8%
increased risk of HF in diabetic patients even after adjusting for other factors, including
CAD227. In a small observational study, insulin-based metabolic control in DMII patients,
improved diastolic LV function and myocardial perfusion reserve228. Conversely, a recent
large prospective randomized trial demonstrated that long-term intensive insulin-based
glycemic control aiming at a HbA1c below 6% was associated with increased 5-year mortality
in DMII patients229. In addition, the DADD (Diabetes mellitus And Diastolic Dysfunction)
trial also failed to confirm improvement of diastolic dysfunction with tight insulin-based
glycemic control in DMII patients230. A statistical type II error could have contributed to the
negative outcome in DADD, because of strict DADD exclusion criteria, which resulted in
patients only having subtle evidence of diastolic dysfuntion with slow relaxation but normal
LV distensibility231. In addition to a type II error, this negative outcome could also suggest
that in DMII, myocardial damage not only results from hyperglycemia but also from
hyperinsulinemia, inflammation and oxidative stress100-102. Indeed, a recent study observed
improvement of diastolic LV function in DMII patients treated with rosiglitazone, to relate
more closely to the fall in malondialdehyde, a plasma marker of oxidative stress, than to the
fall in HbA1c, insulin or IL6232.
Signaling through NO-cGMP-PKG is downregulated in metabolic conditions such as obesity,
metabolic syndrome, IR and DM109,110, while in human diabetic cardiomyopathy, sildenafil
improved measurements of LV geometry, strain and torsion determined by magnetic
resonance imaging111.
Conclusion
In HFpEF, pathophysiological mechanisms, diagnostic and therapeutic strategies remain
uncertain. This is reflected in the lack of improvement of prognosis in HFpEF, over the last
decennia despite the steady increase of HFpEF prevalence. Advances in therapeutic strategies
are only to be expected when more comprehensive insight has been gained into underlying
pathophysiological mechanisms. Recent studies have contributed to improved understanding
of HFpEF pathophysiology demonstrating the importance of elevated cardiomyocyte stiffness
due to reduced myocardial PKG activity and high nitrosative/oxidative stress, which could be
related to the particularly high prevalence of metabolic risk factors in HFpEF.
Pharmacological agents that enhance myocardial NO-cGMP-PKG signaling and reduce
nitrosative/oxidative stress, in conjunction with strategies that improve overall metabolic risk
could therefore have therapeutic potential in HFpEF treatment.
References
1
Owan TE, Hodge DO, Herges RM, Jacobsen SJ, Roger VL, Redfield MM. Trends in prevalence and
outcome of heart failure with preserved ejection fraction. N Engl J Med 2006;355:251-259.
2
Borlaug BA, Paulus WJ. Heart failure with preserved ejection fraction: pathophysiology, diagnosis and
treatment. Eur Heart J 2011;32:670-679.
3
Paulus WJ, van Ballegoij JJM. Treatment of heart failure with normal ejection fraction. An
inconvenient truth! J Am Coll Cardiol 2010;55:526-537.
4
De Keulenaer GW, Brutsaert DL. Systolic and diastolic heart failure are overlapping phenotypes within
the heart failure spectrum. Circulation 2011;123:1996-2005.
5
van Heerebeek L, Borbely A, Niessen HWM, Bronzwaer JG, van der Velden J, Stienen GJ, Linke WA,
Laarman GJ, Paulus WJ. Myocardial structure and function differ in systolic and diastolic heart failure.
Circulation 2006;113:1966-1973.
6
Schwartzenberg S, Redfield MM, From AM, Sorajja P, Nishimura RA, Borlaug BA. Effects of
vasodilation in heart failure with preserved or reduced ejection fraction implications of distinct
pathophysiologies on response to therapy. J Am Coll Cardiol 2012;59:442-451.
7
Borlaug BA, Redfield MM. Diastolic and systolic heart failure are distinct phenotypes within the heart
failure spectrum. Cicrulation 2011;123:2006-2014.
8
Baicu CF, Zile MR, Aurigemma GP, Gaasch WH. Left ventricular systolic performance, function and
contractility in patients with diastolic heart failure. Circulation 2005;111:2306-2312.
9
Aurigemma GP, Zile MR, Gaasch WH. Contractile behavior of the left ventricle in diastolic heart
failure, with emphasis on regional systolic function. Circulation 2006;113:296-304.
10
Zile MR, Baicu CF, Gaasch WH. Diastolic heart failure - abnormalities in active relaxation and passive
stiffness of the left ventricle. N Engl J Med 2004;350:1953-1959.
11
Borbely A, van der Velden J, Papp Z, Bronzwaer JG, Edes I, Stienen GJ, Paulus WJ. Cardiomyocyte
stiffness in diastolic heart failure. Circulation 2005;111:774-781.
12
van Heerebeek L, Hamdani N, Handoko ML, Falcao-Pires I, Musters RJ, Kupreishvili K, Ijsselmuiden
AJ, Schalkwijk CG, Bronzwaer JG, Diamant M, Borbély A, van der Velden J, Stienen GJ, Laarman GJ,
Niessen HW, Paulus WJ. Diastolic stiffness of the failing diabetic heart: Importance of fibrosis,
advanced glycation endproducts, and myocyte resting tension. Circulation 2008;117:43-51.
13
Borbely A, Falcao-Pires I, van Heerebeek, Hamdani N, Edes I, Gavina C, Leite-Moreira AF, Bronzwaer
JG, Papp Z, van der Velden J, Stienen GJ, Paulus WJ. Hypophosphorylation of the stiff N2B titin
isoform raises cardiomyocyte resting tension in failing human myocardium. Circ Res 2009;104:780786.
14
van Heerebeek L, Hamdani N, Falcao-Pires I, Leite-Moreira AF, Begieneman MPV, Bronzwaer JGF,
van der Velden J, Stienen GJ, Laarman GJ, Somsen GA, Verheugt FWA, Niessen HW, Paulus WJ. Low
myocardial protein kinase G activity in heart failure with preserved ejection fraction. Circulation
2012;126:830-839.
15
Linke WA. Sense and stretchability: The role of titin and titin-associated proteins in myocardial stresssensing and mechanical dysfunction. Cardiovasc Res 2008;77:637-648.
16
Krüger M, Linke WA. Titin-based mechanical signaling in normal and failing myocardium. J Mol Cell
Cardiol 2009;46:490-498.
17
LeWinter MM, Granzier H. Cardiac titin: a multifunctional giant. Circulation 2010;121:2137-2145.
18
Makarenko I, Opitz CA, Leake MC, Neagoe C, Kulke M, Gwathmey JK, del Monte F, Hajjar RJ, Linke
WA. Passive stiffness changes caused by upregulation of compliant titin isoforms in human dilated
cardiomyopathy hearts. Circ Res 2004;95:708-716.
19
Nagueh SF, Shah G, Wu Y, Torre-Amione G, King NM, Lahmers S, Witt CC, Becker K, Labeit S,
Granzier HL. Altered titin expression, myocardial stiffness and left ventricular function in patients with
dilated cardiomyopathy. Circulation 2004;110:155-162.
20
Neagoe C, Kulke M, del Monte F, Gwathmey JK, de Tombe PP, Hajjar RJ, Linke WA. Titin isoform
switch in ischemic human heart disease. Circulation 2002;106:1333-1341.
21
Williams L, Howell N, Pagano D, Andreka P, Vertesaljai M, Pecor T, Frenneaux M, Granzier H. Titin
isoform expression in aortic stenosis. Clin Sci 2009;117:237-242.
22
Falcao-Pires I, Hamdani N, Borbely A, Gavina C, Schalkwijk CG, van der Velden J, van Heerebeek L,
Stienen GJ, Niessen HW, Leite-Moreira AF, Paulus WJ. Diabetes mellitus worsens diastolic left
ventricular dysfunction in aortic stenosis through altered myocardial structure and cardiomyocyte
stiffness. Circulation 2011;124:1151-1159.
23
Krüger 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.
24
Krüger M, Kötter S, Grützner A, Lang P, Andresen C, Redfield MM, Butt E, dos Remedios CG, Linke
WA. Protein kinase G modulates human myocardial passive stiffness by phosphorylation of the titin
springs. Circ Res 2009;104:87-94.
25
Hidalgo C, Hudson B, Bogomolovas J, Zhu Y, Anderson B, Greaser M, Labeit S, Granzier H. PKC
phosphorylation of titin’s PEVK element. A novel and conserved pathway for modulating myocardial
stiffness. Circ Res 2009;105:631-638.
26
Grützner A, Garcia-Manyes S, Kötter S, Badilla CL, Fernandez JM, Linke WA. Modulation of titinbased sti ffness by disulfide bonding in the cardiac titin N2-B unique sequence. Biophys J 2009;97:825834.
27
Tsai E, Kass DA. Cyclic GMP signalling in cardiovascular pathology and therapeutics. Pharmacol Ther
2009;122:216-238.
28
Takimoto E, Champion HC, Li M, Belardi D, Ren S, Rodriguez ER, Bedja D, Gabrielson KL, Wang Y,
Kass DA. Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac
hypertrophy. Nat Med 2005;11:214-222.
29
Bishu K, Hamdani N, Mohammed SF, Krüger M, Ohtani T, Ogut O, Brozovich FV, Burnett JC, Linke
WA, Redfield MM. Sildenafil and BNP acutely phosphorylate titin and improve diastolic distensbility
in vivo. Circulation 2011;124:2882-2891.
30
Guazzi M, Vicenzi M, Arena R, Guazzi MD. PDE-5 inhibition with sildenafil improves left ventricular
diastolic function, cardiac geometry and clinical status in patients with stable systolic heart failure:
results of a 1-year prospective, randomized, placebo controlled trial. Circ Heart Fail 2011;4:8-17.
31
Guazzi M, Vicenzi M, Arena R, Guazzi MD. Pulmonary hypertension in heart failure with preserved
ejection fraction: a target of phosphodiesterase-5 inhibition in a 1-year study. Circulation 2011;124:164174.
32
Borlaug BA, Melenovsky V, Marhin T, Fitzgerald P, Kass DA. Sildenafil inhibits beta-adrenergic
stimulated cardiac contractility in humans. Circulation 2005;112:2642-2649.
33
Vlachopoulos C, Hirata K, O’Rourke MF. Effect of sildenafil on arterial stiffness and wave reflection.
Vasc Med 2003;8:243-248.
34
Katz SD, Balidemaj K, Homma S, Wu H, Wang J, Maybaum S. Acute type 5 phosphodiesterase
inhibition with sildenafil enhances flow-mediated vasodilation in patients with chronic heart failure. J
Am Coll Cardiol 2000;36:845-851.
35
Lewis GD, Lachmann J, Camuso J, Lepore JJ, Shin J, Martinovic ME, Systrom DM, Bloch KD,
Semigran MJ. Sildenafil improves exercise hemodynamics and oxygen uptake in patients with systolic
heart failure. Circulation 2007;115:59-66.
36
Hirata K, Adji A, Vlachopoulos V, O’Rourke MF. Effect of sildenafil on cardiac performance in
patients with heart failure. Am J Cardiol 2005;96:1436-1440.
37
Shuai X, Chen Y, Lu Y, Su G, Wang Y, Zhao H, Han J. Diagnosis of heart failure and preserved
ejection fraction: which parameters and diagnostic strategies are more valuable. Eur J Heart Fail
2011;13:737-741.
38
Schulz E, Jansen T, Wenzel P, Daiber A, Münzel T. Nitric oxide, tetrahydrobiopterin, oxidative stress
and endothelial dysfunction in hypertension. Antiox Redox Signal 2008;10:1115-1126.
39
Weber M, Lauer N, Mulsch A, Kojda G. The effect of peroxynitrite on the catalytic activity of soluble
guanylyl cyclase. Free Radic Biol Med 2001;31:1360-1367.
40
Münzel T, Gori T, Bruno RM, Taddei S. Is oxidative stress a therapeutic target in cardiovascular
disease? Eur Heart J 2010;31:2741-2748.
41
Nediani C, Raimondi L, Borchi E, Cerbai E. Nitric oxide/reactive oxygen species generation and
nitroso/redox imbalance in heart failure: from molecular mechanisms to therapeutic implications.
Antioxid Redox Signal 2011;14:289-331.
42
Katz SD, Hryniewicz K, Hriljac I, Balidemaj K, Dimayuga C, Hudaihed A, Yasskiy A. Vascular
endothelial dysfunction and mortality risk in patients with chronic heart failure. Circulation
2005;111:310-314.
43
Katz SD, Khan T, Zeballos GA, Mathew L, Potharlanka P, Knecht M, Whelan J. Decreased activity of
the L-arginine-nitric oxide metabolic pathway in patients with congestive heart failure. Circulation
1999;99:2113-2117.
44
Bauersachs J, Bouloumié A, Fraccarollo D, Hu K, Busse R, Ertl G. Endothelial dysfunction in chronic
myocardial infarction despite increased vascular endothelial nitric oxide synthase and soluble guanylate
cyclase expression: role of enhanced vascular superoxide production. Circulation 1999;100:292-298.
45
Paulus WJ, Bronzwaer JGF. Nitric oxide’s role in the heart: control of beating or breathing? Am J
Physiol Heart Circ Physiol 2004;287:H8-H13.
46
Seddon M, Shah AM, Casadei B. Cardiomyocytes as effectors of nitric oxide signaling. Cardiovasc Res
2007;75:315-326.
47
Hammond J, Balligand JL. Nitric oxide synthase and cyclic GMP signaling in cardiac myocytes: from
contractility to remodeling. J Mol Cell Cardiol 2012;52:330-340.
48
Paulus WJ, Vantrimpont PJ, Shah AM. Acute effects of nitric oxide on left ventricular relaxation and
diastolic distensibility in humans. Assessment by bicoronary sodium nitroprusside infusion. Circulation
1994;89:2070-2078.
49
Heymes C, Vanderheyden M, Bronzwaer JG, Shah AM, Paulus WJ. Endomyocardial nitric oxide
synthase and left ventricular preload reserve in dilated cardiomyopathy. Circulation 1999;99:3009-3016.
50
Matter CM, Mandinov L, Kaufmann PA, Vassalli G, Jiang Z, Hess OM. Effect of NO donors on LV
diastolic function in patients with severe pressure-overload hypertrophy. Circulation 1999;99:23962401.
51
Takimoto E, Belardi D, Tocchetti CG, Vahebi S, Cormaci G, Ketner EA, Moens AL, Champion HC,
Kass DA. Compartmentalization of cardiac beta-adrenergic inotropy modulation by phosphodiesterase
type 5. Circulation 2007;115:2159-2167.
52
Wilson Tang WH, Tong W, Shrestha K, Wang Z, Levison BS, Delfraino B, Hu B, Troughton RW,
Klein AL, Hazen SL. Differential effects of arginine methylation on diastolic dysfunction and disease
progression in patients with chronic systolic heart failure. Eur Heart J 2008;29:2506-2513.
53
Visser M, Paulus WJ, Vermeulen MAR, Richir MC, Davids M, Wisselink W, de Mol BA, van Leeuwen
PA. The role of asymmetric dimethylarginine and arginine in the failing heart and its vasculature. Eur J
Heart Fail 2010;12:1274-1281.
54
Trochu JN, Bouhour JB, Kaley G, Hintze TH. Role of endothelium-derived nitric oxide in the
regulation of cardiac oxygen metabolism: implications in health and disease. Circ Res 2000;87:11081117.
55
Nisoli E, Tonello C, Cardile A, Cozzi V, Bracale R, Tedesco L, Falcone S, Valerio A, Cantoni O,
Clementi E, Moncada S, Carruba MO. Calorie restriction promotes mitochondrial biogenesis by
inducing the expression of eNOS. Science 2005;310:314-317.
56
Phan TT, Abozguia K, Nallur Shivu G, Mahadevan G, Ahmed I, Williams L, Dwivedi G, Patel K,
Steendijk P, Ashrafian H, Henning A, Frenneaux M. Heart failure with preserved ejection fraction is
characterized by dynamic impairment of active relaxation and contraction of the left ventricle on
exercise and associated with myocardial energy deficiency. J Am Coll Cardiol 2009;54:402-409.
57
Lamb HJ, Beyerbracht HP, van der Laarse A, Stoel BC, Doornbos J, van der Wall EE, de Roos A.
Diastolic dysfunction in hypertensive heart disease is associated with altered myocardial metabolism.
Circulation 1999;99:2261-2267.
58
Smith CS, Bottomley PA, Schulman SP, Gerstenblith G, Weiss RG. Altered creatine kinase adenosine
triphosphate kinetics in failing hypertrophied human myocardium. Circulation 2006;114:1151-1158.
59
Neubauer S, Krahe T, Schindler R, Horn M, Hillenbrand H, Entzeroth C, Mader H, Kromer EP, Riegger
GA, Lackner K. 31P magnetic resonance spectroscopy in dilated cardiomyopathy and coronary artery
disease: altered cardiac high-energy phosphate metabolism in heart failure. Circulation 1992;86:18101818.
60
Diamant M, Lamb HJ, Groeneveld Y, Endert EL, Smit JW, Bax JJ, Romijn JA, de Roos A, Radder JK.
Diastolic dysfunction is associated with altered myocardial metabolism in asymptomatic normotensive
patients with well-controlled type 2 diabetes mellitus. J Am Coll Cardiol 2003;42:328-335.
61
Rider OJ, Francis JM, Ali MK, Holloway C, Pegg T, Robson MD, Tyler D, Byrne J, Clarke K,
Neubauer S. Effects of catecholamine stress on diastolic function and myocardial energetics in obesity.
Circulation 2012;125:1511-1519.
62
Cary SP, Winger JA, Marletta MA. Tonic and acute nitric oxide signaling through soluble guanylate
cyclase is mediated by nonheme nitric oxide, ATP and GTP. Proc Natl Acad Sci USA 2005;102:1306413069.
63
Tsai EJ, Liu Y, Koitabaschi N, Bedja D, Danner T, Jasmin JF, Lisanti MP, Friebe A, Takimoto E, Kass
DA. Pressure-overload-induced subcellular relocalization/oxidation of soluble guanylate cyclase in the
heart modulates enzyme stimulation. Circ Res 2012;110:295-303.
64
Zhang M, Kass DA. Phosphodiesterases and cardiac cGMP: evolving roles and controversies. Trends
Pharmacol Sci 2011;32:360-365.
65
Castro LRV, Schittl J, Fischmeister R. Feedback control through cGMP-dependent protein kinase
contributes to differential regulation and compartmentation of cGMP in rat cardiac myocytes. Circ Res
2010;107:1232-1240.
66
Fischmeister R, Castro LRV, Abi-Gerges A, Rochais F, Jurevičius J, Leroy J, Vandecasteele G.
Compartmentation of cyclic nucleotide signaling in the heart. The role of cyclic nucleotide
phosphodiesterases. Circ Res 2006;99:816-828.
67
Zaccolo M, Movsesian MA. cAMP and cGMP signaling cross-talk: Role of phosphodiesterases and
implications for cardiac pathophysiology. Circ Res 2007;100:1569-1578.
68
Kitzman DW, Little WC, Brubaker PH, Anderson RT, Hundley WG, Marburger CT, Brosnihan B,
Morgan TM, Stewart KP. Pathophysiological characterization of isolated diastolic heart failure in
comparison to systolic heart failure. JAMA 2002;288:2144-2150.
69
Gaasch WH, Zile MR. Left ventricular diastolic dysfunction and diastolic heart failure. Annu Rev Med
2004;55:373-394.
70
Aurigemma GP, Gaasch WH. Clinical practice. Diastolic heart failure. N Engl J Med 2004;351:10971105.
71
Levy D, Garrison RJ, Savage DD, Kannel WB, Castelli WP. Prognostic implications of
echocardiographically determined left ventricular mass in the Framingham Heart Study. N Engl J Med.
1990;322:1561-1566.
72
Ritchie RH, Irvine JC, Rosenkranz AC, Patel R, Wendt IR, Horowitz JD, Kemp-Harper BK. Exploiting
cGMP-based therapies for the prevention of left ventricular hypertrophy: NO* and beyond. Pharmacol
Ther 2009;124:279-300.
73
Rohini A, Agrawal N, Koyani CN, Singh R. Molecular targets and regulators of cardiac hypertrophy.
Pharmacol Res 2010;61:269-280.
74
Solomon SD, Janardhanan R, Verma A, Bourgoun M, Daley WL, Purkayastha D, Lacourcière Y,
Hippler SE, Fields H, Naqvi TZ, Mulvagh SL, Arnold JM, Thomas JD, Zile MR, Aurigemma GP;
Valsartan In Diastolic Dysfunction (VALIDD) Investigators. For the Valsartan In Diastolic Dysfunction
(VALIDD) Investigators. Effect of angiotensin receptor blockade and antihypertensive drugs on
diastolic function in patients with hypertension and diastolic dysfunction: a randomised trial. Lancet
2007;369:2079-2087.
75
Krüger M, Linke WA. The giant protein titin: a regulatory node that integrates myocyte signaling
pathways. J Biol Chem 2011;286:9905-9912.
76
Hare JM, Stamler JS. NO/redox disequilibrium in the failing heart and cardiovascular system. J Clin
Invest 2005;115:509-517.
77
Giordano FJ. Oxygen, oxidative stress, hypoxia and heart failure. J Clin Invest 2005;115:500-508.
78
Seddon M, Looi YH, Shah AJ. Oxidative stress and redox signaling in cardiac hypertrophy and heart
failure. Heart 2007;93:903-907.
79
Takimoto E, Kass DA. Role of oxidative stress in cardiac hypertrophy and remodeling. Hypertension
2007;49:241-248.
80
Silberman GA, Fan THM, Liu H, Jiao Z, Xiao HD, Lovelock JD, Boulden BM, Widder J, Fredd S,
Bernstein KE, Wolska BM, Dikalov S, Harrison DG, Dudley SC Jr. Uncoupled cardiac nitric oxide
synthase mediates diastolic dysfunction. Circulation 2010;121:519-528.
81
Westermann D, Riad A, Richter U, Jäger S, Savvatis K, Schuchardt M, Bergmann N, Tölle M,
Nagorsen D, Gotthardt M, Schultheiss HP, Tschöpe C. Enhancement of the endothelial NO synthase
attenuates experimental diastolic heart failure. Basic Res Cardiol 2009;104:499-509.
82
Westermann D, Lindner D, Kasner M, Zietsch C, Savvatis K, Escher F, von Schlippenbach J, Skurk C,
Steendijk P, Riad A, Poller W, Schultheiss HP, Tschöpe C. Cardiac inflammation contributes to
changes in the extracellular matrix in patients with heart failure and normal ejection fraction. Circ Heart
Fail 2011;4:44-52.
83
Hess DT, Matsumoto A, Kim SO, Marshall HE, Stamler JS. Protein S-nitrosylation: purview and
parameters. Nat Rev Mol Cell Biol 2005;6:150-166.
84
Fonorow GC, Stough WG, Abraham WT, Albert NM, Gheorghiade M, Greenberg BH, O'Connor CM,
Sun JL, Yancy CW, Young JB.OPTIMIZE-HF Investigators and Hospitals. Characteristics, treatments
and outcomes of patients with preserved systolic function hospitalized for heart failure: a report from
the OPTIMIZE-HF Registry. J Am Coll Cardiol 2007;50:768-777.
85
Yancy CW, Lopatin M, Stevenson LW, De Marco T, Fonarow GC, for the ADHERE Scientific
Advisory Committee and Investigators. Clinical presentation, management, and in-hospital outcomes of
patients admitted with acute decompensated heart failure with preserved systolic function. A Report
from the Acute Decompensated Heart Failure National Registry (ADHERE). J Am Coll Cardiol
2006;47:76-84.
86
McMurray JJV, Carson PE, Komajda M, McKelvie R, Zile MR, Ptaszynska A, Staiger C, Donovan JM,
Massie BM. Heart failure with preserved ejection fraction: Clinical characteristics of 4133 patients
enrolled in the I-PRESERVE trial. Eur J Heart Fail 2008;10:149-156.
87
Grundy SM, Cleeman JI, Daniels SR, Donato KA, Eckel RH, Franklin BA, Gordon DJ, Krauss RM,
H
Savage PJ, Smith SC Jr, Spertus JA, Costa F. Diagnosis and management of the metabolic syndrome: an
American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation
2005;112:2735-2752.
88
Horwich TB, Fonarow GC. Glucose, obesity, metabolic syndrome and diabetes. J Am Coll Cardiol
2010;55:283-293.
89
Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes. Estimates for the year
2000 and projections for 2030. Diabetes care 2004;27:1047-1053.
90
Must A, Spadano J, Coakley EH, Field AE, Colditz G, Dietz WH. The disease burden associated with
overweight and obesity. JAMA 1999;282:1523-1529.
91
Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults: findings from
the third National Health and Nutrition Examination Survey. JAMA 2002;287:356-359.
92
Ginsberg HN, MacCallum PR. The obesity, metabolic syndrome, and type 2 diabetes mellitus
pandemic: Part I. Increased cardiovascular disease risk and the importance of atherogenic dyslipidemia
in persons with the metabolic syndrome and type 2 diabetes mellitus. J Cardiometab Syndr 2009;4:113119.
93
Kannel WB, Hjortland M, Castelli WP. Role of diabetes in congestive heart failure: the Framingham
study. Am J Cardiol 1974;34:29-34.
94
Kenchaiah S, Evans JC, Levy D, Wilson PW, Benjamin EJ, Larson MG, Kannel WB, Vasan RS.
Obesity and the risk of heart failure. N Engl J Med 2002;347:305-313.
95
Ingelsson E, Sundström J, Ärnlöv J, Zethelius B, Lind L. Insulin resistance and risk of congestive heart
failure. JAMA 2005;294:334-341.
96
Boyer JK, Thanigaraj S, Schechtman KB, Pérez JE. Prevalence of ventricular diastolic dysfunction in
asymptomatic, normotensive patients with diabetes mellitus. Am J Cardiol 2004;93:870-875.
97
De las Fuentes L, Brown AL, Mathews SJ, Waggoner AD, Soto PF, Gropler RJ, Dávila-Román VG.
Metabolic syndrome is associated with abnormal left ventricular diastolic function independent of LV
mass. Eur Heart J 2007;28:553-559.
98
Dinh W, Lankisch M, Nickl W, Scheyer D, Scheffold T, Kramer F, Krahn T, Klein RM, Barroso MC,
Füth R. Insulin resistance and glycemic abnormalities are associated with deterioration of left
ventricular diastolic function: a cross-sectional study. Cardiovasc Diabet 2010;9:63-76.
99
Russo C, Jin Z, Homma S, Rundek T, Elkind MS, Sacco RL, Di Tullio MR. Effect of obesity and
overweight on left ventricular diastolic function. A community-based study in an elderly cohort. J Am
Coll Cardiol 2011;57:1368-1374.
100
Poornima IG, Parikh P, Shannon RP. Diabetic cardiomyopathy: The search for a unifying hypothesis.
Circ Res 2006;98:596-605.
101
Boudina S, Abel ED. Diabetic cardiomyopathy revisited. Circulation 2007;115:3213-3223.
102
Falcao-Pires I, Leite-Moreira AF. Diabetic cardiomyopathy: understanding the molecular and cellular
basis to progress in diagnosis and treatment. Heart Fail Rev 2012;17:325-344.
103
Brooks BA, Franjic B, Ban CR, Swaraj K, Yue DK, Celermajer DS, Twigg SM. Diastolic dysfunction
and abnormalities of the microcirculation in type 2 diabetes. Diabetes Obes Metab 2008;10:739-746.
104
von Bibra H, Thrainsdottir IS, Hansen A, Dounis V, Malmberg K, Rydén L.Tissue Doppler imaging for
the detection and quantitation of myocardial dysfunction in patients with type 2 diabetes mellitus: a
methodological study. Diabetes Vasc Dis Res 2005;2:24-30.
105
Asbun J, Villareal FJ. The pathogenesis of myocardial fibrosis in the setting of diabetic
cardiomyopathy. J Am Coll Cardiol 2006;47:693-700.
106
Hartog JWL, Voors AA, Bakker SJL, Smit AJ, van Veldhuisen DJ. Advanced glycation end-products
(AGEs) and heart failure: Pathophysiology and clinical implications. Eur J Heart Fail 2007;9:11461155.
107
Abel ED, Litwin ES, Sweeney G. Cardiac remodelling in obesity. Physiol Rev 2008;88:389-419.
108
Witteles RM, Fowler MB. Insulin-resistant cardiomyopathy. J Am Coll Cardiol 2008;51:93-102.
109
Liu S, Ma X, Gong M, Shi L, Lincoln T, Wang S. Glucose down-regulation of cGMP-dependent protein
kinase I expression in vascular smooth muscle cells involves NAD(P)H oxidase-derived reactive
oxygen species. Free Radic Biol Med 2007;42:852-863.
110
Rizzo NO, Maloney E, Pham M, Luttrell I, Wessells H, Tateya S, Daum G, Handa P, Schwartz MW,
Kim F. Reduced NO-cGMP signaling contributes to vascular inflammation and insulin resistance
induced by high-fat feeding. Arterioscler Thromb Vasc Biol 2010;30:758-765.
111
Giannetta E, Isidori AM, Galea N, Carbone I, Mandosi E, Vizza CD, Naro F, Morano S, Fedele F,
Lenzi A. Chronic Inhibition of cGMP Phosphodiesterase 5A Improves Diabetic Cardiomyopathy: A
Randomized, Controlled Clinical Trial Using Magnetic Resonance Imaging With Myocardial Tagging.
Circulation 2012;125:2323-2333.
112
Krüger M, Babicz K, von Frieling-Salewsky M, Linke WA. Insulin signaling regulates cardiac titin
properties in heart development and diabetic cardiomyopathy. J Mol Cell Cardiol 2010;48:910-916.
113
Kim JA, Montagnani M, Koh KK, Quon MJ. Reciprocal relationships between insulin resistance and
endothelial dysfunction: molecular and pathophysiological mechanisms. Circulation 2006;113:18881904.
114
Geraldes P, King GL. Activation of protein kinase C isoforms and its impact on diabetic complications.
Circ Res 2010;106:1319-1331.
115
Connelly KA, Kelly DJ, Zhang Y, Prior DL, Advani A, Cox AJ, Thai K, Krum H, Gilbert RE.
Inhibition of protein kinase C-beta by ruboxistaurin preserves cardiac function and reduces extracellular
matrix production in diabetic cardiomyopathy. Circ Heart Fail 2009;2:129-137.
116
Ren J, Pulakat L, Whaley-Connell A, Sowers JR. Mitochondrial biogenesis in the metabolic syndrome
and cardiovascular disease. J Mol Med 2010;88:993-1001.
117
Bugger H, Abel ED. Mitochondria in the diabetic heart. Cardiovasc Res 2010;88:229-240.
118
Moens AL, Kietadisorn R, Lin JY, Kass D. Targeting endothelial and myocardial dysfunction with
tetrahydrobiopterin. J Mol Cell Cardiol 2011;51:559-563.
119
Moens AL, Takimoto E, Tocchetti CG, Chakir K, Bedja D, Cormaci G, Ketner EA, Majmudar M,
Gabrielson K, Halushka MK, Mitchell JB, Biswal S, Channon KM, Wolin MS, Alp NJ, Paolocci N,
Champion HC, Kass DA. Reversal of cardiac hypertrophy and fibrosis from pressure overload by
tetrahydrobiopterin: efficacy of recoupling nitric oxide synthase as a therapeutic strategy. Circulation
2008;117:2626-2636.
120
Stroes E, Kastelein J, Cosentino F, Erkelens W, Wever R, Koomans H, Lüscher T, Rabelink T.
Tetrahydrobiopterin restores endothelial function in hypercholesterolemia. J Clin Invest. 1997;99:41-46.
121
Heitzer T, Krohn K, Albers S, Meinertz . Tetrahydrobiopterin improves endothelium-dependent
vasodilation by increasing nitric oxide activity in patients with Type II diabetes mellitus. Diabetologia
2000;43:1435-1438.
122
Higashi Y, Sasaki S, Nakagawa K, Fukuda Y, Matsuura H, Oshima T, Chayama K. Tetrahydrobiopterin
enhances forearm vascular response to acetylcholine in both normotensive and hypertensive individuals.
Am J Hypertens 2002;15:326-332.
123
Authors/Task Force Members, McMurray JJ, Adamopoulos S, Anker SD, Auricchio A, Böhm M,
Dickstein K, Falk V, Filippatos G, Fonseca C, Gomez-Sanchez MA, Jaarsma T, Køber L, Lip GY,
Maggioni AP, Parkhomenko A, Pieske BM, Popescu BA, Rønnevik PK, Rutten FH, Schwitter J,
Seferovic P, Stepinska J, Trindade PT, Voors AA, Zannad F, Zeiher A; ESC Committee for Practice
Guidelines (CPG), Bax JJ, Baumgartner H, Ceconi C, Dean V, Deaton C, Fagard R, Funck-Brentano C,
Hasdai D, Hoes A, Kirchhof P, Knuuti J, Kolh P, McDonagh T, Moulin C, Popescu BA, Reiner Z,
Sechtem U, Sirnes PA, Tendera M, Torbicki A, Vahanian A, Windecker S; Document Reviewers,
McDonagh T, Sechtem U, Bonet LA, Avraamides P, Ben Lamin HA, Brignole M, Coca A, Cowburn P,
Dargie H, Elliott P, Flachskampf FA, Guida GF, Hardman S, Iung B, Merkely B, Mueller C, Nanas JN,
Nielsen OW, Orn S, Parissis JT, Ponikowski P. ESC Guidelines for the diagnosis and treatment of acute
and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic
Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart
Failure Association (HFA) of the ESC. Eur Heart J 2012;33:1787-1847.
124
Iachini Bellisarii F, Radico F, Muscente F, Horowitz J, De Caterina R. Nitrates and other nitric oxide
donors in cardiology: current positioning and perspectives. Cardiovasc Drugs Ther 2012;26:55-69.
125
Chirkov YY, Horowitz JD. Impaired tissue responsiveness to organic nitrates and nitric oxide: a new
therapeutic frontier? Pharmacol Ther 2007;116:287-305.
126
Cohn JN, Archibald DG, Ziesche S, Franciosa JA, Harston WE, Tristani FE, Dunkman WB, Jacobs W,
Francis GS, Flohr KH. Effect of vasodilator therapy on mortality in chronic congestive heart failure.
Results of a Veterans Administration Cooperative Study. N Engl J Med 1986;314:1547-1552.
127
Taylor AL, Ziesche S, Yancy C, Carson P, D'Agostino R Jr, Ferdinand K, Taylor M, Adams K,
Sabolinski M, Worcel M, Cohn JN; African-American Heart Failure Trial Investigators. Combination of
isosorbide dinitrate and hydralazine in blacks with heart failure. N Engl J Med 2004;351:2049-2057.
128
Münzel T, Kurz S, Rajagopalan S, Thoenes M, Berrington WR, Thompson JA, Freeman BA, Harrison
DG. Hydralazine prevents nitroglycerin tolerance by inhibiting activation of a membrane-bound NADH
oxidase. A new action for an old drug. J Clin Invest 1996;98:1465-1470.
129
Kamath SA, Drazner MH, Wynne J, Fonarow GC, Yancy CW. Characteristics and outcomes in African
American patients with decompensated heart failure. Arch Intern Med 2008;168:1152-1158.
130
Gheorghiade M, Marti CN, Sabbah HN, Roessig L, Greene SJ, Böhm M, Burnett JC, Campia U,
Cleland JG, Collins SP, Fonarow GC, Levy PD, Metra M, Pitt B, Ponikowski P, Sato N, Voors AA,
Stasch JP, Butler J. Soluble guanylate cyclase: a potential therapeutic target for heart failure. Heart Fail
Rev 2013;18;123-134.
131
Evgenov OV, Pacher P, Schmidt PM, Haskó G, Schmidt HH, Stasch JP. NO-independent stimulators
and activators of soluble guanylate cyclase: discovery and therapeutic potential. Nat Rev Drug Discov
2006;5:755-768.
132
Mitrovic V, Jovanovic A, Lehinant S. Soluble guanylate cyclase modulators in heart failure. Curr Heart
Fail Rep 2011;8:38-44.
133
Stasch JP, Schmidt PM, Nedvetsky PI, Nedvetskaya TY, H S AK, Meurer S, Deile M, Taye A, Knorr
A, Lapp H, Müller H, Turgay Y, Rothkegel C, Tersteegen A, Kemp-Harper B, Müller-Esterl W,
Schmidt HH. Targeting the heme-oxidized nitric oxide receptor for selective vasodilatation of diseased
blood vessels. J Clin Invest. 2006;116:2552-2561.
134
Lapp H, Mitrovic V, Franz N, Heuer H, Buerke M, Wolfertz J, Mueck W, Unger S, Wensing G, Frey R.
Cinaciguat (BAY 58-2667) improves cardiopulmonary hemodynamics in patients with acute
decompensated heart failure. Circulation 2009;119:2781-2788.
135
Erdmann E, Semigran MJ, Nieminen MS, Gheorghiade M, Agrawal R, Mitrovic V, Mebazaa A.
Cinaciguat, a soluble guanylate cyclase activator, unloads the heart but also causes hypotension in acute
decompensated heart failure. Eur Heart J 2013;34:57-67.
136
Nagayama T, Zhang M, Hsu S, Takimoto E, Kass DA. Sustained soluble guanylate cyclase stimulation
offsets nitric-oxide synthase inhibition to restore acute cardiac modulation by sildenafil. J Pharm Exp
Ther 2008;326:380-387.
137
Mills RM, LeJemtel TH, Horton DP, Liang C, Lang R, Silver MA, Lui C, Chatterjee K. Sustained
hemodynamic effects of an infusion of nesiritide (human b-type natriuretic peptide) in heart failure: a
randomized, double-blind, placebo-controlled clinical trial. Natrecor Study Group. J Am Coll Cardiol
1999;34:155-162.
138
O'Connor CM, Starling RC, Hernandez AF, Armstrong PW, Dickstein K, Hasselblad V, Heizer GM,
Komajda M, Massie BM, McMurray JJ, Nieminen MS, Reist CJ, Rouleau JL, Swedberg K, Adams KF
Jr, Anker SD, Atar D, Battler A, Botero R, Bohidar NR, Butler J, Clausell N, Corbalán R, Costanzo
MR, Dahlstrom U, Deckelbaum LI, Diaz R, Dunlap ME, Ezekowitz JA, Feldman D, Felker GM,
Fonarow GC, Gennevois D, Gottlieb SS, Hill JA, Hollander JE, Howlett JG, Hudson MP, Kociol RD,
Krum H, Laucevicius A, Levy WC, Méndez GF, Metra M, Mittal S, Oh BH, Pereira NL, Ponikowski P,
Tang WH, Tanomsup S, Teerlink JR, Triposkiadis F, Troughton RW, Voors AA, Whellan DJ, Zannad
F, Califf RM. Effect of nesiritide in patients with acute decompensated heart failure. N Engl J Med
2011;365:32-43.
139
Brandt I, Lambeir AM, Ketelslegers JM, Vanderheyden M, Scharpé S, De Meester I. Dipeptidylpeptidase IV converts intact B-type natriuretic peptide into its des-SerPro form. Clin Chem 2006;52:8287.
140
Boerrigter G, Costello-Boerrigter LC, Harty GJ, Lapp H, Burnett JC Jr. Des-serine-proline brain
natriuretic peptide 3-32 in cardiorenal regulation. Am J Physiol 2007;292:R897-R901.
141
Lam CS, Burnett JC Jr, Costello-Boerrigter L, Rodeheffer RJ, Redfield MM. Alternate circulating proB-type natriuretic peptide and B-type natriuretic peptide forms in the general population. J Am Coll
Cardiol 2007;49:1193-1202.
142
Gomez N, Touihri K, Matheeussen V, Mendes Da Costa A, Mahmoudabady M, Mathieu M, Baerts L,
Peace A, Lybaert P, Scharpé S, De Meester I, Bartunek J, Vanderheyden M, Mc Entee K. Dipeptidyl
peptidase IV inhibition improves cardiorenal function in overpacing-induced heart failure. Eur J Heart
Fail 2012;14:14-21.
143
Rohini A, Agrawal N, Koyani CN, Singh R. Molecular targets and regulators of cardiac hypertrophy.
Pharmacol Res 2010;61:269-280.
144
Abuissa H, Jones PG, Marso SP, O'Keefe JH Jr. Angiotensin-converting enzyme inhibitors or
angiotensin receptor blockers for prevention of type 2 diabetes: a meta-analysis of randomized clinical
trials. J Am Coll Cardiol 2005;46:821-826.
145
McFarlane SI, Kumar A, Sowers JR. Mechanisms by which angiotensin-converting enzyme inhibitors
prevent diabetes and cardiovascular disease. Am J Cardiol 2003;91:30H-37H.
146
Hayashi T, Sohmiya K, Ukimura A, Endoh S, Mori T, Shimomura H, Okabe M, Terasaki F, Kitaura Y.
Angiotensin II receptor blockade prevents microangiopathy and preserves diastolic function in the
diabetic rat heart. Heart 2003;89:1236-1242.
147
Kawasaki D, Kosugi K, Waki H, Yamamoto K, Tsujino T, Masuyama T. Role of activated reninangiotensin system in myocardial fibrosis and left ventricular diastolic dysfunction in diabetic patients-reversal by chronic angiotensin II type 1A receptor blockade. Circ J 2007;71:524-529.
148
Miyata T, van Ypersele de Strihou C. Angiotensin II receptor blockers and angiotensin converting
enzyme inhibitors: implication of radical scavenging and transition metal chelation in inhibition of
advanced glycation end product formation. Arch Biochem Biophys 2003;419:50-54.
149
Granger CB, McMurray JJ, Yusuf S, Held P, Michelson EL, Olofsson B, Ostergren J, Pfeffer MA,
Swedberg K; CHARM Investigators and Committees. Effects of candesartan in patients with chronic
heart failure and reduced left-ventricular systolic function intolerant to angiotensin-converting-enzyme
inhibitors: the CHARM-Alternative trial. Lancet 2003;362:772-776.
150
Sharpe N, Smith H, Murphy J, Greaves S, Hart H, Gamble G. Early prevention of left ventricular
dysfunction after myocardial infarction with angiotensin-converting-enzyme inhibition. Lancet
1991;337:872-876.
151
Lonn E, Shaikholeslami R, Yi Q, Bosch J, Sullivan B, Tanser P, Magi A, Yusuf S. Effects of ramipril
on left ventricular mass and function in cardiovascular patients with controlled blood pressure and with
preserved left ventricular ejection fraction: a substudy of the Heart Outcomes Prevention Evaluation
(HOPE) Trial. J Am Coll Cardiol 2004;43:220-2206.
152
Ciulla MM, Paliotti R, Esposito A, Dìez J, López B, Dahlöf B, Nicholls MG, Smith RD, Gilles L,
Magrini F, Zanchetti A. Different effects of antihypertensive therapies based on losartan or atenolol on
ultrasound and biochemical markers of myocardial fibrosis: results of a randomized trial. Circulation
2004;110:552-557.
153
Dandona P, Dhindsa S, Ghanim H, Chaudhuri A. Angiotensin II and inflammation: the effect of
angiotensin-converting enzyme inhibition and angiotensin II receptor blockade. J Hum Hypertens
2007;21:20-27.
154
Williams HC, Griendling KK. NADPH oxidase inhibitors: new antihypertensive agents? J Cardiovasc
Pharmacol 2007;50:9-16.
155
Wittstein IS, Kass DA, Pak PH, Maughan WL, Fetics B, Hare JM. Cardiac nitric oxide production due
to angiotensin-converting enzyme inhibition decreases beta-adrenergic myocardial contractility in
patients with dilated cardiomyopathy. J Am Coll Cardiol 2001;38:429-435.
156
Huisamen B, Pêrel SJ, Friedrich SO, Salie R, Strijdom H, Lochner A. ANG II type I receptor
antagonism improved nitric oxide production and enhanced eNOS and PKB/Akt expression in hearts
from a rat model of insulin resistance. Mol Cell Biochem 2011;349:21-31.
157
Yalçin F, Aksoy FG, Muderrisoglu H, Sabah I, Garcia MJ, Thomas JD. Treatment of hypertension with
perindopril reduces plasma atrial natriuretic peptide levels, left ventricular mass, and improves
echocardiographic parameters of diastolic function. Clin Cardiol 2000;23:437-441.
158
Wachtell K, Bella JN, Rokkedal J, Palmieri V, Papademetriou V, Dahlöf B, Aalto T, Gerdts E,
Devereux RB. Change in diastolic left ventricular filling after one year of antihypertensive treatment:
The Losartan Intervention For Endpoint Reduction in Hypertension (LIFE) Study. Circulation
2002;105:1071-1076.
159
Mattioli AV, Zennaro M, Bonatti S, Bonetti L, Mattioli G. Regression of left ventricular hypertrophy
and improvement of diastolic function in hypertensive patients treated with telmisartan. Int J Cardiol
2004;97:383-388.
160
Cleland JG, Tendera M, Adamus J, Freemantle N, Polonski L, Taylor J; PEP-CHF Investigators. The
perindopril in elderly people with chronic heart failure (PEP-CHF) study. Eur Heart J 2006;27:23382345.
161
Yusuf S, Pfeffer MA, Swedberg K, Granger CB, Held P, McMurray JJV, Michelson EL, Olofsson B,
Östergren J, for the CHARM Investigators and Committees. Effects of candesartan in patients with
chronic heart failure and preserved left-ventricular ejection fraction: the CHARM-Preserved Trial.
Lancet. 2003;362:777–781.
162
Massie BM, Carson PE, McMurray JJ, Komajda M, McKelvie R, Zile MR, Anderson S, Donovan M,
Iverson E, Staiger C, Ptaszynska A. Irbesartan in patients with heart failure and preserved ejection
fraction. N Engl J Med 2008;359:2456-2467.
163
Shahin Y, Khan JA, Samuel N, Chetter I. Angiotensin converting enzyme inhibitors effect on
endothelial dysfunction: a meta-analysis of randomised controlled trials. Atherosclerosis 2011;216:7-16.
164
Mullen MJ, Clarkson P, Donald AE, Thomson H, Thorne SA, Powe AJ, Furuno T, Bull T, Deanfield
JE. Effect of enalapril on endothelial function in young insulin-dependent diabetic patients: a
randomized, double-blind study. J Am Coll Cardiol 1998;31:1330-1335.
165
Williams IL, Chowienczyk PJ, Wheatcroft SB, Patel AG, Sherwood RA, Shah AM, Kearney MT.
Divergent effects of angiotensin-converting enzyme inhibition on blood pressure and endothelial
function in obese humans. Diab Vasc Dis Res 2006;3:34-38.
166
Davis BR, Kostis JB, Simpson LM, Black HR, Cushman WC, Einhorn PT, Farber MA, Ford CE, Levy
D, Massie BM, Nawaz S; ALLHAT Collaborative Research Group. Heart failure with preserved and
reduced left ventricular ejection fraction in the antihypertensive and lipid-lowering treatment to prevent
heart attack trial. Circulation 2008;118:2259-2267.
167
Kitzman DW, Hundley WG, Brubaker PH, Morgan TM, Moore JB, Stewart KP, Little WC. A
randomized double-blind trial of enalapril in older patients with heart failure and preserved ejection
fraction: effects on exercise tolerance and arterial distensibility. Circ Heart Fail 2010;3:477-485.
168
Jay D, Hitomi H, Griendling KK. Oxidative stress and diabetic cardiovascular complications. Free
Radic Biol Med 2006;40:183-192.
169
Geraldes P, King GL. Activation of protein kinase C isoforms and its impact on diabetic complications.
Circ Res 2010;106:1319-1331.
170
Connelly KA, Kelly DJ, Zhang Y, Prior DL, Advani A, Cox AJ, Thai K, Krum H, Gilbert RE.
Inhibition of protein kinase C-beta by ruboxistaurin preserves cardiac function and reduces extracellular
matrix production in diabetic cardiomyopathy. Circ Heart Fail 2009;2:129-137.
171
Weber KT. Aldosterone in congestive heart failure. N Engl J Med 2001;345:1689-1697.
172
Whaley-Connell A, Johnson MS, Sowers JR. Aldosterone: role in the cardiometabolic syndrome and
resistant hypertension. Prog Cardiovasc Dis 2010;52:401-409.
173
Choi EY, Ha JW, Yoon SJ, Shim CY, Seo HS, Park S, Ko YG, Kang SM, Choi D, Rim SJ, Jang Y,
Chung N. Increased plasma aldosterone-to-renin ratio is associated with impaired left ventricular
longitudinal functional reserve in patients with uncomplicated hypertension. J Am Soc Echocardiogr
2008;21:251-256.
174
Daniel KR, Wells G, Stewart K, Moore B, Kitzman DW. Effect of aldosterone antagonism on exercise
tolerance, Doppler diastolic function, and quality of life in older women with diastolic heart failure.
Congest Heart Fail 2009;15:68-74.
175
Mottram PM, Haluska B, Leano R, Cowley D, Stowasser M, Marwick TH. Effect of aldosterone
antagonism on myocardial dysfunction in hypertensive patients with diastolic heart failure. Circulation
2004;110:558-565.
176
Edelmann F, Wachter R, Schmidt A, Kraigher-Krainer, Colantonio C, Kamke W, Duvinage A,
Stahrenberg R, Durstewitz K, Löffler M, Düngen HD, Tschöpe C, Herrmann-Lingen C, Halle M,
Hasenfuss G, Gelbrich G, Pieske B, for the Aldo-DHF Investigators. Effect of spironolactone on
diastolic function and exercise capacity in patients with heart failure with preserved ejection fraction.
The Aldo-DHF randomized controlled trial. JAMA 2013;309:781-791.
177
Little WC, Zile MR, Kitzman DW, Hundley WG, O'Brien TX, Degroof RC. The effect of alagebrium
chloride (ALT-711), a novel glucose cross-link breaker, in the treatment of elderly patients with
diastolic heart failure. J Card Fail 2005;11:191-195.
178
Thohan V, Koerner MM, Pratt CM, Torre GA. Improvements in diastolic function among patients with
advanced systolic heart failure utilizing alagebrium (an oral advanced glycation end-product cross-link
breaker). Circulation 2005;112(Suppl 2):2647.
179
Kass DA, Shapiro EP, Kawaguchi M, Capriotti AR, Scuteri A, deGroof RC, Lakatta EG. Improved
arterial compliance by a novel advanced glycation end-product crosslink breaker. Circulation
2001;104:1464-1470.
180
Hartog JW, Willemsen S, van Veldhuisen DJ, Posma JL, van Wijk LM, Hummel YM, Hillege HL,
Voors AA; BENEFICIAL investigators. Effects of alagebrium, an advanced glycation endproduct
breaker, on exercise tolerance and cardiac function in patients with chronic heart failure. Eur J Heart
Fail 2011;13:899-908.
181
Laufs U. Beyond lipid-lowering: effects of statins on endothelial nitric oxide. Eur J Clin Pharmacol
2003;58:719-731.
182
Ramasubbu K, Estep J, White DL, Deswal A, Mann DL. Experimental and clinical basis for the use of
statins in patients with ischemic and nonischemic cardiomyopathy. J Am Coll Cardiol 2008;51:415-426.
183
Mathur N, Ramasubbu K, Mann DL. Spectrum of pleiotropic effects of statins in heart failure. Heart
Fail Clin 2008;4:153-161.
184
Fukuta H, Sane DC, Brucks S, Little WC. Statin therapy may be associated with lower mortality in
patients with diastolic heart failure: a preliminary report. Circulation 2005;112:357-363.
185
Tehrani F, Morrissey R, Phan A, Chien C, Schwarz ER. Statin therapy in patients with diastolic heart f
ailure. Clin Cardiol 2010;33:E1-E5.
186
Gomez-Soto FM, Romero SP, Bernal JA, Escobar MA, Puerto JL, Andrey JL, Ruiz P, Gomez F.
Mortality and morbidity of newly diagnosed heart failure treated with statins: a propensity-adjusted
cohort study. Int J Cardiol 2010;140:210-218.
187
Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, Nathan DM;
Diabetes Prevention Program Research Group. Reduction in the incidence of type 2 diabetes with
lifestyle intervention or metformin. N Engl J Med 2002;346:393-403.
188
Kardassis D, Bech-Hanssen O, Schönander M, Sjöström L, Karason K. The influence of body
composition, fat distribution, and sustained weight loss on left ventricular mass and geometry in
obesity. Obesity 2012;20:605-611.
189
Peterson LR, Herrero P, Schechtman KB, Racette SB, Waggoner AD, Kisrieva-Ware Z, Dence C, Klein
S, Marsala J, Meyer T, Gropler RJ. Effect of obesity and insulin resistance on myocardial substrate
metabolism and efficiency in young women. Circulation 2004;109:2191-2196.
190
Cook SA, Varela-Carver A, Mongillo M, Kleinert C, Khan MT, Leccisotti L, Strickland N, Matsui T,
Das S, Rosenzweig A, Punjabi P, Camici PG. Abnormal myocardial insulin signalling in type 2 diabetes
and left-ventricular dysfunction. Eur Heart J 2010;31:100-111.
191
Kardassis D, Bech-Hanssen O, Schönander M, Sjöström L, Petzold M, Karason K. Impact of body
composition, fat distribution and sustained weight loss on cardiac function in obesity. Int J Cardiol.
2012;159:128-133.
192
Wong CY, Byrne NM, O'Moore-Sullivan T, Hills AP, Prins JB, Marwick TH. Effect of weight loss due
to lifestyle intervention on subclinical cardiovascular dysfunction in obesity (body mass index >30
kg/m2). Am J Cardiol 2006;98:1593-1598.
193
Leichman JG, Wilson EB, Scarborough T, Aguilar D, Miller CC 3rd, Yu S, Algahim MF, Reyes M,
Moody FG, Taegtmeyer H. Dramatic reversal of derangements in muscle metabolism and left
ventricular function after bariatric surgery. Am J Med 2008;121:966-973.
194
de las Fuentes L, Waggoner AD, Mohammed BS, Stein RI, Miller BV 3rd, Foster GD, Wyatt HR, Klein
S, Davila-Roman VG. Effect of moderate diet-induced weight loss and weight regain on cardiovascular
structure and function. J Am Coll Cardiol 2009;54:2376-2381.
195
Hammer S, Snel M, Lamb HJ, Jazet IM, van der Meer RW, Pijl H, Meinders EA, Romijn JA, de Roos
A, Smit JW. Prolonged caloric restriction in obese patients with type 2 diabetes mellitus decreases
myocardial triglyceride content and improves myocardial function. J Am Coll Cardiol 2008;52:10061012.
196
Viljanen AP, Karmi A, Borra R, Pärkkä JP, Lepomäki V, Parkkola R, Lautamäki R, Järvisalo M,
Taittonen M, Rönnemaa T, Iozzo P, Knuuti J, Nuutila P, Raitakari OT. Effect of caloric restriction on
myocardial fatty acid uptake, left ventricular mass, and cardiac work in obese adults. Am J Cardiol
2009;103:1721-1726.
197
Ikonomidis I, Mazarakis A, Papadopoulos C, Patsouras N, Kalfarentzos F, Lekakis J, Kremastinos DT,
Alexopoulos D. Weight loss after bariatric surgery improves aortic elastic properties and left ventricular
function in individuals with morbid obesity: a 3-year follow-up study. J Hypertens 2007;25:439-447.
198
Garza CA, Pellikka PA, Somers VK, Sarr MG, Collazo-Clavell ML, Korenfeld Y, Lopez-Jimenez F.
Structural and functional changes in left and right ventricles after major weight loss following bariatric
surgery for morbid obesity. Am J Cardiol 2010;105:550-556.
199
Lin CH, Kurup S, Herrero P, Schechtman KB, Eagon JC, Klein S, Dávila-Román VG, Stein RI, Dorn
GW 2nd, Gropler RJ, Waggoner AD, Peterson LR. Myocardial oxygen consumption change predicts
left ventricular relaxation improvement in obese humans after weight loss. Obesity 2011;19:1804-1812.
200
Kemppainen J, Tsuchida H, Stolen K, Karlsson H, Björnholm M, Heinonen OJ, Nuutila P, Krook A,
Knuuti J, Zierath JR. Insulin signalling and resistance in patients with chronic heart failure. J Physiol
2003;550:305-315.
201
Piepoli MF, Flather M, Coats AJ. Overview of studies of exercise training in chronic heart failure: the
need for a prospective randomized multicentre European trial. Eur Heart J 1998;19:830-841.
202
Smart N, Marwick TH. Exercise training for patients with heart failure: a systematic review of factors
that improve mortality and morbidity. Am J Med 2004;116:693-706.
203
Piepoli MF, Davos C, Francis DP, Coats AJ; ExTraMATCH Collaborative. Exercise training metaanalysis of trials in patients with chronic heart failure (ExTraMATCH). BMJ 2004;328:189.
204
Adamopoulos S, Parissis J, Kroupis C, Georgiadis M, Karatzas D, Karavolias G, Koniavitou K, Coats
AJ, Kremastinos DT. Physical training reduces peripheral markers of inflammation in patients with
chronic heart failure. Eur Heart J 2001;22:791-797.
205
Boulé NG, Haddad E, Kenny GP, Wells GA, Sigal RJ. Effects of exercise on glycemic control and body
mass in type 2 diabetes mellitus: a meta-analysis of controlled clinical trials. JAMA 2001;286:12181227.
206
Linke A, Schoene N, Gielen S, Hofer J, Erbs S, Schuler G, Hambrecht R. Endothelial dysfunction in
patients with chronic heart failure: systemic effects of lower-limb exercise training. J Am Coll Cardiol
2001;37:392-397.
207
Arbab-Zadeh A, Dijk E, Prasad A, Fu Q, Torres P, Zhang R, Thomas JD, Palmer D, Levine BD. Effect
of aging and physical activity on left ventricular compliance. Circulation 2004;110:1799-1805.
208
Edelmann F, Gelbrich G, Düngen HD, Fröhling S, Wachter R, Stahrenberg R, Binder L, Töpper A,
Lashki DJ, Schwarz S, Herrmann-Lingen C, Löffler M, Hasenfuss G, Halle M, Pieske B. Exercise
training improves exercise capacity and diastolic function in patients with heart failure with preserved
ejection fraction: results of the Ex-DHF (Exercise training in Diastolic Heart Failure) pilot study. J Am
Coll Cardiol 2011;58:1780-1791.
209
Smart NA, Haluska B, Jeffriess L, Leung D. Exercise Training in Heart Failure With Preserved Systolic
Function: A Randomized Controlled Trial of the Effects on Cardiac Function and Functional Capacity.
Congest Heart Fail 2012;18:295-301.
210
Rodrigues B, Cam MC, McNeill JH. Metabolic disturbances in diabetic cardiomyopathy. Mol Cell
Biochem 1998;180:53-57.
211
Russell RR 3rd, Yin R, Caplan MJ, Hu X, Ren J, Shulman GI, Sinusas AJ, Young LH. Additive effects
of hyperinsulinemia and ischemia on myocardial GLUT1 and GLUT4 translocation in vivo. Circulation
1998;98:2180-2186.
212
Liedtke AJ, DeMaison L, Eggleston AM, Cohen LM, Nellis SH. Changes in substrate metabolism and
effects of excess fatty acids in reperfused myocardium. Circ Res 1988;62:535-542.
213
Rijzewijk LJ, van der Meer RW, Smit JW, Diamant M, Bax JJ, Hammer S, Romijn JA, de Roos A,
Lamb HJ. Myocardial steatosis is an independent predictor of diastolic dysfunction in type 2 diabetes
mellitus. J Am Coll Cardiol 2008;52:1793-1799.
214
Szwed H, Sadowski Z, Pachocki R, Domzał-Bocheńska M, Szymczak K, Szydłowski Z, Paradowski A,
Gajos G, Kałuza G, Kulon I, Wator-Brzezińska A, Elikowski W, Kuźniak M. The antiischemic effects
and tolerability of trimetazidine in coronary diabetic patients. A substudy from TRIMPOL-1.
Cardiovasc Drugs Ther 1999;13:217-222.
215
Fragasso G, Palloshi A, Puccetti P, Silipigni C, Rossodivita A, Pala M, Calori G, Alfieri O, Margonato
A. A randomized clinical trial of trimetazidine, a partial free fatty acid oxidation inhibitor, in patients
with heart failure. J Am Coll Cardiol 2006;48:992-998.
216
Kantor PF, Lucien A, Kozak R, Lopaschuk GD. The antianginal drug trimetazidine shifts cardiac
energy metabolism from fatty acid oxidation to glucose oxidation by inhibiting mitochondrial longchain 3-ketoacyl coenzyme A thiolase. Circ Res 2000;86:580-588.
217
Williams FM, Tanda K, Kus M, Williams TJ. Trimetazidine inhibits neutrophil accumulation after
myocardial ischaemia and reperfusion in rabbits. J Cardiovasc Pharmacol 1993;22:828-833.
218
Ruixing Y, Wenwu L, Al-Ghazali R. Trimetazidine inhibits cardiomyocyte apoptosis in a rabbit model
of ischemia-reperfusion. Transl Res 2007;149:152-160.
219
Di Napoli P, Chierchia S, Taccardi AA, Grilli A, Felaco M, De Caterina R, Barsotti A. Trimetazidine
improves post-ischemic recovery by preserving endothelial nitric oxide synthase expression in isolated
working rat hearts. Nitric Oxide 2007;16:228-236.
220
Zhang L, Lu Y, Jiang H, Zhang L, Sun A, Zou Y, Ge J. Additional use of trimetazidine in patients with
chronic heart failure: a meta-analysis. J Am Coll Cardiol 2012;59:913-922.
221
Masoudi FA, Inzucchi SE, Wang Y, Havranek EP, Foody JM, Krumholz HM. Thiazolidinediones,
metformin, and outcomes in older patients with diabetes and heart failure: an observational study.
Circulation 2005;111;583-590.
222
Shioji K, Kishimoto C, Nakamura H, Masutani H, Yuan Z, Oka S, Yodoi J. Overexpression of
thioredoxin-1 in transgenic mice attenuates adriamycin-induced cardiotoxicity. Circulation
2002;106:1403-1409.
223
Salpeter S, Greyber E, Pasternak G, Salpeter E. Risk of fatal and nonfatal lactic acidosis with metformin
use in type 2 diabetes mellitus. Cochrane Database Syst Rev. 2006 Jan 25;(1):CD002967.
224
Eurich DT, McAlister FA, Blackburn DF, Majumdar SR, Tsuyuki RT, Varney J, Johnson JA. Benefits
and harms of antidiabetic agents in patients with diabetes and heart failure: systematic review. BMJ
2007;335:497.
225
Evans JM, Doney AS, AlZadjali MA, Ogston SA, Petrie JR, Morris AD, Struthers AD, Wong AK, Lang
CC. Effect of Metformin on mortality in patients with heart failure and type 2 diabetes mellitus. Am J
Cardiol 2010;106:1006-1010.
226
Wong AK, Symon R, Alzadjali MA, Ang DS, Ogston S, Choy A, Petrie JR, Struthers AD, Lang CC.
The effect of metformin on insulin resistance and exercise parameters in patients with heart failure. Eur
J Heart Fail 2012;14:1303-1310.
227
Iribarren C, Karter AJ, Go AS, Ferrara A, Liu JY, Sidney S, Selby JV. Glycemic control and heart
failure among adult patients with diabetes. Circulation 2001;103:2668-1673.
228
von Bibra H, Hansen A, Dounis V, Bystedt T, Malmberg K, Rydén L. Augmented metabolic control
improves myocardial diastolic function and perfusion in patients with non-insulin dependent diabetes.
Heart 2004;90:1483-1484.
229
ACCORD Study Group, Gerstein HC, Miller ME, Genuth S, Ismail-Beigi F, Buse JB, Goff DC Jr,
Probstfield JL, Cushman WC, Ginsberg HN, Bigger JT, Grimm RH Jr, Byington RP, Rosenberg YD,
Friedewald WT. Long-term effects of intensive glucose lowering on cardiovascular outcomes. New
Engl J Med 2011;364:818-828.
230
Jarnert C, Landstedt-Hallin L, Malmberg K, Melcher A, Ohrvik J, Persson H, Rydén L. A randomized
trial of the impact of strict glycaemic control on myocardial diastolic function and perfusion reserve: a
report from the DADD (Diabetes mellitus And Diastolic Dysfunction) study. Eur J Heart Fail
2009;11:39-47.
231
van Heerebeek L, Paulus WJ. The dialogue between diabetes and diastole. Eur J Heart Fail 2009;11:3-5.
232
von Bibra H, Diamant M, Scheffer PG, Siegmund T, Schumm-Draeger PM. Rosiglitazone, but not
glimepiride, improves myocardial diastolic function in association with reduction in oxidative stress in
type 2 diabetic patients without overt heart disease. Diab Vasc Dis Res 2008;5:310-318.
Summary
This thesis investigates several aspects of structural, functional and biochemical remodeling at
the myocardial and cardiomyocyte level in patients with heart failure with preserved ejection
fraction (HFpEF) and compared these measurements to measurements obtained in patients
with HF with reduced EF (HFrEF) as well as in patients operated for severe aortic stenosis
(AS). We used a clinical to basic translational “bed-to-bench” approach, relating clinical,
hemodynamic and echocardiographic parameters to structural, functional and biochemical
characteristics of the myocardium and cardiomyocytes through procurement of left ventricular
(LV) endomyocardial biopsies. In addition, the importance of metabolic risk factors, such as
diabetes mellitus type II, for diastolic LV dysfunction is investigated. According to the results
of this thesis, raised diastolic LV stiffness in the different patient groups results from distinct
pathophysiological mechanisms, which provides evidence that HFpEF and HFrEF actually
represent distinct HF phenotypes.
Heart failure with preserved ejection fraction (HFpEF) is widely prevalent and is associated
with a cumbersome prognosis, while its pathophysiological mechanisms are incompletely
understood and specific therapeutic strategies remain uncertain. It is however still debated
whether HFpEF and HFrEF are indeed distinct HF phenotypes despite prominent differences
in myocardial remodeling and clinical characteristics. To support the clinical distinction
between HFpEF and HFrEF, LV myocardial structure and function were compared in LV
endomyocardial biopsy samples of patients with HFpEF and HFrEF, which is described in
Chapter 2. Patients hospitalized for worsening HF were classified as having HFpEF (n=22;
LVEF 62 ± 2%) or HFrEF (n=22; LVEF 34 ± 2%). None of the patients had significant
coronary artery disease and transvascularly procured LV endomyocardial biopsies did not
demonstrate evidence of infiltrative or inflammatory myocardial disease. Biopsy samples
were analyzed with histomorphometry and electron microscopy. Single, permeabilized
cardiomyocytes were isolated from the biopsies, stretched to a sarcomere length of 2.2 μm to
measure intrinsic stiffness (passive force (Fpassive)), and activated with calcium (Ca2+)containing solutions to measure total force. HFpEF and HFrEF patients demonstrated
important differences in clinical, hemodynamic and echocardiographic characteristics. HFpEF
patients were more often hypertensive and obese and tended to be older than HFrEF patients.
LV peak systolic pressure, EF, LV wall thickness and myocardial stiffness were higher in
HFpEF. Furthermore, HFpEF patients demonstrated concentric remodeling with increased LV
mass index/LV end-diastolic volume index ratio (LVMI/LVEDVI), whereas HFrEF patients
had eccentric remodeling with lowered LVMI/LVEDVI ratio. HFpEF and HFrEF patients
also demonstrated important differences in myocellular structural and functional
characteristics. In HFpEF, cardiomyocyte diameter, myofibrillar density, myofilament Ca2+
sensitivity and cardiomyocyte Fpassive were higher than in HFrEF, while myocardial collagen
volume fraction (CVF) and total cardiomyocyte force were comparable. Following in-vitro
administration of protein kinase A (PKA), cardiomyocyte Fpassive was reduced in both HF
groups, but the drop in cardiomyocyte Fpassive was significantly larger in HFpEF, suggesting a
larger myofilament phosphorylation deficit in HFpEF. The giant sarcomeric protein titin
importantly determines cardiomyocyte Fpassive through shifts in expression of its stiff N2B and
compliant N2BA isoforms and through shifts in isoform phosphorylation status. In HFpEF,
the titin N2BA/N2B isoform expression ratio was lower than in HFrEF. We concluded that
important differences exist in both macroscopic and microscopic myocardial structural and
functional properties in LV myocardium of HFpEF and HFrEF patients, which suggests
HFpEF and HFrEF to be associated with phenotypically distinct myocardial and
cardiomyocyte abnormalities. These differences support the clinical discrimination of HF
patients into HFpEF and HFrEF groups.
Chapter 3. Excessive diastolic LV stiffness is an important contributor to HF in patients with
diabetes mellitus (DM). Diabetes is presumed to increase stiffness through myocardial
deposition of collagen and advanced glycation end products (AGEs). Cardiomyocyte Fpassive
also elevates stiffness, especially in HFpEF. We assessed the contribution to diastolic stiffness
of fibrosis, AGEs and cardiomyocyte Fpassive in diabetic HF patients with preserved or reduced
LVEF. In this study, LV endomyocardial biopsies were procured in both HFpEF and HFrEF
patients without significant coronary disease, who were hospitalized for worsening HF.
Biopsy samples were used for quantification of CVF and AGEs and for isolation of
cardiomyocytes to measure Fpassive. Diabetic HF patients had higher diastolic LV stiffness
irrespective of LVEF. The underlying mechanisms for increased diastolic LV stiffness in
diabetic HFrEF consisted predominantly of increased myocardial CVF and AGEs deposition.
In contrast, increased cardiomyocyte Fpassive was the most important contributor to raised
diastolic LV stiffness in diabetic HFpEF, with similar CVF and only a trend towards
increased myocardial AGEs deposition in diabetic HFpEF compared to non-diabetic HFpEF
patients. Furthermore, the higher Fpassive in diabetic HFpEF than in non-diabetic HFpEF
cardiomyocytes was paralleled by widening of the sarcomeric Z-disc, which was significantly
larger in diabetic HFpEF than in non-diabetic HFpEF cardiomyocytes. This study was the
first to report Z-disc widening in humans and because of the simultaneous elevation of Fpassive,
it suggests that Z-disc widening results from altered elastic properties of cytoskeletal proteins,
which pull at and open up adjacent Z-discs. Raised cardiomyocyte Fpassive was acutely lowered
with in-vitro administration of PKA with the decrement of Fpassive lowering being greatest in
diabetic HFpEF cardiomyocytes, suggesting the largest phosphorylation deficit in this
condition.
In this study, we concluded that the mechanisms responsible for the increased diastolic
stiffness of the diabetic heart differ in HFpEF and HFrEF. In diabetic HFpEF patients,
increased cardiomyocyte Fpassive more importantly underlies raised diastolic LV stiffness,
whereas in diabetic HFrEF patients, elevated fibrosis and myocardial AGEs deposition are
more important.
Chapter 4. In this editorial comment, the implications of transcriptional and posttranslational
modifications of the giant sarcomeric protein titin for diastolic function are discussed. Titin
importantly determines myocardial stiffness through modulation of cardiomyocyte Fpassive.
Titin spans a half sarcomere, running from the Z-disc to the M-band and functions as a
bidirectional spring responsible for early diastolic recoil and late diastolic resistance to
stretch. Cardiomyocyte titin-based elasticity can be adjusted through transcriptional and
posttranslational modifications. At the transcriptional level, titin modulates stiffness through
shifts in expression of its compliant N2BA (3.2-3.7 MDa) and stiff N2B (3.0MDa) isoforms,
which are co-expressed in the sarcomere. Titin-based stiffness is posttranslationally modified
by PKA and protein kinase G (PKG) mediated phosphorylation of the stiff N2B segment of
titin, which acutely lowers cardiomyocyte Fpassive in skinned human LV muscle strips, in
isolated human LV myofibrils and in isolated cardiomyocytes from HFpEF and HFrEF
patients. This acute adjustment of titin-based stiffness by PKA and PKG mediated
phosphorylation provides the cardiomyocyte with an “adaptive suspension”, regulating short
term modulation of myocardial stiffness, while extracellular matrix changes provide long term
modulation of myocardial stiffness.
Chapter 5. Transcriptional and posttranslational changes in the giant sarcomeric protein titin
underlie increased cardiomyocyte Fpassive, which importantly contributes to high diastolic
stiffness of failing myocardium. Shifts in titin isoform expression ratio and isoform
phosphorylation can alter cardiomyocyte Fpassive. In LV biopsies of HF patients, AS patients
and controls, we therefore related Fpassive of isolated cardiomyocytes to expression of titin
isoforms and to phosphorylation of titin and titin isoforms. Biopsies were procured by
transvascular technique in HF patients and perioperatively in AS patients or from explanted
hearts. All patients were free of significant coronary disease. Isolated, permeabilized
cardiomyocytes were stretched to 2.2 μm sarcomere length to measure Fpassive. Expression and
phosphorylation of titin isoforms were analyzed using gel electrophoresis with ProQ Diamond
and SYPRO Ruby stains. Cardiomyocyte Fpassive was higher in HF than in AS or controls,
while in-vitro administration of PKA acutely lowered Fpassive only in HF cardiomyocytes.
High Fpassive of HF cardiomyocytes also fell on administration of PKG and remained unaltered
on subsequent PKA treatment. The titin N2BA/N2B isoform expression ratio was higher in
HF than in controls and comparable in HF and AS patients, while a significant relation was
observed between N2BA/N2B ratio and LV end-diastolic wall stress. Overall titin
phosphorylation was comparable in HF and AS, but relative phosphorylation of the stiff N2B
titin isoform was significantly lower in HF than in AS. Because of comparable expression of
titin isoforms and similar overall phosphorylation of titin, the higher Fpassive of HF
cardiomyocytes compared to AS cardiomyocytes was attributed to a shift in titin isoform
phosphorylation with relative hypophosphorylation of the stiff N2B titin isoform.
Contribution to Fpassive of the thin filament and crossbridge interaction were ruled out because
of unaltered Fpassive after exposure to gelsolin (which removes the thin filament) and 2,3butanedione monoxine (which prevents cross bridge interactions). Cardiomyocyte Fpassive,
PKA-induced decrease in Fpassive and N2B phosphorylation deficit rose from AS to HFrEF to
HFpEF. A larger N2B titin isoform phosphorylation deficit in HFpEF explains both their
higher Fpassive and larger PKA-induced fall of Fpassive. We concluded that relative
hypophosphorylation of the stiff N2B titin isoform represents a novel mechanism responsible
for raised Fpassive of human HF cardiomyocytes. In HF, phosphorylation of the stiff N2B titin
isoform acutely lowers cardiomyocyte Fpassive, which could therefore have important
therapeutic implications, as stimulation of titin phosphorylation may represent a means to
relax the stiffened heart.
Chapter 6. In this review pathophysiological mechanisms for increased diastolic LV stiffness
in diabetic heart failure are discussed. Increased diastolic LV stiffness is recognized as the
earliest manifestation of DM-induced LV dysfunction. Mechanisms responsible for increased
diastolic LV stiffness in diabetic HF include myocardial fibrosis, AGEs, metabolic
disturbances, impaired Ca2+ homeostasis, endothelial dysfunction, myocardial oxidative
stress, impaired nitric oxide (NO) bioavailability, downregulated myocardial cyclic guanosine
monophosphate (cGMP) signaling and altered titin regulation. Furthermore, mechanisms
responsible for the elevated diastolic LV stiffness differ in diabetic HFpEF and diabetic
HFrEF patients. Myocardial fibrosis and deposition of AGEs are important determinants of
increased diastolic LV stiffness in diabetic HFrEF, whereas increased Fpassive of hypertrophied
cardiomyocytes is the main determinant of increased diastolic LV stiffness in diabetic HFpEF.
Downregulation of NO-mediated cGMP-PKG signaling, probably induced by vascular
inflammation and oxidative stress, could account for the high Fpassive of hypertrophied
cardiomyocytes observed in diabetic HFpEF patients.
Chapter 7. Diabetes mellitus (DM)-induced diastolic LV dysfunction is recognized as an
important determinant of morbidity and mortality in HF. In this chapter, the effects of DM on
diastolic LV function and its underlying mechanisms were identified in patients with severe
AS with or without DM who were referred for aortic valve replacement in the absence of
significant coronary artery disease. Preoperative Doppler echocardiography and
hemodynamics were implemented with perioperative LV biopsies. Myocardial CVF and
AGEs deposition were quantified by histomorphometry and immunohistochemistry, whereas
Fpassive was determined in isolated, permeabilized cardiomyocytes stretched to a sarcomere
length of 2.2 μm. Expression and phosphorylation of titin isoforms were analyzed with gel
electrophoresis with ProQ Diamond and SYPRO Ruby stains. Diabetic AS patients had
reduced LV end-diastolic distensibility as evident from higher LV end-diastolic pressure at
comparable LV end-diastolic volume index and attributed to higher myocardial CVF, more
AGEs deposition and higher cardiomyocyte Fpassive. The higher cardiomyocyte Fpassive in
diabetic AS patients was related to significant hypophosphorylation of the stiff N2B titin
isoform, while in-vitro administration of PKA normalized cardiomyocyte Fpassive. We
concluded that an additional impairment of LV end-diastolic distensibility is observed when
severe, symptomatic AS was associated with DM. This additional impairment related to both
structural and functional alterations of LV myocardium consisting of increased interstitial
collagen deposition, augmented intramyocardial AGE accumulation and higher intrinsic
cardiomyocyte stiffness. The contribution of increased cardiomyocyte Fpassive to raised
diastolic LV stiffness could be amenable to novel therapeutic strategies modulating
posttranslational titin-based stiffness through stimulation of titin phosphorylation.
Chapter 8. Prominent features of myocardial remodeling in HFpEF are high cardiomyocyte
Fpassive and cardiomyocyte hypertrophy. In experimental models, both reacted favourably to
raised PKG activity. In this chapter, we assessed myocardial PKG activity, its downstream
effects on cardiomyocyte Fpassive and cardiomyocyte diameter, and its upstream control by
cGMP, nitrosative/oxidative stress, and brain natriuretic peptide (BNP). To discern altered
control of myocardial remodeling by PKG, HFpEF was compared with HFrEF and AS. All
patients were free of significant coronary disease. More HFpEF patients were obese or had
DM. LV myocardial biopsies were procured transvascularly in HFpEF and HFrEF and
perioperatively in AS. Fpassive was measured in isolated, permeabilized cardiomyocytes before
and after PKG administration. Myocardial homogenates were used for assessment of PKG
activity, cGMP concentration and expression of proBNP-108 and nitrotyrosine. Nitrotyrosine
represents an indirect measure of nitrosative/oxidative stress. Additional quantitative
immunohistochemical analysis was performed for PKG activity, nitrotyrosine expression and
expression of soluble guanylate cyclase (sGC) and phosphodiesterase type 5A (PDE5A),
which breaks down cGMP, thereby terminating cGMP-PKG signaling. Our study showed that
relative to both AS and HFrEF, HFpEF patients had reduced myocardial PKG activity and
lower cGMP concentration, which related to higher cardiomyocyte Fpassive and increased
myocardial nitrotyrosine levels, indicative of raised nitrosative/oxidative stress. In all groups,
cardiomyocyte Fpassive was acutely lowered by in vitro administration of PKG, whereas
cardiomyocytes from HFpEF patients demonstrated the largest PKG-induced fall in Fpassive.
Reduced PKG activity and lower myocardial cGMP concentration in HFpEF did not result
from altered myocardial sGC or PDE5A expression, which were similar in all groups, or from
unequal natriuretic peptide (NP) expression, which was comparable in HFpEF and AS.
Downregulation of cGMP-PKG signaling in HFpEF was therefore most likely related to low
myocardial NO bioavailability because of high nitrosative/oxidative stress, which was almost
fourfold higher in HFpEF than in both HFrEF and AS. Oxidative stress is indeed known to
lower NO bioavailability and downregulate cGMP-PKG signaling. Increased
nitrosative/oxidative stress in HFpEF could have resulted from the high prevalence in HFpEF
of metabolic comorbidities. We concluded that HFpEF myocardium is characterized by
downregulated NO-cGMP-PKG signaling probably resulting from high nitrosative/oxidative
stress. Low PKG activity raises cardiomyocyte intrinsic stiffness and could be a potential
target for a specific HFpEF treatment strategy.
Chapter 9. In this review underlying molecular and cellular mechanisms for diastolic LV
dysfunction are discussed. HFpEF is widely prevalent, associated with a dismal prognosis and
incompletely understood pathophysiology, which precludes specific therapy. Prominent
features of HFpEF are impaired relaxation, increased diastolic LV stiffness and distinct
cardiac remodeling processes at the macroscopic, microscopic and ultrastructural level. In
HFpEF, increased diastolic LV stiffness can be explained by maladaptive changes in the
extracellular matrix and by elevated cardiomyocyte stiffness, which seem to be related to
myocardial inflammation, nitrosative/oxidative stress, endothelial dysfunction and
downregulation of NO-cGMP-PKG signaling. NO is crucially important for diastolic
function, since it potently enhances LV relaxation and distensibility through a variety of
mechanisms including cGMP-PKG dependent and independent mechanisms and modulation
of β-adrenergic responsiveness, S-nitrosylation signaling and myocardial bioenergetics.
Patients with HFpEF demonstrate a high prevalence of metabolic disorders, which may
contribute to enhanced myocardial inflammation, nitrosative/oxidative stress, downregulation
of NO bioavailability and NO-mediated cGMP-PKG signaling, thereby triggering diastolic
LV dysfunction. We concluded that targeting metabolic risk and improving endothelial
function, NO bioactivity and cGMP-PKG signaling can be promising strategies for specific
HFpEF treatment.
Future perspectives
Heart failure with preserved ejection fraction (HFpEF) demonstrates an increasing prevalence,
and currently approximately 50% of HF patients present with this type of HF. Although
clinical outcome is slightly better for HFpEF than for HFrEF, mortality associated with
HFpEF is considerable and most patients with HFpEF die from cardiovascular causes.
Unfortunately, in HFpEF, pathophysiological mechanisms, diagnostic and therapeutic
strategies remain uncertain and this is reflected in the lack of improvement of prognosis in
HFpEF over the last decennia. Advances in therapeutic strategies are only to be expected
when more comprehensive insight has been gained into underlying pathophysiological
mechanisms responsible for development and progression of HFpEF. HFpEF appears a
complex syndrome characterized by maladaptive changes in myocardial and cardiomyocyte
structural, functional and biochemical characteristics, probably induced in a multifactorial
fashion by a combination of adverse effects of myocardial inflammation, nitrosative/oxidative
stress, endothelial dysfunction, downregulation of NO bioavailability and NO-mediated
signaling, impaired myocardial bioenergetics, disturbed calcium handling and concentric
hypertrophy. Our recent translational studies have contributed to improved understanding of
HFpEF pathophysiology and exciting insights with potential direct applications for
therapeutic strategies have emerged. For instance, pharmacological agents that enhance
myocardial NO-cGMP-PKG signaling and reduce nitrosative/oxidative stress, in conjunction
with strategies that improve overall metabolic risk could have therapeutic potential in HFpEF
treatment. Nevertheless, because of the high complexity of HFpEF pathophysiology,
continuing research efforts, based on an integrative translational approach are undisputably
valuable to keep on moving this exciting field forward. This importance of integrative
physiology to gain insight into clinical pathophysiology in order to improve HFpEF outcome
has recently been acknowledged and awarded a significant grant from the European
Committee, to launch a large collaborative project, including 20 centers across Europe,
coordinated by Prof Dr. W.J. Paulus. This large scale integrating project includes both
experimental models of diastolic dysfunction as well as HFpEF patients and focuses on
pathophysiological mechanisms, improved diagnostic modalities and potential specific
HFpEF therapeutic strategies.