SUMMARY
Acute decompensated heart failure (ADHF) is responsible for more than 1 million hospital admissions each year in the US. Clinicians and scientists have developed therapeutic strategies that reduce mortality in patients with chronic heart failure (HF). Despite the widely appreciated magnitude of the ADHF problem, there is still a critical gap in our understanding of the cellular mechanisms involved and effective treatment strategies for hospitalized patients. Irrespective of the etiology, patients with ADHF present with similar symptoms (e.g. edema, altered hemodynamics and congestion) as multiple signaling pathways converge in a common phenotypic presentation. Investigations have shown that patients with ADHF have increased catecholamine levels, which cause chronic stimulation of β-adrenergic receptors. This overstimulation leads to chronic G-protein activation and perturbations in myocyte signaling, as the patient’s heart attempts to adapt to progressive HF. Over time, these compensatory signaling mechanisms ultimately fail, and maladaptive signaling prevails with progressive worsening of symptoms. This Review summarizes some of the changes that occur during chronic adrenergic stimulation, and examines how downstream contractile dysfunction and myocyte death can alter the prognosis of patients with HF hospitalized for acute events.
Keywords: apoptosis, calcium, cardiac, necrosis, nitric oxide
INTRODUCTION
Heart failure (HF) is the most frequent hospital diagnosis in the US and the final common pathway for a variety of pathologic conditions.1 Two key components of HF progression are structural and functional adaptations of cardiac myocytes. Initially, these changes in signal-transduction mechanisms are compensatory in light of increased cardiac demand; however, with time and as the disease progresses, they fail to correct the unfavorable changes and ultimately become maladaptive. β-Adrenergic-receptor (βAR) signaling has a key role in myocyte performance, and alterations in these signaling pathways are well-established disease pathways in HF.2,3
Despite extensive guidelines for managing patients with chronic HF, few prospective randomized trials have studied patients with acute decompensated HF (ADHF); hence, there is no consensus on how these patients should be managed. The lack of effective treatment options is partly a consequence of the poor understanding we have of mechanisms in acute HF and the lack of prospective clinical trials demonstrating a change in mortality in ADHF with treatment. The 5-year mortality for HF is roughly 50%,1 but recent data suggest that 1-year mortality in patients with HF who are admitted to hospital with ADHF could be as high as 33%.4 Conventionally, patients with ADHF can be divided into those who have a new diagnosis of HF, and those patients who have an acute decompensated event in the context of chronic HF. This Review focuses on the latter, larger group of HF patients who present with an acute decompensated event. The etiology of these patients is varied and a variety of circumstances can precipitate their hospitalization (Box 1).
Box 1 Possible reasons for hospitalization of patients with an acute decompensated event in the context of chronic heart failure
New-onset ischemia (macrovascular or microvascular)
Increased wall strain or increased intracardiac filling pressures (can be secondary to diastolic or systolic ventricular dysfunction)
Nonadherence (e.g. medications, fluid or diet)
Tachyarrhythmia ( e.g. recurrent atrial fibrillation, ventricular tachycardia)
Uncontrolled hypertension
NSAID usage
Excessive ethanol consumption or illicit drug usage
Endocrinopathies (e.g. diabetes, thyroid disease)
Worsening valvular disease with chronic pathological remodeling
Concurrent infection or upregulation of metabolic needs with a fixed cardiac output (e.g. urinary tract infection in a NYHA class IV patient)
Gastrointestinal bleed
Pregnancy
Chemotherapy
Constrictive disease and/or restrictive physiology
Infiltrative cardiomyopathy with an inability to compensate
Complications of hypertrophic cardiomyopathy
Uncompensated pulmonary hypertension or right-sided ventricular function
Progressively decreasing cardiac output (‘cold and dry’)
In this Review, we attempt to present a nonpartisan view of ADHF highlighting βAR signaling as one of the final common pathways that may directly contribute to ADHF and the high mortality associated with this condition. We focus on the decreasing number of myocytes and altered excitation–contraction coupling (ECC) seen in patients with ADHF, as these two processes are likely to have a considerable impact upon the disease process and the patient’s future constitution. We recognize, however, that these are not the only mechanisms that contribute to the disorder. Additional events that further exacerbate HF, such as fibrosis, pathological remodeling, dyssynchrony and hypertrophy, occur following the decrease in number of myocytes and worsening ECC associated with ADHF, leading to the progressive irreversibility of the disease.3 For demonstrative purposes only, we have selected the βAR to illustrate how its interactions with other signaling cascades can increase cell death through necrosis, apoptosis and perhaps autophagy (all three processes referred to as ‘cell death’, hereafter) and decrease contractile function (i.e. ECC; Figure 1). The mechanisms examined in this Review are not inclusive, nor is it the intent of the authors to imply that these mechanisms are more important than others not discussed.
Figure 1.

Altered adrenergic signaling following an acute episode of ADHF initiates a maladaptive cellular response. Although hemodynamics are usually improved before discharge, mortality subsequent to hospital discharge is increased. aSee Box 1. bSee reference 4. Abbreviation: ECC, excitation–contraction coupling.
HYPOTHESIS
Irrespective of the stage at which adverse changes in cell signaling occur (whether they involve G-protein coupled receptors, tyrosine kinase receptors, cytokine signaling or the nitroso–redox balance), the majority of these unique initiators converge on a limited number of downstream pathways to produce similar clinical end points—decrement in left ventricular (LV) performance, LV remodeling and congestion with increased systemic vascular resistance and in more infirm patients, a decrement in cardiac output.5 The observed decrease in cardiac function is, in part, a consequence of decreased contractile function and accelerated cell death. We propose that with each episode of ADHF, cardiac cell function does not completely recover to the prehospitalized level. Although patients may seem to be recovered at hospital discharge, they will have progressive deleterious myocyte signaling with each acute decompensated event. We propose that this pathological signaling will continue to accelerate, irrespective of whether the outward phenotype suggests improved hemodynamics. We hypothesize that ADHF is part of the progressive pathophysiology of chronic HF, not just an inevitable consequence of the disease. In fact, the current standard of care could actually worsen a patient’s long-term cardiac function, despite some perceived recovery in the short term (Table 1).
Table 1.
Medical therapies commonly used to treat patients with acute decompensated heart failure.
| Drug class | Short-term effect (i.e. 1–3 days) | Intermediate-term effect (i.e. 4–14 days) | Long-term effect (i.e. >14 days) |
|---|---|---|---|
| ACE inhibitors | ± | ++ | +++ |
| Digoxin | ± | + | ½+ |
| Loop diuretics | ++ | + | − |
| Aldosterone antagonist | ± | + | ++ |
| Morphine sulfate | ++ | + | + |
| Vasodilators | +++ | ++ | ++ |
| Inotropes | ++ | − − | − − − |
| β-Blockers | ± | + | +++ |
| Arterial dilators | ± | + | + |
Abbreviations: +, favorable effect; −, detrimental effect; ±, favorable and detrimental effects; ACE, angiotensin-converting enzyme.
Maladaptive adrenergic signaling and increased cell death
Despite intensive investigation, the complete role of adrenergic signaling in HF development and the mechanism of action of β-blockers as a HF therapy have not yet been completely elucidated. Originally, it was hypothesized that β-blockers repopulated cell membranes with βARs. This concept was partly based on the observation that the density of βARs was reduced in patients with HF, and functional coupling of the remaining receptors with their G proteins was impaired. This concept has not been generally supported, however, as some of the most efficacious β-blockers, such as carvedilol, have not been shown to resensitize βARs.6 Nevertheless, many β-blocker trials have demonstrated consistent improvement in HF with treatment, measured as improved LV function and attenuation in chamber size (‘reverse-remodeling’).7–9 These dichotomous results have led many investigators to conclude that the improvements seen in these clinical trials were mediated by more complicated and as yet poorly delineated, molecular and cellular signaling pathways.
β-Adrenergic receptor signaling
Aside from changes in myocyte cell size and tissue composition, an important component of LV remodeling during HF progression involves alteration of β-adrenergic signaling pathways. These pathways are important modulators of cardiac performance in normally functioning hearts and contribute to the equipoise between ECC and cell death. Catecholamines serve as endogenous agonists for both β1ARs and β2ARs, which when stimulated activate Gs proteins. Stimulation of Gs proteins increases cyclic AMP levels via activation of adenylyl cyclase. Of note, β1ARs use only Gs proteins, while β2ARs use two G proteins, Gi and Gs; β1ARs have been implicated as being pathological in HF, whereas β2ARs have generally been perceived to be protective.10,11 This paradigm may or may not be true in chronic human HF. With progressive chronic HF and increased sympathetic tone, chronic increases in agonist exposure leads to a decrease in the density of β1ARs, while the density of β2ARs actually increases as the patient’s condition worsens.12 This change in receptor density might change signaling preferences, as well as the efficacy or mechanism of action of specific β-blockers. Biased agonists may utilize different G-protein signaling pathways preferentially to maximize the effect of a given drug class.13 Despite downregulation of β1ARs in HF (the AR most implicated in worsening HF), empirical evidence has shown that the overall condition of patients with ADHF usually worsens, suggesting that β2AR and α1AR signaling could have a greater role in the progression of HF than previously appreciated. Perhaps the key to success with the next generation of β-blockers will lie with their ability to favorably alter the Gi–Gs protein balance and subsequent downstream signaling. Some newer therapies might actually circumvent the βARs or use G-protein-independent pathways to selectively activate more favorable signaling.13,14 Of note, clinical trials have demonstrated that time-dependent administration of selective and nonselective β-blockers leads to chronic ventricular reverse-remodeling and an attenuation in mortality not attributable to hemodynamic changes alone.6,8,9 Data also suggest that chronic administration (i.e. at least 3–6 months, preferably years) of β-blockers is essential for functional changes and a survival benefit to be seen.15
Following receptor activation, second-messenger-dependent protein kinases, including cyclic-AMP-dependent protein kinase (PKA), phosphorylate serine and threonine residues within the cytoplasmic loops and C-terminal-tail domains of the βARs. Phosphorylation of these sites is sufficient to impair receptor–G-protein coupling.16 An additional mechanism for desensitizing the receptor, which does not require agonist activation, involves G-protein receptor kinases (GRKs) and β-arrestin, a regulatory protein that initiates and attenuates various signaling pathways. One of the principle functions of GRKs in adrenergic signaling is to increase receptor affinity for β-arrestin through serine/threonine phosphorylation.17,18 Following GRK-mediated phosphorylation of the receptor, it is the binding of β-arrestin to the AR complex that leads to receptor desensitization and internalization. Irrespective of the HF model, chronic downstream adrenergic activation leads to an upregulation of GRKs, protein phosphatases, Gi protein and PKA expression, which is usually paralleled by a decrement in Gs expression and a reduction in sarcoplasmic reticulum calcium (Ca2+) levels and an increase in cytosolic Ca2+ concentration.3 Mediated through PKA and calcium/calmodulin-dependent protein kinase II (CaMKII) mechanisms, chronic β-adrenergic stimulation increases apoptosis by increasing sarcoplasmic reticulum stress, deregulating mitogen-activated protein kinases, and unbalancing or making phosphoinositide 3-kinase–protein kinase B (AKT) signaling ineffective by inducing changes in the location and phosphorylation of AKT, thus, altering its protective mechanisms (Figure 2).19,20 These intracellular events lead to an observable change in the functional capacity of the failing heart with an observed decrement in myocyte contractile function and a decrease in the total number of myocytes.
Figure 2.
Chronic βAR stimulation perturbs normal signaling (increasing PKA, CaMKII, and diastolic Ca2+ levels, and making PI3K/AKT signaling ineffective) leading to many maladaptive mechanisms including mitochondrial swelling and cytochrome c release. The release of cytochrome c ultimately activates the caspases, resulting in increased apoptosis and decreased contractile function. With persistent decompensated heart failure there is progressive decrease in myocyte contractile function, while the total number of myocytes decreases secondary to apoptosis, adrenergic-mediated necrosis and autophagy. Blue lines represent direct βAR activation, while green lines represent G-protein-independent signaling. Abbreviations: AKT, protein kinase B; βAR, β-adrenergic receptor; Ca2+, calcium ions; CaMKII, calcium/calmodulin-dependent protein kinase II; cAMP, cyclic AMP; GRK, G-protein receptor kinases; IP3R, inositol triphosphate receptor; PI3K, phosphoinositide 3-kinase; PKA, cyclic-AMP-dependent protein kinase; PLB, phospholamban; RGS, regulator of G-protein signaling; RyR, ryanodine receptor; SR, sarcoplasmic reticulum.
Myocardial tissue changes and cell death
A hallmark feature of HF progression in both animal models and humans is the time-dependent adaptation of myocardium that leads to changes in cardiac myocyte performance and subsequent changes in myocardial tissue composition.21 For example, ventricular chamber dilation, myocyte hypertrophy and interstitial fibrosis are all well-recognized features of HF.22 Elevated sympathetic nervous system activity is also common, as the body attempts to maintain cardiac output and systemic perfusion. Each of these changes can be considered as adaptations for acute increases in cardiac demand, but it is likely that they also contribute to progressive pump failure and are considered deleterious over the long term.23 Recent studies have shown evidence of myocyte ‘drop out’ during HF progression, suggesting that myocyte populations continuously decline in the failing myocardium, increasing the demand on those remaining.24,25 These findings are consistent with increasing levels of cell death as a result of progressive detrimental molecular signaling and not as a consequence of hemodynamic dysregulation.26 During pathological remodeling, an absolute reduction in the total number of myocytes contributes to the outward phenotype. This cell loss leads to decreased wall thickness, increased wall strain and dyssynchrony, all of which add to subsequent additional cell loss and progressive maladaptive physiology as the patient’s ventricular function continues to decline.27
Apoptosis
Several investigators have suggested that apoptotic signaling is increased 232-fold in patients with progressive HF compared with matched controls.27,28 We believe the rate of apoptosis is also increased in patients with ADHF. The majority of apoptotic signaling cascades coalesce with the activation of a group of constitutively active proteases called caspases that are activated by proteolytic cleavage or dimerization. When activated, caspases coordinate signaling cascades that ultimately lead to myocyte death. Caspase activation can occur through two interwoven pathways within myocytes and fibroblasts. The extrinsic pathway is associated with increasing inflammatory response and cytokine signaling, and is known to be upregulated in some types of ADHF. The activation of this pathway usually requires agonists to bind to cell-surface receptors (‘death receptors’) such as the Fas and tumor necrosis factor (TNF) receptors, which then interact with adaptor molecules, transducing the signal. This receptor signaling then activates pro-caspase-8, which subsequently acts upon BH3-interacting domain death agonist (BID) and caspase-3. In turn, caspase-3 activates caspase-7 and, thereafter, both act as ‘executioner’ or effector caspases by translocating to the nucleus and degrading essential components. Some data suggest that nonspecific β-blockers decrease TNF-induced apoptosis in human cells.29
The other ‘intrinsic’ apoptotic pathway is mediated though perturbations in Ca2+ signaling and mitochondrial and sarcoplasmic reticulum stress. Eloquent, early studies in humans demonstrated aggressive apoptosis and upregulation of proteases in specific cardiomyopathies. Through a variety of signaling mechanisms, cytochrome c translocates from the mitochondria to the cytosol. Once in the cytosol, cytochrome c binds to apoptosis protease activating factor 1 (APAF-1) and pro-caspase-9, forming the apoptosome. This protein complex then cleaves pro-caspase-9 releasing active caspase-9, which subsequently activates caspase-3, leading to DNA fragmentation and cell death.23,28,30–33
CONTRACTILITY IN ACUTE DECOMPENSATED HEART FAILURE
New data suggest that apoptotic signaling is not as simple as first thought—that the apoptotic cascade was either active (cell death) or inactive (no cell death).34 It is now recognized that apoptotic signaling cascades can decrease contractile function before cell death by cleaving contractile proteins.35,36 In this regard, overexpression of caspase-3 can directly reduce LV contractile function.36 In addition, caspase inhibition has been shown to improve contractile function by attenuating contractile protein loss.37 A decreased contractile state with activated apoptotic signaling could be an intermediate state of ‘cell death’ in which cells can progress towards death, continue to be dysfunctional or, in the correct environment, recover. In transgenic animals, overexpression of caspase-8 revealed a direct correlation between expression of this protein and increased myocyte death.24 In addition, even at low levels of caspase-8 expression, significant failure was induced in the animal model of chronic HF.24 Similar lines of investigation have indicated that activating apoptosis intermediates also results in development of severe cardiomyopathies and contributes to decreased contractile function.35,36 Of note, altered Ca2+ handling can lead to apoptosis, but also contribute to contractile dysfunction.37,38 The caspase cascade and Ca2+ mishandling are examples of how apoptosis and contractile dysfunction intersect.
Excitation–contraction coupling
The initiating event in ECC is firing of the action potential, which leads to Ca2+ influx via the L-type Ca2+ current (ICa-L). This Ca2+ influx, also known as the trigger Ca2+, activates the Ca2+ release channels found on the sarcoplasmic reticulum—the ryanodine receptors—causing a graded release of Ca2+ from the sarcoplasmic reticulum, a process termed Ca2+-induced Ca2+ release. The myofilaments are, in turn, activated by this increase in Ca2+concentration such that there is a direct, though nonlinear, relationship between free Ca2+ concentration and contraction. The Ca2+ sensitivity of the myofilaments can also be altered (e.g. by β-adrenergic stimulation) to further regulate contraction and relaxation. After activation, the Ca2+ concentration returns to diastolic levels via uptake by the sarcoplasmic/endoplasmic reticulum calcium ATPase/phospholamban (SERCA/PLB) complex and the Na+–Ca2+ exchanger (NCX), a process that also contributes to the action potential waveform.39
The cardiac dysfunction in HF can be observed at the cellular level as a blunted force-frequency response.40 Much of this dysfunction is due to changes in the expression of certain proteins such as transient outward potassium channels, inward rectifier potassium channels, NCX, ryanodine receptors and SERCA, among others.39 These protein expression changes lead to increased action potential duration, reduced Ca2+ transient and shortening amplitude, and slowed rates of cytosolic Ca2+ decline and relaxation.41 Clinically, medications that further attenuate Ca2+ handling (e.g. calcium-channel blockers) increase mortality in patients who already have decreased ventricular function.
Calcium/calmodulin-dependent protein kinase II
The δ isoform of CaMKII is the predominant isoform found within myocytes. CaMKII is a serine/threonine kinase that is activated by Ca2+ and calmodulin. Within myocytes, the Ca2+ transient leads to oscillations of Ca2+–calmodulin concentration,42 which can activate CaMKII on a beat-per-beat basis. In addition, autophosphorylation of CaMKII keeps calmodulin bound to the kinase to retain activity even after Ca2+ levels decrease. Intriguingly, independent of its effects on Ca2+ handling, sustained βAR stimulation can increase CaMKII activity directly.43 While CaMKII can modulate many proteins involved in ECC, it’s major action is the regulation of ryanodine receptors.44 CaMKII-mediated phosphorylation of ryanodine receptors increases diastolic Ca2+ leak from the sarcoplasmic reticulum, which is disadvantageous as it reduces sarcoplasmic reticulum Ca2+ load, a main determinant of contractility,45 and increases the propensity for arrhythmias.46 Interestingly, the leak associated with increased βAR stimulation is attributable to increased activity of CaMKII and not PKA.43 Importantly, increasing Ca2+ flux via ryanodine receptors alone is not sufficient to cause arrhythmogenic Ca2+ waves. Another stimulus is, therefore, needed to keep sarcoplasmic reticulum Ca2+ content at a level sufficient for Ca2+ wave production.47 We believe that in ADHF, continued βAR stimulation activates CaMKII, causing diastolic Ca2+ leak from the sarcoplasmic reticulum via ryanodine receptor phosphorylation, and that PKA activation maintains sarcoplasmic reticulum Ca2+ at a level sufficiently high for the production of arrhythmogenic Ca2+ waves. In addition to alterations in Ca2+ handling, CaMKII can also phosphorylate ion channels. For example, CaMKII can phosphorylate Na+ channels, enhancing the late (persistent) Na+ current, which leads to a prolonged QT interval and increased Na+ concentration.48 These effects lead to monomorphic and polymorphic ventricular tachycardia and arrhythmias, which can be further exacerbated by hypokalemia and hypomagnesemia in patients with ADHF. Sustained CaMKII activation seen in ADHF could, therefore, lead to electrophysiological changes that make the patient more prone to sudden cardiac death (Figure 3).49
Figure 3.
Possible mechanisms of reduced contractility in acute decompensated heart failure. Chronic βAR stimulation activates CaMKII, NOS2 expression and increased ROS production. In addition, NOS1 and NOS3 expression and localization might be altered. The ensuing difference in spatial and temporal nitric oxide production, coupled with increased ROS production, results in RNS formation and altered nitroso–redox balance. Stimulation (blue) or inhibition (red) of targeted excitation–contraction coupling proteins will result in contractile dysfunction and increased diastolic Ca2+ concentration. Abbreviations: βAR, β-adrenergic receptor; Ca2+, calcium ions; CaMKII, calcium/calmodulin-dependent protein kinase II; GRK, G-protein receptor kinase; Na+, sodium ions; NCX, Na+–Ca2+ exchanger; NOS1, neuronal nitric oxide synthase; NOS2, inducible nitric oxide synthase; NOS3, endothelial nitric oxide synthase; PLB, phospholamban; RNS, reactive nitrogen species; ROS, reactive oxygen species; RyR, ryanodine receptor; SR, sarcoplasmic reticulum.
Reactive oxygen species
Studies have shown that chronic activation of the βAR pathway can lead to increased production of reactive oxygen species (ROS). This effect is most probably mediated via downregulation of copper–zinc-superoxide dismutase.50 Moreover, during acute decompensated episodes there is an inflammatory response caused by an increase in diastolic wall stress because of pressure and volume overload. This increase in wall stress leads to the production of the inflammatory cytokines TNF, interleukin 1 and interleukin 6.51 Another possible cause of cytokine production is mesenteric venous congestion or bowel-wall edema, or hypoperfusion, which lead to intestinal bacterial translocation, endotoxin release, and subsequently an immune response.52 Indeed, a study demonstrated a transient increase in cytokine production in patients admitted for ADHF that peaked 12 h after admission and steadily decreased until steady-state was achieved 2 weeks after admission.53 An increase in cytokine levels also leads to increased production of ROS.54 We, therefore, believe that ROS production is greatly increased during ADHF.
Many studies have shown that direct exposure to ROS will lead to contractile dysfunction.55,56 This negative inotropic effect of ROS occurs via modulation of sarcoplasmic reticulum function, membrane currents, myofilaments and direct G-protein signaling. ROS can decrease SERCA activity, reducing Ca2+ uptake and sarcoplasmic reticulum Ca2+ load.57 An increase in cellular ROS concentration can also increase NCX activity.58 This increase in NCX activity when coupled with an increase in cytosolic Na+ can lead to an increase in reverse-mode NCX. Consistent with this hypothesis, inhibition of reverse-mode NCX inhibits the increase in diastolic force that normally occurs after ROS exposure.56 In addition, raised ROS levels lead to an increase in ryanodine receptor open probability (as with CaMKII).59 The threshold stimulating Ca2+ leak from the sarcoplasmic reticulum can, thus, be reduced via ROS-mediated (and CaMKII) modulation of ryanodine receptor activity.
The Na+ channel is also affected by changes in ROS concentration. ROS-induced activation of the late Na+ current leads to prolonged action potential duration, increased cellular Na+ concentration and early afterdepolarizations.60 Effects related to both Na+ and Ca2+ channel changes occur early in ROS-induced dysfunction, and the alterations in Ca2+ handling outlined previously are responsible for the early cellular injury. The late phase of injury is most probably caused by myofilament changes; it is hypothesized that the increase in Ca2+ activates calpain, leading to the degradation of the troponin complex, which influences myofilament sensitivity.56 ROS (H2O2 in particular) can also decrease GS protein activity, leading to decreased adenylyl cyclase activation following βAR stimulation.55 The net effects of increased ROS concentration are, thus, a shift in the Ca2+ pool location, from the sarcoplasmic reticulum to the cytosol, a decrease in Ca2+-induced Ca2+ release, an increase in Na+ concentration (which ultimately leads to increased diastolic tension), reduced developed tension, a decreased βAR responsive, and ultimately the development of arrhythmias (Figure 3).
Reactive nitrogen species
Nitric oxide (NO) and related congeners are signaling molecules that modulate cardiac contractility. Cardiac myocytes express three isoforms of NO synthase. Neuronal NO synthase (nNOS or NOS1) and endothelial NO synthase (eNOS or NOS3) are constitutively expressed and produce low amounts of NO, while inducible NO synthase (iNOS or NOS2) is not normally expressed in cardiac myocytes. NOS2 expression is induced during an inflammatory response, common in patients with HF,61 and once expressed, produces much higher levels of NO than do NOS1 and NOS3.62 Sustained βAR stimulation can upregulate NOS2 expression and the formation of reactive nitrogen species (e.g. peroxynitrite).63 Interestingly, cyclic AMP can stabilize NOS2 mRNA, resulting in even greater NOS2 concentrations.64 We believe that NOS2 expression is increased substantially during ADHF as a result of the activated inflammatory-mediated mechanisms and sustained βAR stimulation. Studies have shown that once expressed, NOS2 diminishes the cellular response to βAR stimulation via alterations in Ca2+ handling.62,65,66 Overexpression of NOS2 and the resulting elevated production of NO and ROS will in turn lead to the formation of peroxynitrite. We have found that the peroxynitrite donor 3-morpholinosyndnomine (SIN-1) decreases the βAR response, predominantly through activation of protein phosphatases that decrease the level of PLB phosphorylation.67 This pathway might prove to be central in the βAR hyporesponsiveness observed in ADHF. Interestingly, failing human myocytes treated with an NOS2 inhibitor had a near-normal response to βAR stimulation,66 indicating that NOS2 signaling could also have a role in βAR receptor desensitization, possibly via altered palmitoylation.68
NOS1 and NOS3 signaling might also be involved in the pathogenesis of ADHF. NOS1 and NOS3 signaling are in fact dissimilar, targeting different ECC proteins and leading to diverse functional effects.61 NOS1 and NOS3 provide some degree of protection for patients with HF;69,70 however, as HF develops the expression of NOS3 decreases,71 resulting in a negation of its protective role. Interestingly, in the failing heart, NOS1 translocates from the sarcoplasmic reticulum to the cavealoe.71 We believe that this translocation is due to the loss of NOS3—with decreased NOS3 expression, NOS1 takes up a functional role similar to that of NOS3 in order to maintain protection and decrease the detrimental effects of sustained βAR stimulation. In fact, a study has shown that translocation of NOS1 leads to a reduced βAR response.72 With the increased NO production via NOS2, we envision that the protective roles of NOS1 and NOS3 are further diminished in HF. Specifically, the compartmentalized and tightly regulated NO production via NOS1 and NOS3 seen in the normal heart will be lost with the high NO concentration resulting from increased NOS2 expression (Figure 3). This altered NOS1 and NOS3 signaling may also enhance GRK-mediated desensitization of the βAR receptors.73
It is important to note that the nitroso–redox balance—the equilibrium between NO and ROS concentrations—is tightly regulated in normal hearts and altered in HF.74 The altered spatial and temporal production of NO and ROS leads to peroxynitrite formation, which has profound functional effects on the response to βAR stimulation.
CONCLUSIONS
During pathological remodeling, it is now known that an absolute reduction in the total number of myocytes contributes to HF progression.27,28 Furthermore, apoptotic signaling cascades may decrease contractile function before cell death by cleaving contractile proteins.34,35 Additional changes in intracellular Ca2+ handling have a profound effect in ADHF. The decrease in internal sarcoplasmic reticulum Ca2+ levels40,75 and concordant increase in cytosolic Ca2+concentration leads to a decrease in contractile function and an increase in apoptotic signaling. The result is fewer myocytes, and a decrease in contractile function of the remaining myocytes. In addition, progressive nitroso–redox biology activates mitochondrial apoptotic machinery, while simultaneously having a deleterious effect on ECC.66 A switch in βAR signaling is a key feature of progressive HF. In order for patients to truly recover, there must be a change in the chronic deleterious signaling mechanisms in the remaining myocytes. Our data suggest that fibrosis is not significantly reversed with physiologic or functional recovery, nor does the number of myocytes increase significantly.76 That clinicians and basic scientists will be able to retread these maladaptive signaling pathways is unlikely. As such, innovative ways to ‘reverse’ signaling in sick hearts is required. Future therapies targeted towards cell death and altered ECC present hope for improved reverse remodeling and better patient outcomes.
KEY POINTS.
Acute decompensated heart failure (ADHF) is a large clinical problem, yet little is known about the basic mechanisms associated with disease progression; the final common pathway in ADHF is increased adrenergic signaling, independent of ADHF etiology
Long-term (or maladaptive) adrenergic signaling in ADHF will lead to activation of calcium/calmodulin-dependent protein kinase, increased cytokine levels, nitroso–redox imbalance, and a shift in Ca2+ pools from the sarcoplasmic reticulum to the cytosol
Maladaptive adrenergic signaling leads to caspase activation, cytochrome c translocation and progressive cell death, leading in turn to progressive worsening of heart function
Maladaptive adrenergic signaling alters cardiac excitation–contraction coupling, which leads to decreased contractility and an increased propensity for arrhythmias
Therapeutic targeting of these two common pathways (cell death and altered excitation–contraction coupling) of ADHF may improve patient recovery
Acknowledgments
We thank Dr Jonathan Davis for critically reading the manuscript. Dr TS Elton (R01 HL048848), Dr DS Feldman (R01 HL084498), and Dr MT Ziolo (R01 HL079283) are supported by the National Institutes of Health.
Footnotes
Competing interests
The authors declared no competing interests.
References
- 1.Heart Failure Society of America. HFSA 2006 Comprehensive heart failure practice guideline. J Card Fail. 2006;12:e1–2. doi: 10.1016/j.cardfail.2005.11.005. [DOI] [PubMed] [Google Scholar]
- 2.Tilley DG, Rockman HA. Role of β-adrenergic receptor signaling and desensitization in heart failure: new concepts and prospects for treatment. Expert Rev Cardiovasc Ther. 2006;4:417–432. doi: 10.1586/14779072.4.3.417. [DOI] [PubMed] [Google Scholar]
- 3.Feldman DS, et al. Mechanisms of disease: β-adrenergic receptors—alterations in signal transduction and pharmacogenomics in heart failure. Nat Clin Pract Cardiovasc Med. 2005;2:475–483. doi: 10.1038/ncpcardio0309. [DOI] [PubMed] [Google Scholar]
- 4.Jong P, et al. Prognosis and determinants of survival in patients newly hospitalized for heart failure: a population-based study. Arch Intern Med. 2002;162:1689–1694. doi: 10.1001/archinte.162.15.1689. [DOI] [PubMed] [Google Scholar]
- 5.Feldman D, et al. Management strategies for stage-d patients with acute heart failure. Clin Cardiol. 2007 doi: 10.1002/clc.20251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Doughty RN, et al. Left ventricular remodeling with carvedilol in patients with congestive heart failure due to ischemic heart disease. Australia–New Zealand Heart Failure Research Collaborative Group. J Am Coll Cardiol. 1997;29:1060–1066. doi: 10.1016/s0735-1097(97)00012-0. [DOI] [PubMed] [Google Scholar]
- 7.CIBIS-II Investigators and Committees. The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial. Lancet. 1999;353:9–13. [PubMed] [Google Scholar]
- 8.MERIT-HF Study Group. Effect of metoprolol CR/XL in chronic heart failure: Metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF) Lancet. 1999;353:2001–2007. [PubMed] [Google Scholar]
- 9.Packer M, et al. The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. US Carvedilol Heart Failure Study Group. N Engl J Med. 1996;334:1349–1355. doi: 10.1056/NEJM199605233342101. [DOI] [PubMed] [Google Scholar]
- 10.Lohse MJ, et al. What is the role of β-adrenergic signaling in heart failure? Circ Res. 2003;93:896–906. doi: 10.1161/01.RES.0000102042.83024.CA. [DOI] [PubMed] [Google Scholar]
- 11.Vatner SF, et al. β-Adrenergic receptor signaling: an acute compensatory adjustment-inappropriate for the chronic stress of heart failure? Insights from Gsalpha overexpression and other genetically engineered animal models. Circ Res. 2000;86:502–506. doi: 10.1161/01.res.86.5.502. [DOI] [PubMed] [Google Scholar]
- 12.Bristow MR, et al. β-Adrenergic pathways in nonfailing and failing human ventricular myocardium. Circulation. 1990;82(2 suppl):I12–I25. [PubMed] [Google Scholar]
- 13.Rajagopal K, et al. β-Arrestin2-mediated inotropic effects of the angiotensin II type 1A receptor in isolated cardiac myocytes. Proc Natl Acad Sci USA. 2006;103:16284–16289. doi: 10.1073/pnas.0607583103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Noma T, et al. β-Arrestin-mediated β1-adrenergic receptor transactivation of the EGFR confers cardioprotection. J Clin Invest. 2007;117:2445–2458. doi: 10.1172/JCI31901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ojeda S, et al. Short- and long-term results of a programme for the prevention of readmissions and mortality in patients with heart failure: are effects maintained after stopping the programme? Eur J Heart Fail. 2005;7:921–926. doi: 10.1016/j.ejheart.2005.05.009. [DOI] [PubMed] [Google Scholar]
- 16.Lefkowitz RJ. G protein-coupled receptor kinases. Cell. 1993;74:409–412. doi: 10.1016/0092-8674(93)80042-d. [DOI] [PubMed] [Google Scholar]
- 17.Iaccarino G, et al. Elevated myocardial and lymphocyte GRK2 expression and activity in human heart failure. Eur Heart J. 2005;26:1752–1758. doi: 10.1093/eurheartj/ehi429. [DOI] [PubMed] [Google Scholar]
- 18.Luttrell LM, et al. β-Arrestin-dependent formation of β2 adrenergic receptor-Src protein kinase complexes. Science. 1999;283:655–661. doi: 10.1126/science.283.5402.655. [DOI] [PubMed] [Google Scholar]
- 19.Dent MR, et al. Alterations in both death and survival signals for apoptosis in heart failure due to volume overload. J Mol Cell Cardiol. 2007;43:726–732. doi: 10.1016/j.yjmcc.2007.09.001. [DOI] [PubMed] [Google Scholar]
- 20.Muraski JA, et al. Pim-1 regulates cardiomyocyte survival downstream of Akt. Nat Med. 2007;13:1467–1475. doi: 10.1038/nm1671. [DOI] [PubMed] [Google Scholar]
- 21.Singh K, et al. Adrenergic regulation of cardiac myocyte apoptosis. J Cell Physiol. 2001;189:257–265. doi: 10.1002/jcp.10024. [DOI] [PubMed] [Google Scholar]
- 22.Beltrami CA, et al. Structural basis of end-stage failure in ischemic cardiomyopathy in humans. Circulation. 1994;89:151–163. doi: 10.1161/01.cir.89.1.151. [DOI] [PubMed] [Google Scholar]
- 23.Communal C, Colucci WS. The control of cardiomyocyte apoptosis via the β-adrenergic signaling pathways. Arch Mal Coeur Vaiss. 2005;98:236–241. [PubMed] [Google Scholar]
- 24.Wencker D, et al. A mechanistic role for cardiac myocyte apoptosis in heart failure. J Clin Invest. 2003;111:1497–1504. doi: 10.1172/JCI17664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Xiang Y, Kobilka BK. Myocyte adrenoceptor signaling pathways. Science. 2003;300:1530–1532. doi: 10.1126/science.1079206. [DOI] [PubMed] [Google Scholar]
- 26.Beltrami CA, et al. The cellular basis of dilated cardiomyopathy in humans. J Mol Cell Cardiol. 1995;27:291–305. doi: 10.1016/s0022-2828(08)80028-4. [DOI] [PubMed] [Google Scholar]
- 27.Olivetti G, et al. Apoptosis in the failing human heart. N Engl J Med. 1997;336:1131–1141. doi: 10.1056/NEJM199704173361603. [DOI] [PubMed] [Google Scholar]
- 28.Narula J, et al. Apoptosis in myocytes in end-stage heart failure. N Engl J Med. 1996;335:1182–1189. doi: 10.1056/NEJM199610173351603. [DOI] [PubMed] [Google Scholar]
- 29.Rossig L, et al. Congestive heart failure induces endothelial cell apoptosis: protective role of carvedilol. J Am Coll Cardiol. 2000;36:2081–2089. doi: 10.1016/s0735-1097(00)01002-0. [DOI] [PubMed] [Google Scholar]
- 30.Chang J, et al. Activation of Rho-associated coiled-coil protein kinase 1 (ROCK-1) by caspase-3 cleavage plays an essential role in cardiac myocyte apoptosis. Proc Natl Acad Sci USA. 2006;103:14495–14500. doi: 10.1073/pnas.0601911103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Cardone MH, et al. Regulation of cell death protease caspase-9 by phosphorylation. Science. 1998;282:1318–1321. doi: 10.1126/science.282.5392.1318. [DOI] [PubMed] [Google Scholar]
- 32.Mallat Z, et al. Evidence of apoptosis in arrhythmogenic right ventricular dysplasia. N Engl J Med. 1996;335:1190–1196. doi: 10.1056/NEJM199610173351604. [DOI] [PubMed] [Google Scholar]
- 33.Bing OH. Hypothesis: apoptosis may be a mechanism for the transition to heart failure with chronic pressure overload. J Mol Cell Cardiol. 1994;26:943–948. doi: 10.1006/jmcc.1994.1115. [DOI] [PubMed] [Google Scholar]
- 34.Kitsis RN, Mann DL. Apoptosis and the heart: a decade of progress. J Mol Cell Cardiol. 2005;38:1–2. doi: 10.1016/j.yjmcc.2004.11.008. [DOI] [PubMed] [Google Scholar]
- 35.Communal C, et al. Functional consequences of caspase activation in cardiac myocytes. Proc Natl Acad Sci USA. 2002;99:6252–6256. doi: 10.1073/pnas.092022999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Narula J, et al. Apoptosis and the systolic dysfunction in congestive heart failure: story of apoptosis interruptus and zombie myocytes. Cardiol Clin. 2001;19:113–126. doi: 10.1016/s0733-8651(05)70198-3. [DOI] [PubMed] [Google Scholar]
- 37.Chandrashekhar Y, et al. Long-term caspase inhibition ameliorates apoptosis, reduces myocardial troponin-I cleavage, protects left ventricular function, and attenuates remodeling in rats with myocardial infarction. J Am Coll Cardiol. 2004;43:295–301. doi: 10.1016/j.jacc.2003.09.026. [DOI] [PubMed] [Google Scholar]
- 38.Laugwitz KL, et al. Blocking caspase-activated apoptosis improves contractility in failing myocardium. Hum Gene Ther. 2001;12:2051–2063. doi: 10.1089/10430340152677403. [DOI] [PubMed] [Google Scholar]
- 39.Bers DM. Excitation-Contraction Coupling and Cardiac Contractile Force. Dordrecht: Kluwer Academic Publishers; 2001. [Google Scholar]
- 40.Ziolo MT, et al. Adenoviral gene transfer of mutant phospholamban rescues contractile dysfunction in failing rabbit myocytes with relatively preserved SERCA function. Circ Res. 2005;96:815–817. doi: 10.1161/01.RES.0000163981.97262.3b. [DOI] [PubMed] [Google Scholar]
- 41.Piacentino V, III, et al. Cellular basis of abnormal calcium transients of failing human ventricular myocytes. Circ Res. 2003;92:651–658. doi: 10.1161/01.RES.0000062469.83985.9B. [DOI] [PubMed] [Google Scholar]
- 42.Maier LS, et al. Dynamic changes in free Ca–calmodulin levels in adult cardiac myocytes. J Mol Cell Cardiol. 2006;41:451–458. doi: 10.1016/j.yjmcc.2006.04.020. [DOI] [PubMed] [Google Scholar]
- 43.Curran J, et al. β-Adrenergic enhancement of sarcoplasmic reticulum calcium leak in cardiac myocytes is mediated by calcium/calmodulin-dependent protein kinase. Circ Res. 2007;100:391–398. doi: 10.1161/01.RES.0000258172.74570.e6. [DOI] [PubMed] [Google Scholar]
- 44.Kohlhaas M, et al. Increased sarcoplasmic reticulum calcium leak but unaltered contractility by acute CaMKII overexpression in isolated rabbit cardiac myocytes. Circ Res. 2006;98:235–244. doi: 10.1161/01.RES.0000200739.90811.9f. [DOI] [PubMed] [Google Scholar]
- 45.Bassani JW, et al. Fractional SR Ca release is regulated by trigger Ca and SR Ca content in cardiac myocytes. Am J Physiol. 1995;268:C1313–C1319. doi: 10.1152/ajpcell.1995.268.5.C1313. [DOI] [PubMed] [Google Scholar]
- 46.Pogwizd SM, et al. Arrhythmogenesis and contractile dysfunction in heart failure: roles of sodium-calcium exchange, inward rectifier potassium current, and residual β-adrenergic responsiveness. Circ Res. 2001;88:1159–1167. doi: 10.1161/hh1101.091193. [DOI] [PubMed] [Google Scholar]
- 47.Venetucci LA, et al. Increasing ryanodine receptor open probability alone does not produce arrhythmogenic calcium waves: threshold sarcoplasmic reticulum calcium content is required. Circ Res. 2007;100:105–111. doi: 10.1161/01.RES.0000252828.17939.00. [DOI] [PubMed] [Google Scholar]
- 48.Wagner S, et al. Ca2+/calmodulin-dependent protein kinase II regulates cardiac Na+ channels. J Clin Invest. 2006;116:3127–3138. doi: 10.1172/JCI26620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Olshansky B, et al. Where patients with mild to moderate heart failure die: results from the Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT) Am Heart J. 2007;153:1089–1094. doi: 10.1016/j.ahj.2007.02.036. [DOI] [PubMed] [Google Scholar]
- 50.Srivastava S, et al. Downregulation of CuZn-superoxide dismutase contributes to β-adrenergic receptor-mediated oxidative stress in the heart. Cardiovasc Res. 2007;74:445–455. doi: 10.1016/j.cardiores.2007.02.016. [DOI] [PubMed] [Google Scholar]
- 51.Baumgarten G, et al. Load-dependent and -independent regulation of proinflammatory cytokine and cytokine receptor gene expression in the adult mammalian heart. Circulation. 2002;105:2192–2197. doi: 10.1161/01.cir.0000015608.37608.18. [DOI] [PubMed] [Google Scholar]
- 52.Niebauer J, et al. Endotoxin and immune activation in chronic heart failure: a prospective cohort study. Lancet. 1999;353:1838–1842. doi: 10.1016/S0140-6736(98)09286-1. [DOI] [PubMed] [Google Scholar]
- 53.Suzuki H, et al. Time-course of changes in the levels of interleukin 6 in acutely decompensated heart failure. Int J Cardiol. 2005;100:415–420. doi: 10.1016/j.ijcard.2004.08.041. [DOI] [PubMed] [Google Scholar]
- 54.Csont T, et al. The involvement of superoxide and iNOS-derived NO in cardiac dysfunction induced by pro-inflammatory cytokines. J Mol Cell Cardiol. 2005;39:833–840. doi: 10.1016/j.yjmcc.2005.07.010. [DOI] [PubMed] [Google Scholar]
- 55.Persad S, et al. Modification of cardiac β-adrenoceptor mechanisms by H2O2. Am J Physiol. 1998;274:H416–H423. doi: 10.1152/ajpheart.1998.274.2.H416. [DOI] [PubMed] [Google Scholar]
- 56.Zeitz O, et al. Hydroxyl radical-induced acute diastolic dysfunction is due to calcium overload via reverse-mode Na(+)-Ca(2+) exchange. Circ Res. 2002;90:988–995. doi: 10.1161/01.res.0000018625.25212.1e. [DOI] [PubMed] [Google Scholar]
- 57.Xu KY, et al. Hydroxyl radical inhibits sarcoplasmic reticulum Ca(2+)-ATPase function by direct attack on the ATP binding site. Circ Res. 1997;80:76–81. doi: 10.1161/01.res.80.1.76. [DOI] [PubMed] [Google Scholar]
- 58.Goldhaber JI. Free radicals enhance Na+/Ca2+ exchange in ventricular myocytes. Am J Physiol. 1996;271:H823–H833. doi: 10.1152/ajpheart.1996.271.3.H823. [DOI] [PubMed] [Google Scholar]
- 59.Zima AV, et al. Effects of cytosolic NADH/NAD(+) levels on sarcoplasmic reticulum Ca(2+) release in permeabilized rat ventricular myocytes. J Physiol. 2004;555:727–741. doi: 10.1113/jphysiol.2003.055848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Song Y, et al. Blocking late sodium current reduces hydrogen peroxide-induced arrhythmogenic activity and contractile dysfunction. J Pharmacol Exp Ther. 2006;318:214–222. doi: 10.1124/jpet.106.101832. [DOI] [PubMed] [Google Scholar]
- 61.Ziolo MT, Bers DM. The real estate of NOS signaling: location, location, location. Circ Res. 2003;92:1279–1281. doi: 10.1161/01.RES.0000080783.34092.AF. [DOI] [PubMed] [Google Scholar]
- 62.Ziolo MT, et al. Myocytes isolated from rejecting transplanted rat hearts exhibit a nitric oxide-mediated reduction in the calcium current. J Mol Cell Cardiol. 2001;33:1691–1699. doi: 10.1006/jmcc.2001.1420. [DOI] [PubMed] [Google Scholar]
- 63.Hu A, et al. Chronic β-adrenergic receptor stimulation induces cardiac apoptosis and aggravates myocardial ischemia/reperfusion injury by provoking inducible nitric-oxide synthase-mediated nitrative stress. J Pharmacol Exp Ther. 2006;318:469–475. doi: 10.1124/jpet.106.102160. [DOI] [PubMed] [Google Scholar]
- 64.Oddis CV, et al. cAMP enhances inducible nitric oxide synthase mRNA stability in cardiac myocytes. Am J Physiol. 1995;269:H2044–H2050. doi: 10.1152/ajpheart.1995.269.6.H2044. [DOI] [PubMed] [Google Scholar]
- 65.Ziolo MT, et al. Expression of inducible nitric oxide synthase depresses β-adrenergic-stimulated calcium release from the sarcoplasmic reticulum in intact ventricular myocytes. Circulation. 2001;104:2961–2966. doi: 10.1161/hc4901.100379. [DOI] [PubMed] [Google Scholar]
- 66.Ziolo MT, et al. Myocyte nitric oxide synthase 2 contributes to blunted β-adrenergic response in failing human hearts by decreasing Ca2+ transients. Circulation. 2004;109:1886–1891. doi: 10.1161/01.CIR.0000124231.98250.A8. [DOI] [PubMed] [Google Scholar]
- 67.Kohr MJ, et al. Targeting of phospholamban by peroxynitrite decreases β-adrenergic stimulation in cardiomyocytes. Cardiovasc Res. doi: 10.1093/cvr/cvm018. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Adam L, et al. Nitric oxide modulates β(2)-adrenergic receptor palmitoylation and signaling. J Biol Chem. 1999;274:26337–26343. doi: 10.1074/jbc.274.37.26337. [DOI] [PubMed] [Google Scholar]
- 69.Saraiva RM, et al. Deficiency of neuronal nitric oxide synthase increases mortality and cardiac remodeling after myocardial infarction: role of nitroso-redox equilibrium. Circulation. 2005;112:3415–3422. doi: 10.1161/CIRCULATIONAHA.105.557892. [DOI] [PubMed] [Google Scholar]
- 70.Buys ES, et al. Cardiomyocyte-restricted restoration of nitric oxide synthase 3 attenuates left ventricular remodeling after chronic pressure overload. Am J Physiol Heart Circ Physiol. 2007;293:H620–H627. doi: 10.1152/ajpheart.01236.2006. [DOI] [PubMed] [Google Scholar]
- 71.Damy T, et al. Increased neuronal nitric oxide synthase-derived NO production in the failing human heart. Lancet. 2004;363:1365–1367. doi: 10.1016/S0140-6736(04)16048-0. [DOI] [PubMed] [Google Scholar]
- 72.Bendall JK, et al. Role of myocardial neuronal nitric oxide synthase-derived nitric oxide in β-adrenergic hyporesponsiveness after myocardial infarction-induced heart failure in rat. Circulation. 2004;110:2368–2375. doi: 10.1161/01.CIR.0000145160.04084.AC. [DOI] [PubMed] [Google Scholar]
- 73.Whalen EJ, et al. Regulation of β-adrenergic receptor signaling by S-nitrosylation of G-protein-coupled receptor kinase 2. Cell. 2007;129:511–522. doi: 10.1016/j.cell.2007.02.046. [DOI] [PubMed] [Google Scholar]
- 74.Hare JM, Stamler JS. NO/redox disequilibrium in the failing heart and cardiovascular system. J Clin Invest. 2005;115:509–517. doi: 10.1172/JCI200524459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kubalova Z, et al. Abnormal intrastore calcium signaling in chronic heart failure. Proc Natl Acad Sci USA. 2005;102:14104–14109. doi: 10.1073/pnas.0504298102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Nishijima Y, et al. Chronic cardiac resynchronization therapy and reverse ventricular remodeling in a model of nonischemic cardiomyopathy. Life Sci. 2007;81:1152–1159. doi: 10.1016/j.lfs.2007.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]


