Article

Cardiac Resynchronisation Therapy and Cellular Bioenergetics: Effects Beyond Chamber Mechanics

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Information image
Average (ratings)
No ratings
Your rating

Abstract

Cardiac resynchronisation therapy is a cornerstone in the treatment of advanced dyssynchronous heart failure. However, despite its widespread clinical application, precise mechanisms through which it exerts its beneficial effects remain elusive. Several studies have pointed to a metabolic component suggesting that, both in concert with alterations in chamber mechanics and independently of them, resynchronisation reverses detrimental changes to cellular metabolism, increasing energy efficiency and metabolic reserve. These actions could partially account for the existence of responders that improve functionally but not echocardiographically. This article will attempt to summarise key components of cardiomyocyte metabolism in health and heart failure, with a focus on the dyssynchronous variant. Both chamber mechanics-related and -unrelated pathways of resynchronisation effects on bioenergetics – stemming from the ultramicroscopic level – and a possible common underlying mechanism relating mechanosensing to metabolism through the cytoskeleton will be presented. Improved insights regarding the cellular and molecular effects of resynchronisation on bioenergetics will promote our understanding of non-response, optimal device programming and lead to better patient care.

Disclosure:The authors have no conflicts of interest to declare.

Received:

Accepted:

Published online:

Correspondence Details:Polychronis Dilaveris, National and Kapodistrian University of Athens, 22 Miltiadou St, 155 61 Athens, Greece. E: hrodil1@yahoo.com

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

Cardiac resynchronisation therapy (CRT) has been a cornerstone in the treatment of select advanced heart failure cases since its introduction to our armamentarium in the early 2000s.1 Indeed, 30–60% of advanced heart failure patients exhibit evidence of dyssynchrony, when defined electrocardiographically or mechanically.2–4 The latter is a consequence of the former. CRT has several unique features, summarised in Table 1.

Although the main mechanisms through which CRT is thought to act are improvements in chamber mechanics, a more subtle effect has been recognised, linking resynchronisation to cellular metabolism and energy efficiency.

Full comprehension of this connection could help us to interpret non-response and lead to more sophisticated criteria for CRT use, help detect concealed responders – patients who do not have increases in mechanical output but do have improvements in bioenergetics and may have improved functional reserve – and alter CRT programming approach.

This article will present cellular bioenergetics in myocardial cells and subsequent alterations in heart failure. Cellular and molecular aspects of CRT effects on bioenergetics will be discussed, along with their potential implications for clinical practice.

Cardiomyocyte Metabolism in Health and Heart Failure

Normal Conditions

The human heart has been called a metabolic omnivore because of its ability to use all common oxidative substrates. Under normal conditions, 60–90% of adenosine triphosphate (ATP) produced is a product of fatty acid oxidation with the rest attributed to glucose oxidation.5 Notably, these metabolic pathways are mutually inhibitory (the Randle cycle) and the relative flow is determined by the fed or fasted state of the cell; fed leading to preferential use of glucose and fasted to fatty acid oxidation.6 Given the much higher efficiency of aerobic oxidation compared with anaerobic metabolism, the heart predictably uses the former (it accounts for 90% of energy production).7,8 Oxidative metabolism begins in the sarcoplasm but continues in the mitochondria, which occupy 30% of cardiomyocyte volume, strategically placed near myofibrils and sarcoplasmic reticulum (SR) to minimise the diffusion distance of the high-energy bond-containing molecules from production to consumption sites.8 This highlights the crucial dependency of cellular function on mitochondrial integrity, placement and output.

The burden of cardiac work is such that there is no energy production reserve. At maximal exercise the heart is operating at >90% of maximum oxidative capacity and recycles an amount of ATP (6 kg) more than 20 times its weight.6,9

Unique Features In Development

Article image

Complexes Of The Mitochondrial Respiratory

Article image

As such, establishing effective coupling between energy producing and consuming organelles is crucial. Calcium ions are a strong candidate for this role inasmuch as they participate in both actin-myosin crosstalk regulation and in activation of major mitochondrial complexes (Table 2) and ATP synthase.10–12

One of the key aspects of cellular bioenergetics lies in the recognition that the sarcoplasm is not a homogeneous solution of molecules; rather, local concentrations of metabolites are essential in determining metabolic efficiency, as dictated by the laws of thermodynamics.7 More specifically, the energy yield from ATP dissociation is determined by the equation:

Where ΔG denotes change in Gibbs free energy, a measure of the energy yield and so of the spontaneous nature of a reaction, combining both enthalpy (thermal output) and entropy (measure of randomness) through the equation:

With H denoting enthalpy, T absolute temperature and S entropy. ΔG0 is the standard free energy change at a temperature of 25°C (298 K), R is the universal gas constant in J mol-1 K-1, ln(X) is the natural logarithm of X and [Y] denotes the local concentration – allowed to freely diffuse in the vicinity without barriers such as membranes – of substance Y.

Consequently, the free energy yield (ΔG) is critically dependent on local concentrations of products and reactants and fast regeneration of ATP, along with rapid removal of adenosine diphosphate (ADP) from the vicinity to ensure optimal energy output. Simply put, the same intracellular task may require more ATP as a result of lower energy from every ATP dissociation owing to the mismanagement of reactants. Reduced yield may cause reactivation of the foetal myosin isoform (beta-myosin), producing less shortening per power stroke, but requiring less energy to undergo conformational changes than the adult one (alpha-myosin).13

In addition to the electron transport chain/ATP synthase proximity in mitochondria, all the enzymes of glycolysis are organised in complexes attached to energy-consuming structures (SR/myofibrils). Phosphocreatine (PCr) kinase also exhibits high concentrations near myosin head regions, SR calcium transport ATPase (SERCA) pumps and ADP/ATP mitochondrial antiporter, where it ensures rapid ATP regeneration and – in the latter case – rapid ATP shuttling to the sarcoplasm, maintaining optimal yield from ATP dissociation.14–17

Prerequisites for optimal and efficient cellular bioenergetics are listed in Table 3.

Alterations in Heart Failure

In heart failure, the heart switches from an omnivore to preferentially using glucose oxidation for energy production, initially by increasing glucose consumption and ultimately by decreasing fatty acid oxidation.18–20 In the short term, glucose and fatty acids compete with each other for use as substrates for energy production (the Randle cycle).21 In the fasted state, beta-oxidation of the relatively abundant fatty acids – released by the liver – causes an increase in the mitochondrial ratios of acetyl-coenzyme A/coenzyme A and reduced nicotinamide adenine dinucleotide (NADH)/oxidised nicotinamide adenine dinucleotide (NAD+), which both inhibit pyruvate dehydrogenase activity.22 Conversely, in the fed state, malonyl-coenzyme A, a by-product of glucose oxidation, inhibits fatty acid oxidation and promotes lipogenesis by inhibiting carnitine palmitoyltransferase.23 It appears that in heart failure, stressors – in the form of oxidative stress or adrenergic signalling – continuously promote glucose use as a substrate for energy production, because of its effects beyond ATP production.24

In the long term, changes in enzyme levels or activity are involved in regulating substrate use. Indeed, enzymes involved in fatty acid oxidation are downregulated in heart failure; a return to the foetal pattern.25,26 More specifically, entry of glucose catabolism products into the Krebs cycle is facilitated through a reduction of pyruvate dehydrogenase kinase – which phosphorylates and inhibits pyruvate dehydrogenase, necessary for the conversion of pyruvate to acetyl-coenzyme A – while transcript levels of carnitine palmitoyltransferase – necessary for acylcarnitine reconversion into acyl-coenzyme A and carnitine after entry into mitochondria – are downregulated.25,27 Furthermore, beta-oxidation is itself inhibited by reduced levels of some acyl-coenzyme A dehydrogenases, both in foetal and failing hearts.

Citrate synthase messenger RNA levels are also found to be reduced in the foetal gene-expression pattern, leading not merely to a metabolism based on glucose, but more specifically a glycolytic one. Increased glucose availability – at least prior to development of insulin resistance – may underlie the myosin isoform switch, through O-glycosylation of transcriptional factors.28 Furthermore, the foetal pattern promotes cell survival by means of innate antiapoptotic pathway activation, such as that mediated by the protein kinase B/mammalian target of rapamycin.29 The exact triggers for this switch are not well understood, but it is thought that exposure to a hypoxic milieu – caused, in heart failure, by vascular disease, fibrosis and increased workload – is reminiscent of the in utero environment and is the underlying cause of this change, leading to faster adaptation to changing stimuli mediated by a multitude of mechanisms acting on both the transcriptional and epigenetic level.28,30

The main activator of fatty acid oxidation is peroxisome proliferator-activated receptor-alpha (PPAR-alpha), which increases expression of genes involved in the entry, transport to mitochondria and beta-oxidation of fatty acids and downregulates expression of genes of proteins participating in glucose metabolism. Reduced PPAR-alpha expression has been described in heart failure and has been linked to fibrosis and mitochondrial fragmentation.31 This phenomenon may represent an adaptational attempt given that, stoichiometrically, the amount of oxygen consumed per ATP produced is higher for fatty acids than for glucose.32 Potential causes of lipotoxicity are detailed in Table 4.

In fact, increased glycolytic capacity has been related to increased survival in heart failure.33 Furthermore, localisation of glycolytic enzymes in the sarcoplasm and a preferentially glycolytic metabolism allows for ATP use in housekeeping processes – such as sodium–potassium ATPases and SERCAs – and provides substrates for the pentose phosphate pathway. The pentose phosphate pathway is critical for synthesis of antioxidants such as NADPH, allowing cellular survival – maintaining proper ionic concentrations and membrane potential – albeit at the cost of functionality.24,34 This is advantageous in the short term but detrimental in chronic conditions. Given the much higher amount of ATP produced per fatty acid molecule, many glucose molecules must be oxidated to match this yield. However, in advanced heart failure a reduced glucose oxidation capacity is found, primarily attributed to the insulin resistance of the failing heart because of an increased concentration of non-metabolised fatty acids and sympathetic system and renin-angiotensin-aldosterone axis activation.25,35–37 Downregulation of sarcoplasmic membrane glucose transporters has also been noted.38 This is especially true for septal segments in dyssynchronous heart failure where decreased workload – contraction against relaxed lateral segments – allows for reduced glucose consumption.39,40 Moreover, beta-oxidation leads to the production of Krebs cycle intermediates (anaplerotic reactions).33 Obviously this deprives the heart of its fuel, leading to energy starvation.9

Features Necessary For Normal Effectiveness

Article image

Proposed Mechanisms Of Lipotoxicity

Article image

As such, myocardial substrate metabolism tuning crucially affects left ventricular energetics in vivo, to the extent that preservation of fatty acid metabolism preference has been found to be linked with lack of clinical response to CRT.41,42 A twofold explanation may be given; either the failing myocardium has not switched to glucose metabolism as a compensatory mechanism and so continues to use an ineffective fuel, or full metabolic compensation at the microscopic level has been achieved so normal preference to fatty acids is maintained and no potential for further improvement exists.43 The latter interpretation is more likely given that CRT has been found to increase fatty acid metabolism, in itself facilitating oxidative metabolism.

PCr kinase levels are decreased in heart failure.44,45 This leads to a lower ATP/ADP ratio which – aside from the obvious negative effects discussed previously – may serve an unexpected adaptive purpose. That is, ensuring – by the opening of ATP-gated potassium channels of the mitochondrial membrane, which hyperpolarise the organelle – that cytochrome c will not be released and apoptotic processes will not be triggered.46 Furthermore, the mitochondrial isoform of PCr kinase partakes in a feedback mechanism ensuring that energy production is matched to consumption; ADP from the sarcoplasm is pumped into the mitochondria by the adenosine nucleotide transporter, converted to ATP by the mitochondrial PCr kinase and shuttled back through the same path. Loss of PCr means that the mitochondria can no longer respond accordingly to increased sarcoplasmic ADP.47

A complementary ATP-replenishing system involves adenylate kinase, which converts two ADP molecules into ATP and adenosine monophosphate (AMP). In addition, increased AMP levels lead to AMP-dependent protein kinase (AMPK) activation, which constitutes a central element of adaptational mechanisms in low-fuel conditions.48 Specifically, AMPK, when acutely activated, shuts down ATP-consuming processes such as fatty acid and glucose synthesis, activates ATP-producing pathways such as fatty acid oxidation and glucose intake through glucose transporter type 4, and increases insulin sensitivity, ensuring cellular survival.17,48–50 However, chronic activation is detrimental because fatty acid metabolism is reduced through mitochondrial transport inhibition and apoptosis is activated.51

Heart failure is a state of severe energy wasting, i.e. energy consumption not leading to useful work but lost as heat.52 Increased uncoupling protein concentration has been observed, leading to proton leak back into the matrix.9,33 Altered calcium homeostasis may lead to the myosin head performing the power stroke while unbound to actin (troponin C dysfunction), so not leading to sarcomere shortening.53,54 Finally, abnormalities in proteins connecting sarcomeres to the extracellular matrix lead to decoupling and prevent sarcomere shortening translating into cardiomyocyte contraction.55

Mitochondrial dysfunction in heart failure merits further consideration. Increased reactive oxygen species (ROS) concentrations in heart failure lead to electron transport chain dysfunction and damage mitochondrial proteins and genome.34,46 However, a burst of ROS may actually prove beneficial inasmuch as it triggers activation of the master regulator of mitochondrial biogenesis and oxidative capacity; peroxisome proliferator-activated receptor-gamma coactivator-1 (PGC-1). This protein allows for increased expression of nucleus-encoded mitochondrial transcription factor 1, increasing mitochondrial biogenesis, oxidative capacity and fatty acid use.8,56 Furthermore, PGC-1 increases synthesis of antioxidant enzymes (catalase, superoxide dismutase and glutathione peroxidase) in response to redox signalling.57

Chronically elevated catecholamine and angiotensin levels and increased TNF-alpha and endothelin-1 lead to protein kinase B activation, which, in turn, downregulates PGC-1, causing mitochondrial fragmentation (inability to replicate) and decreased concentrations of electron transport chain complexes and ATP synthase.5,7,9,10,33 Although CRT does increase protein kinase B phosphorylation/activity, this should be viewed in light of global prosurvival effects of this kinase, rather than its adverse actions on cellular bioenergetics, which are mitigated through different pathways by resynchronisation.58,59 Finally, and especially pertaining to dyssynchronous heart failure, with abnormal stretching of non-activated cells, an intimate crosstalk exists between cardiomyocyte stretch and apoptosis. This appears to be mediated by mitochondria, as evidenced by proapoptotic molecule (p53 and Bax) increases, leading to mitochondrial membrane depolarisation and release of cytochrome c and other complexes triggering apoptosis upon cellular stretching.60

In short, teleologically, heart failure initially redirects cardiomyocyte metabolism towards glucose use, which ensures cellular survival at the cost of function. However, insulin resistance in advanced stages of heart failure prevents use of glucose as efficiently.61 Studies have suggested that the failing heart may attempt to switch to ketone bodies oxidation in an effort to sustain its metabolism and increase its efficiency (conversion into usable substrate not requiring much energy).27,62,63 Ketone bodies oxidation is a rapidly usable and relatively dense – requiring two steps to enter the Krebs cycle as acetyl-coenzyme A and each yielding two acetyl-coenzyme A molecules – source of energy, supplemented by the liver.

Branched chain amino acid catabolic pathways have been found to be downregulated in a murine heart failure model – with reduced expression of key enzymes such as the branched-chain alpha-keto acid dehydrogenase complex – presumably because of oversupply of ketone bodies by the liver (two out of three proteinogenic branched chain amino acids are ketogenic).64 However, this leads to an accumulation of branched-chain keto acids, which induces inhibition of mitochondrial respiration.65 Moreover, factors promoting fatty acid oxidation (PPAR-alpha and PGC-1) are also regulators of cellular oxidative capacity in general.34 As such, their downregulation leads not only to preferential glucose use but also to reduced oxidative potential of the cell. Several of the previously mentioned disturbances are (partially) reversed by effective resynchronisation, as discussed in the following sections.

Chamber Mechanics and Energy Consumption in Dyssynchronous Heart Failure

It should be noted that most studies attempting to clarify effects of resynchronisation on chamber mechanics, substrate metabolism and energy efficiency have been conducted in dilated cardiomyopathy models with concomitant left bundle branch block because of the perceived homogeneous myocardial involvement that leads to more predictable behaviour, i.e. excitation sequence, after CRT initiation. Conversely, ischaemic cardiomyopathy of such severity (ejection fraction ≤35%) usually involves the presence of extensive dense scar regions, which critically modify electrophysiological properties (the stimulus may need to bypass a fixed line of block and, in the process, depolarise different myocardial segments). This is reflected by ischaemic aetiology being considered a negative prognosticator for response to CRT.66–74 The patients expected to gain the greatest benefits from chamber resynchronisation are those with the most pronounced basal dyssynchrony, exhibiting the widest QRS complexes.75–77

Modern views on dyssynchronous heart failure include the notion of adverse mechanoenergetics, leading to reduced energy efficiency, evidenced by a reduced ratio between energy consumed (in the form of oxygen) and external work performed (stroke work).78–80 Several explanations can be offered (Table 5).

Compared with inotropes, CRT has been found to acutely improve systolic function (increased) while reducing oxygen consumption assessed by arterial/coronary sinus oxygen difference and minute oxygen consumption).81 Chamber segments resynchronisation offers a plausible interpretation – by negating increased interior work – for improved energetics upon CRT introduction. An additional beneficial effect of CRT could lie in the reversal of the shortening of diastolic period and diastolic dysfunction, both present in dyssynchronous heart failure in general and particularly so in the left bundle branch block variety.82–84 More specifically, divergence in the timing of systole between myocardial segments in left bundle branch block dilated cardiomyopathy has been shown to worsen microvascular function of the left anterior descending artery territory (perfusion occurring during diastole) and consequently flow reserve, raising the potential for therapeutic interventions.85 In the mechanoenergetics context, although not improving efficiency, CRT could improve overall available energy by increasing oxygen and substrate delivery to myocardium.

The most robust method of assessing substrate metabolism relies on the use of glucose and fatty acid radioactive isotopes and their in vivo turnover rates.43,86–89 Substrate metabolism, but not perfusion, is also altered in dyssynchronous heart failure. The septal wall uses less glucose than lateral segments, potentially owing to the preferential conversion of mechanical work to stretching of inactive myocardium, as opposed to blood expulsion through the aortic valve, mediated molecularly by reduced glucose transporter expression.38,90,91 Conversely, upon contraction, the lateral wall consumes higher amounts of glucose-derived energy because of its pre-stretched condition (exceeding Frank-Starling law limits). Furthermore, this is in accordance with preferential glucose use in advanced heart failure, as the lateral wall could be considered to be at a more locally advanced heart failure stage.

Consequently, glucose uptake by 13-fluorodeoxyglucose – measured by PET – may represent increases in glycolysis rather than oxidative phosphorylation and prioritisation of survival over function. As such, this pendulum-like internal energy transfer accounts for significant macroscopic causes of reduced mechanical efficiency. When oxidative metabolism is specifically assessed by means of acetate clearance – a precursor to acetyl-coenzyme A that enters the Krebs cycle – an increase in septal and a decrease in lateral oxidative metabolism upon CRT was noted, leading to global homogenisation without increases in metabolic demand, despite increases in mechanical work output.92 This appears to contradict previous interpretations of CRT effects on substrate metabolism, yet it can be hypothesised that the increased septal work reactivated oxidative phosphorylation to increase energy output, whereas the lateral wall operates at less-than-maximal oxidative capacity and can reduce oxidative metabolism, also reducing ROS burden in the process.

Predictably, CRT has been found to rectify and homogenise glucose use throughout the myocardium without affecting perfusion to the same degree; septal:lateral glucose use ratio increased from 0.62 to 0.91 after CRT, p<0.001.39,93,94 Regarding the septum in particular, a slower – anticipated by the fact the lateral segments are also simultaneously actively contracting – yet more effective contraction has been reported.95 The importance of myocyte length has been further demonstrated by the dependence of generated pressure and oxygen consumption (both increasing, the former more than the latter, leading to improved efficiency) on atrioventricular delay during CRT, with extremely short delays completely negating haemodynamic effects of resynchronisation.96 Moreover, CRT appeared to confer a benefit with regard to the metabolic reserve and mechanical efficiency (response to beta-stimulation) of the failing heart, even after long-term application.72 The same increases in output were noted upon dobutamine administration compared with dyssynchronous hearts, yet were accompanied by increased efficiency and acetate extraction (operation at less-than-maximal oxidative capacity at rest).93 This long-term trend – present even 13 months after CRT initiation – alludes to intrinsic metabolism alterations after prolonged CRT application, consistent with the notion of detrimental effects of dyssynchrony itself on energy efficiency.72,97 Additionally, a trend for beneficial effects of CRT on fluid dynamics, leading to increased direct conversion of inflow to outflow kinetic energy, has been reported. This complements previous reports on the effects of dyssynchrony on rheodynamics and potentially offers a novel marker for predicting response to CRT.98–101

Possible Mechanisms Underlying

Article image

Although similar metabolism alterations have been reported in ischaemic cardiomyopathy and analogous CRT effects are indeed observed in the majority of dyssynchronous cases, a sizeable minority (32%) do not exhibit septal reverse mismatch (lower glucose use relative to perfusion).102 This is attributable to perfusion deficits of the lateral wall – present in 91% of the subgroup discussed – precluding higher glucose use there and, in effect, leaving the septum responsible for cardiac output.

An important inherent limitation of this approach for metabolism assessment lies in the assumption of intracellular homogeneity that ignores the high degree of compartmentalisation and sequestration in the sarcoplasm.9,103 That is, intracellular presence of a metabolite does not account for the site and mode of its breakdown, its vicinity to the energy-consuming areas of the cell and ultimately its energy yield, alterations of which may crucially affect efficiency.

A novel pacing modality, multisite pacing – constituting an advanced form of CRT – has started to yield interesting results regarding the previously mentioned parameters. It entails administration of two, rather than one, left ventricular pacing pulses, followed by a stimulus to the right ventricle. As such, it allows for sculpting of the activation sequence of the myocardium, potentially bypassing limitations posed by dense scar presence and allowing for further optimisation.104 Interesting results have been obtained regarding haemodynamics and stroke work improvement, and clinical studies attempting to correlate multisite pacing optimisation with energy efficiency improvement at the cardiovascular system level, through use of ventriculoarterial coupling, are underway.105,106

Dyssynchronous heart failure, specifically in the form of left bundle branch block presence, leads to mechanical dispersion and altered stretching of the different myocardial segments, affecting calcium ion homeostasis – potentially via mechanosensitive ion channels – leading to proarrhythmia, theoretically reversible by CRT.107,108 However, CRT can, itself, under certain conditions, exhibit proarrhythmic effects as a result of:109

  • increased transmural dispersion of repolarisation (pulse delivered at left ventricular epicardium);
  • wave break at the collision area of the two pulses (left and right ventricular) and potential re-entry; and
  • in-scar pacing, amenable to time-dependent alteration, potentially underlying electrical storm events.110,111

As as result of the first pulse being delivered with increased width, multisite pacing may not be involved in re-entry occurring because of localised in-scar pacing.104

This implies that CRT, besides its acute haemodynamic effects – stroke volume increases were noted in the previously mentioned studies – has profound effects on myocardial energy management.72,93 This raises the intriguing possibility that, depending on currently undetermined individual parameters, response to CRT may not manifest as an overt improvement in ejection fraction, chamber volumes and pressures, but in covert alterations in metabolism, increasing efficiency and reserve, rendering the heart more able to respond to abrupt increases in output demand.112 Such effects have been reported even when classical adverse prognosticators – QRS widening upon biventricular pacing – are observed, suggesting a degree of independence between effects on mechanics and bioenergetics, owing to differing underlying molecular pathways.113 The inhomogeneous myocardial involvement in ischaemic cardiomyopathy, leading to divergent and unpredictable effects of dyssynchrony on metabolism, may also affect these covert responders, at least at the macroscopic (chamber) level.102

CRT Effects on Bioenergetics at the Ultramicroscopic Level

Mitochondria are one of the key sites of CRT effects at the ultramicroscopic level. Proteomic analysis in a canine model has revealed levels of 31 mitochondrial proteins to be altered after CRT application, with almost half constituting subunits of the respiratory chain.114 A concerted effect is observed whereby CRT increases activity of key enzymes in anaplerotic pathways, providing more intermediates of the Krebs cycle, which can otherwise be consumed to biosynthetic processes such as amino acid synthesis, increasing its activity, facilitating acetyl-coenzyme A entry into the cycle and consequently NADH/flavin adenine dinucleotide (FADH2) formation and funnelling of electrons into their transport chain to complete aerobic oxidation.115

Moreover, all complexes of the respiratory (electron transport) chain, with the exception of cytochrome c oxidase (complex IV), have critical subunits upregulated. This includes complex II (succinate dehydrogenase), which links the Krebs cycle with electron transport chain; it catalyses succinate to fumarate conversion along with FADH2 formation, immediately introduces FADH2-derived electrons into the chain and allows for the flow of NADH-derived electrons that enter the chain further upstream – complex I – as a result of higher energy levels towards complex III/cytochrome bc1 complex.116 Finally, ATP synthase (complex V) subunit degradation is inhibited and, following phosphorylation, complex formation and ATP yield per proton flowing is improved (2.5 ADP molecules phosphorylated per oxygen atom consumed with CRT versus 1.4 in dyssynchronous heart failure – due to electron entry now more often occurring at complex I). Complex V activity increases 20% during CRT compared with dyssynchronous heart failure. As such, ultimately, electrons exiting the chain and protons flowing down their electrochemical gradient through complex V are paired with molecular oxygen, forming water.

Several complementary effects have been noted, such as upregulation of fatty acid binding protein that allows their entry into mitochondrial matrix and subsequent beta-oxidation and downregulation of uncoupling proteins.117 The latter may be a defence mechanism, reducing energy production and – through substrate alteration – reducing contraction of the myocyte to prevent the occurrence of extreme stretches, especially during contraction against already shortened segments (lateral wall). Cytochrome c has, unexpectedly, been reported to be downregulated following biventricular pacing, owing to either methodological issues (post-translational modifications that alter its pI and so its localisation following protein electrophoresis) or in the context of preventing apoptotic cell death, promoted by several pathways in dyssynchronous heart failure, as previously discussed.114

Although electron and proton coupling with molecular oxygen is tightly controlled by complex IV, there is always the possibility of ROS formation through release of intermediates from complex IV or electron interaction with molecules other than oxygen. These can induce post-translational modifications to a multitude of mitochondrial energy production-related proteins and membrane lipids, severely impairing their functionality and even leading to cell death.118–122 Most importantly, endothelial nitric oxide synthase, under oxidative stress, becomes uncoupled from tetrahydrobiopterin (itself oxidated) and yields significant amounts of ROS.121

To negate this potentially detrimental side-effect of increased function and efficiency of oxidative metabolism, an increase in ROS-scavenging protein levels is necessary and sufficient. Indeed, CRT has been found to be linked to a significant rise in thioredoxin-dependent peroxide reductase, critical for the formation of reduced disulphide bonds that either constitute the reversal of oxidative events or may be coupled – as in the case of glutathione – to the reduction of a variety of oxidised molecules.114,123 Interestingly, post-translational modification of regulatory ATP synthase subunits (alpha subunits) has been reported (S-glutathionylation and S-nitrosation), along with novel disulphide bond formation. S-glutathionylation reversal by CRT was linked to a twofold increase in enzyme activity, providing a further mechanism for its effects on complex V.124 Despite this, downregulation of ATP synthase activity may be part of a feedback loop, where increases in oxidative stress, such as those caused by heart failure, prevent aerobic oxidation and ultimately limit ROS production.125–130 The ensuing increase in oxidative stress is alleviated by the aforementioned increases in ROS-scavenging proteins.

These changes in protein levels are coupled with increased expression of proteins that translocate into mitochondria – synthesis of most energy production-related proteins has been relinquished to the nucleus – chief among which is prohibitin 2. Prohibitins are assembled into a ring-like structure in the inner mitochondrial membrane and their presumed roles involve being chaperones for respiration chain proteins (ensuring proper protein folding) or acting as general structuring scaffolds required for optimal mitochondrial morphology and function.131,132 Mitochondrial protein proteases have conversely been found to be downregulated post-CRT.133

Of note, according to transcriptomic analyses, gene level-related effects of dyssynchrony were sixfold more pronounced at the anterior than the lateral wall.134 Appropriately, CRT leads to pronounced local but not global changes in gene expression.26 Chronically increased stresses such as those observed upon dyssynchrony lead to profound alterations in key elements of the contractile apparatus, namely the transverse tubules (T-tubules), causing their disruption. This is mediated by downregulation of structural proteins such as junctophilin, and consequently impairs calcium handling (T-tubules bring into proximity the sarcolemmal and SR membranes).135,136 This indirectly affects energy use as much larger (potentially proarrhythmic) calcium spikes are necessary to cause calcium release from the SR and accomplish the excitation-contraction coupling; a disturbance that CRT can rectify.

A potential master regulator of protein activity affected by CRT has been recognised in the form of CK2 (formerly casein kinase II), a serine-threonine protein kinase, using both ATP and guanosine-5’-triphosphate as substrates and indispensable to cell survival and growth by preventing caspase access to cleavage sites of proteins, promoting DNA repair and suppressing p53 activity.137–139 In a canine model of tachypacing- and left bundle branch-related dyssynchronous heart failure, phosphoproteomics analysis identified CK2 as the most likely kinase whose downregulation is involved in protein phosphorylation alterations observed in dyssynchronous heart failure, a pattern reversed by the introduction of CRT. Although an intriguing finding, it cannot be inferred whether CRT effects are mediated by upregulation of CK2 or whether improved energetics and triphosphate nucleotide availability (used by CK2 as a phosphate donor during phosphorylation) account for increased kinase activity, promoting cell survival and function in a virtuous cycle. Finally, similar effects leading to increases in oxidative potential have been noted in peripheral muscles as well, potentially resulting in further improvement of patients’ functional status and exercise tolerance.140

Increased adrenergic signalling is usually thought of as deleterious in heart failure; evidenced by the benefits of its blockade with beta-blockers. Accordingly, there is intrinsically reduced responsiveness of the myocardium to adrenergic stimuli as a defence mechanism involving increased activation of inhibitory G-alpha subunits (deactivating rather than activating protein kinase A; PKA) and internalisation/degradation of receptors through phosphorylation by G-protein receptor kinase 2.141–144 However, the response of the myocardium to a transient increase in adrenergic signalling may be crucial in maintaining exercise capacity. CRT has been found to restore this parameter by upregulating regulator proteins of G-protein signalling (RGS) that inhibit inhibitory alpha-subunits of G-proteins (Gais).145 As such, SR-bound PKA may activate calcium-handling proteins, increasing calcium ion transient flux during depolarisation and its subsequent sequestration, ultimately enhancing the associated myocyte shortening, reversing the severely blunted calcium spikes of dyssynchronous heart failure.146–148 This post-translational effect on calcium-handling proteins is supported by the observation that their encoding genes are not upregulated by CRT.149 Consequently, exercise tolerance may be further improved.

Depiction Of Cardiomyocyte Metabolism

Article image

Alterations Induced Under Cardiac Resynchronisation Therapy

Article image

The most intriguing observation stemming from a canine model study by Chakir et al. lies in the triggering of increased response to beta-adrenergic signalling following dyssynchrony introduction into synchronous heart failure.145 This allowed for radical hypotheses claiming that a CRT holiday (analogous to diuretic holiday) may enhance CRT effects – particularly in non-responders – and that purposeful transient dyssynchrony introduction in heart failure patients without an indication for CRT or a pacemaker may trigger the same beneficial results. Animal studies in the tachypacing-induced cardiomyopathy setting have since confirmed this assumption, reporting improved cardiac response to beta-adrenergic stimuli and suppression of heart failure in terms of chamber dilatation and cellular dysfunction, even in the context of synchronous heart failure.150 An obvious noted issue stems from potential intersubject differences in the required holiday period.

A further path linking substrate preference to contractile function has been reported to be affected by CRT, namely phosphorylation of Z-disk and M-band proteins by the upregulated glycogen synthase kinase.151 Activation of this kinase leads to inactivation of glycogen synthase, potentially in the context of restoring the omnivorous nature of the cardiomyocyte. Target sarcomeric proteins (troponins I and T, myosin-binding protein C and myosin light chain isoforms) are involved in calcium sensing and consequently their activation sensitises the contractile apparatus to calcium ions’ presence and facilitates contraction.

Moreover, CRT has been shown to increase messenger RNA levels of the alpha-myosin isoform and its relative ratio to those of beta-myosin in advanced dyssynchronous heart failure patients, possibly because of improvements in energy metabolism that allow for a more ATP-consuming yet more efficient isoform to be chosen, restoring functionality.152 Interestingly, another subset of glycogen synthase kinase target sarcomeric proteins are involved in mechanosensing and may actually partake in the core mechanisms through which CRT exerts its effects. Figure 1 summarises the most important effects of CRT application to metabolism in dyssynchronous heart failure at the ultramicroscopic level.

Linking Mechanics to Metabolomics

A fundamental issue regarding all the described effects of CRT on cellular bioenergetics is raised concerning mechanisms and signal transduction pathways underlying the translation of mechanical alterations to metabolic effects. A strong candidate is the cytoskeleton, linking sarcomeres – the contractile units of cardiomyocytes – with the extracellular matrix and ensuring that cellular contraction causes tissue shortening. As a result of changes in myocardial lamellae orientation and chamber architecture, mechanical deformation of the failing heart is significantly altered.153,154 The aforementioned changes in calcium concentration, owing to alterations in proteins such as SERCA, decrease peak contractile force at the same time as modified extracellular matrix synthesis (different collagen isoforms) and sarcomere architecture (reduced levels of titin, a spring-like protein conferring elasticity) render myocardial tissue stiff.155,156

Concomitantly, changes in gap junction proteins – connexin-43 is redistributed laterally rather than longitudinally – reduce both conduction velocity and synchronisation between adjacent cardiomyocytes.157,158 Protein kinase B, which is upregulated by CRT, has been found to phosphorylate connexin-43 and facilitate incorporation into gap junctions, allowing for initiation and coordination of the ischaemic injury response.21,159 Consequently, mitogen-activated protein kinase further phosphorylates connexin-43 leading to gap junction closure and severing communication between adjacent cells’ sarcoplasms, potentially preventing apoptosis spreading.159 As such, stiff cardiomyocytes exhibiting reduced functionality attempt to contract over a stiffer matrix, not amenable to deformation, leading to abnormally high tension to the cytoskeletal structures connecting them (focal adhesions and integrins).160 This condition is further aggravated by local (between adjacent cells) and regional (in dyssynchronous heart failure) lack of coordination.156,161

Focal adhesions are so intricately linked with sarcomeres – to ensure anchoring to sarcolemma/extracellular matrix and bidirectional mechanotransduction – that a specialised structure emerges called a costamere.162–164 Costameres are sub-sarcolemmal multiprotein complexes initially found to localise over Z-disks, perpendicularly to the cardiomyocyte axis, but subsequently shown to extend over M-lines and along the myocyte length.163,165,166 They can be thought of as a specialised striated and cardiac muscle version of focal adhesions. Two main constituent protein complexes have been identified; the dystrophin/glycoprotein complex that provides a link between laminin – an extracellular matrix protein – and filamentous actin-based cytoskeleton, and the integrin/talin/vinculin complex, which is potentially more involved in signal transduction given its association with integrin-linked and focal adhesion kinases.167–170

The importance of these protein complexes in the biomechanical stability of the heart, especially under mechanical stress, can be inferred by the fact that mice with cardiac-specific vinculin deletion display a dilated cardiomyopathy phenotype.171 Moreover, another integrin-associated protein, melusin – a chaperone, assisting in proper protein folding and assembly – performs a highly specialised role in cardiomyocyte response to stress stimuli by promoting cellular survival and hypertrophy through both mitogen-activated protein kinase family pathways and protein kinase B (also affecting energy metabolism and, as mentioned previously, upregulated by CRT).58,59,172–174 Indeed, in aortic stenosis patients, melusin levels have been found to correlate with systolic function preservation.175 As such, it could be theorised that persistent supraphysiological stress (stretch) levels, such as those in dyssynchronous heart failure, render melusin levels insufficient to induce and maintain proper cellular responses, leading to apoptosis and myocardial wall thinning.

Similar findings have been reported in the case of integrins.176 Changes in integrin isoform gene expression profile have been noted during reverse remodelling of chambers upon left ventricular assist device therapy.176 CRT has been shown to lead to alterations in levels or activity of several sarcomeric and focal adhesions-related proteins involved in mechanosensing, such as cap-Z (reduced expression), muscle LIM protein (acetylation-activation), tensin, desmin and filamin-C.151,177–180 Spatial distribution of alpha-actinin – a microfilament protein necessary for the attachment of actin filaments to the Z-lines in skeletal muscle cells, coordinating sarcomeric contraction and providing links to focal adhesions and stress actin fibres – has been found to be disrupted in dyssynchronous heart failure, losing its periodicity and forming depositions, with CRT partially rectifying its pattern.181–183

Mitochondria, being linked to the cytoskeleton – both actin and microtubules – for reasons of localisation and transport, also sense the increased tension of dyssynchronous heart failure.55,184,185 This leads to increased expression of proapoptotic Bcl-2 family proteins, including Bax and Bad, which mediate cytochrome c release and apoptosis triggering, and to reduced complex IV activity (reduced by 21%) leading to reduced ATP formation and ROS accumulation because electrons are unable to combine with molecular oxygen and protons.60,186,187 In fact, the type of stretch caused by dyssynchrony (monotonous stretch) further accentuates these changes and directly reduces ATP synthase activity and mitochondrial biogenesis by reducing PGC-1alpha.188 In this framework, shutting down oxidative metabolism – with all its detrimental consequences – may again constitute a defensive mechanism of the cell to prevent apoptosis, in part caused by increased mechanical stretch. As such, not only is metabolism affected by anomalous stretch, but also its ability to adapt in response to such stressors is impaired.189

As such, an overarching hypothesis could be that alleviation by resynchronisation of the increased strain imposed on cardiomyocytes by dyssynchrony, which led to compensatory alterations/remodelling of sarcomeres and triggered changes in bioenergetics, now leads to altered input from specialised mechanosensors that is transduced to the cell, triggering the beneficial effects.53,190 A depiction of our current partial understanding of this link is attempted in Figure 2.

Conclusion

CRT can be thought of as constituting a metabolic therapy, acting on two levels. Firstly, at the chamber level, it rehomogenises substrate use and improving mechanical output and energy efficiency by reducing energy spent on internal work. Secondly, at the cellular level, it increases oxidative cell capacity by acting at virtually all stages of oxidative metabolism. The latter effect may not be inseparable from the former, creating the potential for the existence of covert responders.

Whether this is true for cases that do not have an indication for CRT or can be achieved through more advanced resynchronisation modalities, such as multisite pacing, or even by allowing (in dyssynchronous heart failure) or introducing (in synchronous heart failure) intermittent dyssynchrony, requires further study. It is possible that advanced computational models will be needed to determine the optimal site of leads and timing parameters to achieve conventional CRT/multisite pacing optimisation. Certain models can already integrate projected changes in bioenergetics, in the form of ATP use homogenisation assessed concomitantly with stroke work maximisation, to propose adequate lead implantation sites.156,191 Although challenging, especially to the clinician, comprehending these principles will probably prove necessary to promote our insight into the effects of resynchronisation beyond chamber mechanics, improving care for our patients.

Linking Mechanical Stretch

Article image

References

  1. Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J 2016;37:2129–200.
    Crossref | PubMed
  2. McAlister FA, Ezekowitz J, Hooton N, et al. Cardiac resynchronization therapy for patients with left ventricular systolic dysfunction: a systematic review. JAMA 2007;297:2502–14.
    Crossref | PubMed
  3. Chalil S, Stegemann B, Muhyaldeen S, et al. Intraventricular dyssynchrony predicts mortality and morbidity after cardiac resynchronization therapy: a study using cardiovascular magnetic resonance tissue synchronization imaging. J Am Coll Cardiol 2007;50:243–52.
    Crossref | PubMed
  4. Yu CM, Lin H, Zhang Q, Sanderson JE. High prevalence of left ventricular systolic and diastolic asynchrony in patients with congestive heart failure and normal QRS duration. Heart 2003;89:54–60.
    PubMed
  5. Huss JM, Kelly DP. Nuclear receptor signaling and cardiac energetics. Circ Res 2004;95:568–78.
    Crossref | PubMed
  6. Fillmore N, Mori J, Lopaschuk GD. Mitochondrial fatty acid oxidation alterations in heart failure, ischaemic heart disease and diabetic cardiomyopathy. Br J Pharmacol 2014;171:2080–90.
    Crossref | PubMed
  7. Alberts B, Johnson A, Lewis J, et al., Energy conversion: mitochondria and chloroplasts. In: Molecular Biology of the Cell. 5th ed, New York: Garland Science, 2008:813–78.
  8. Ventura-Clapier R, Garnier A, Veksler V. Energy metabolism in heart failure. J Physiol 2004;555:1–13.
    Crossref | PubMed
  9. Neubauer S. The failing heart – an engine out of fuel. N Engl J Med 2007;356:1140-51.
    Crossref | PubMed
  10. Stanley WC, Recchia FA, Lopaschuk GD. Myocardial substrate metabolism in the normal and failing heart. Physiol Rev 2005;85:1093–129.
    Crossref | PubMed
  11. Balaban RS. Cardiac energy metabolism homeostasis: role of cytosolic calcium. J Mol Cell Cardiol 2002;34:1259–71.
    Crossref | PubMed
  12. Nickel A, Loffler J, Maack C. Myocardial energetics in heart failure. Basic Res Cardiol 2013;108:358.
    Crossref | PubMed
  13. Boulpaep EL. The heart as a pump. In: Boron WF, Boulpaep EL, eds. Medical Physiology: A cellular and Molecular Approach, Philadelphia: Saunders, 2003:508–33.
  14. Wallimann T, Eppenberger HM. Localization and function of M-line-bound creatine kinase. M-band model and creatine phosphate shuttle. Cell Muscle Motil 1985;6:239–85.
    PubMed
  15. Ventura-Clapier R, Veksler V, Hoerter JA. Myofibrillar creatine kinase and cardiac contraction. Mol Cell Biochem 1994;133–134:125–44.
    PubMed
  16. Ventura-Clapier R, Kuznetsov A, Veksler V, et al. Functional coupling of creatine kinases in muscles: species and tissue specificity. Mol Cell Biochem 1998;184:231–47.
    PubMed
  17. Saks VA, Khuchua ZA, Vasilyeva EV, et al. Metabolic compartmentation and substrate channelling in muscle cells. Role of coupled creatine kinases in in vivo regulation of cellular respiration – a synthesis. Mol Cell Biochem 1994;133–134:155–92.
    PubMed
  18. Remondino A, Rosenblatt-Velin N, Montessuit C, et al. Altered expression of proteins of metabolic regulation during remodeling of the left ventricle after myocardial infarction. J Mol Cell Cardiol 2000;32:2025–34.
    Crossref | PubMed
  19. Nascimben L, Ingwall JS, Lorell BH, et al. Mechanisms for increased glycolysis in the hypertrophied rat heart. Hypertension 2004;44:662–7.
    Crossref | PubMed
  20. Osorio JC, Stanley WC, Linke A, et al. Impaired myocardial fatty acid oxidation and reduced protein expression of retinoid X receptor-alpha in pacing-induced heart failure. Circulation 2002;106:606–12.
    PubMed
  21. Dunn CA, Lampe PD. Injury-triggered Akt phosphorylation of Cx43: a ZO-1-driven molecular switch that regulates gap junction size. J Cell Sci 2014;127:455–64.
    Crossref | PubMed
  22. Bowker-Kinley MM, Davis WI, Wu P, et al. Evidence for existence of tissue-specific regulation of the mammalian pyruvate dehydrogenase complex. Biochem J 1998;329:191–6.
    Crossref | PubMed
  23. Hue L, Taegtmeyer H. The Randle cycle revisited: a new head for an old hat. Am J Physiol Endocrinol Metab 2009;297:E578–91.
    Crossref | PubMed
  24. Doenst T, Nguyen TD, Abel ED. Cardiac metabolism in heart failure: implications beyond ATP production. Circ Res 2013;113:709–24.
    Crossref | PubMed
  25. Razeghi P, Young ME, Alcorn JL, et al. Metabolic gene expression in fetal and failing human heart. Circulation 2001;104:2923–31.
    PubMed
  26. Barth AS, Chakir K, Kass DA, et al. Transcriptome, proteome, and metabolome in dyssynchronous heart failure and CRT. J Cardiovasc Transl Res 2012;5:180–7.
    Crossref | PubMed
  27. Chen L, Song J, Hu S. Metabolic remodeling of substrate utilization during heart failure progression. Heart Fail Rev 2019;24:143–54.
    Crossref | PubMed
  28. Taegtmeyer H, Sen S, Vela D. Return to the fetal gene program: a suggested metabolic link to gene expression in the heart. Ann N Y Acad Sci 2010;1188:191–8.
    Crossref | PubMed
  29. Sack MN, Yellon DM. Insulin therapy as an adjunct to reperfusion after acute coronary ischemia: a proposed direct myocardial cell survival effect independent of metabolic modulation. J Am Coll Cardiol 2003;41:1404–7.
    Crossref | PubMed
  30. Dirkx E, da Costa Martins PA, De Windt LJ. Regulation of fetal gene expression in heart failure. Biochim Biophys Acta 2013;1832:2414–24.
    PubMed
  31. Garnier A, Fortin D, Deloménie C, et al. Depressed mitochondrial transcription factors and oxidative capacity in rat failing cardiac and skeletal muscles. J Physiol 2003;551:491–501.
    Crossref | PubMed
  32. Jaswal J, Ussher J. Myocardial fatty acid utilization as a determinant of cardiac efficiency and function. Clin Lipidol 2009;4:379–89.
    Crossref
  33. Ingwall JS. Energy metabolism in heart failure and remodelling. Cardiovasc Res 2009;81:412–9.
    Crossref | PubMed
  34. van Bilsen M, van Nieuwenhoven FA, van der Vusse GJ. Metabolic remodelling of the failing heart: beneficial or detrimental? Cardiovasc Res 2009;81:420–8.
    Crossref | PubMed
  35. An D, Rodrigues B. Role of changes in cardiac metabolism in development of diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol 2006;291:H1489–506.
    Crossref | PubMed
  36. Kalsi KK, Smolenski RT, Pritchard RD, et al. Energetics and function of the failing human heart with dilated or hypertrophic cardiomyopathy. Eur J Clin Invest 1999;29:469–77.
    Crossref | PubMed
  37. Taylor M, Wallhaus TR, Degrado TR, et al. An evaluation of myocardial fatty acid and glucose uptake using PET with [18F]fluoro-6-thia-heptadecanoic acid and [18F]FDG in patients with congestive heart failure. J Nucl Med 2001;42:55–62.
    PubMed
  38. Depre C, Shipley GL, Chen W, et al. Unloaded heart in vivo replicates fetal gene expression of cardiac hypertrophy. Nat Med 1998;4:1269–75.
    Crossref | PubMed
  39. Nowak B, Sinha AM, Schaefer WM, et al. Cardiac resynchronization therapy homogenizes myocardial glucose metabolism and perfusion in dilated cardiomyopathy and left bundle branch block. J Am Coll Cardiol 2003;41:1523–8.
    Crossref | PubMed
  40. Prinzen FW, Hunter WC, Wyman BT, et al. Mapping of regional myocardial strain and work during ventricular pacing: experimental study using magnetic resonance imaging tagging. J Am Coll Cardiol 1999;33:1735–42.
    Crossref | PubMed
  41. Korvald C, Elvenes OP, Myrmel T. Myocardial substrate metabolism influences left ventricular energetics in vivo. Am J Physiol Heart Circ Physiol 2000;278:H1345–51.
    Crossref | PubMed
  42. Obrzut S, Tiongson J, Jamshidi N, et al. Assessment of metabolic phenotypes in patients with non-ischemic dilated cardiomyopathy undergoing cardiac resynchronization therapy. J Cardiovasc Transl Res 2010;3:643–51.
    Crossref | PubMed
  43. Wallhaus TR, Taylor M, DeGrado TR, et al. Myocardial free fatty acid and glucose use after carvedilol treatment in patients with congestive heart failure. Circulation 2001;103:2441–6.
    Crossref | PubMed
  44. Shen W, Spindler M, Higgins MA, et al. The fall in creatine levels and creatine kinase isozyme changes in the failing heart are reversible: complex post-transcriptional regulation of the components of the CK system. J Mol Cell Cardiol 2005;39:537–44.
    Crossref
  45. Hermann G, Decherd GM. The chemical nature of heart failure. Ann Intern Med 1939;12:1233–44.
    Crossref
  46. Scolletta S, Biagioli B. Energetic myocardial metabolism and oxidative stress: let’s make them our friends in the fight against heart failure. Biomed Pharmacother 2010;64:203–7.
    Crossref | PubMed
  47. Ingwall JS. Is cardiac failure a consequence of decreased energy reserve? Circulation 1993;87(VII):58–62.
  48. Ingwall JS, Weiss RG. Is the failing heart energy starved? On using chemical energy to support cardiac function. Circ Res 2004;95:135–45.
    Crossref | PubMed
  49. Kim TT, Dyck JR. Is AMPK the savior of the failing heart? Trends Endocrinol Metab 2015;26:40–8.
    Crossref | PubMed
  50. Young LH, Coven DL, Russell RR 3rd. Cellular and molecular regulation of cardiac glucose transport. J Nucl Cardiol 2000;7:267–76.
    PubMed
  51. Tian R, Musi N, D’Agostino J, et al. Increased adenosine monophosphate-activated protein kinase activity in rat hearts with pressure-overload hypertrophy. Circulation 2001;104:1664–9.
    PubMed
  52. Saavedra WF, Paolocci N, St John ME, et al. Imbalance between xanthine oxidase and nitric oxide synthase signaling pathways underlies mechanoenergetic uncoupling in the failing heart. Circ Res 2002;90:297–304.
    PubMed
  53. Hein S, Kostin S, Heling A, et al. The role of the cytoskeleton in heart failure. Cardiovasc Res 2000;45:273–8.
    PubMed
  54. Teerlink JR. A novel approach to improve cardiac performance: cardiac myosin activators. Heart Fail Rev 2009;14:289–98.
    Crossref | PubMed
  55. Bartolák-Suki E, Imsirovic J, Nishibori Y, et al. Regulation of mitochondrial structure and dynamics by the cytoskeleton and mechanical factors. Int J Mol Sci 2017;18.
    Crossref | PubMed
  56. Schilling J, Kelly DP. The PGC-1 cascade as a therapeutic target for heart failure. J Mol Cell Cardiol 2011;51:578–83.
    Crossref | PubMed
  57. St-Pierre J, Drori S, Uldry M, et al. Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell 2006;127:397–408.
    Crossref | PubMed
  58. Chakir K, Daya SK, Tunin RS, et al. Reversal of global apoptosis and regional stress kinase activation by cardiac resynchronization. Circulation 2008;117:1369–77.
    Crossref | PubMed
  59. D’Ascia C, Cittadini A, Monti MG, et al. Effects of biventricular pacing on interstitial remodelling, tumor necrosis factor-alpha expression, and apoptotic death in failing human myocardium. Eur Heart J 2006;27:201–6.
    Crossref | PubMed
  60. Liao XD, Wang XH, Jin HJ, et al. Mechanical stretch induces mitochondria-dependent apoptosis in neonatal rat cardiomyocytes and G2/M accumulation in cardiac fibroblasts. Cell Res 2004;14:16–26.
    Crossref | PubMed
  61. Kolwicz SC Jr, Tian R. Metabolic therapy at the crossroad: how to optimize myocardial substrate utilization? Trends Cardiovasc Med 2009;19:201–7.
    Crossref | PubMed
  62. Aubert G, Martin OJ, Horton JL, et al. The failing heart relies on ketone bodies as a fuel. Circulation 2016;133:698–705.
    Crossref | PubMed
  63. Cotter DG, Schugar RC, Crawford PA. Ketone body metabolism and cardiovascular disease. Am J Physiol Heart Circ Physiol 2013;304:H1060–76.
    Crossref | PubMed
  64. Sun H, Olson KC, Gao C, et al. Catabolic defect of branched-chain amino acids promotes heart failure. Circulation 2016;133:2038–49.
    Crossref | PubMed
  65. Wang W, Zhang F, Xia Y, et al. Defective branched chain amino acid catabolism contributes to cardiac dysfunction and remodeling following myocardial infarction. Am J Physiol Heart Circ Physiol 2016;311:H1160–9.
    Crossref | PubMed
  66. St John Sutton MG, Plappert T, Abraham WT, et al. Effect of cardiac resynchronization therapy on left ventricular size and function in chronic heart failure. Circulation 2003;107:1985–90.
    Crossref | PubMed
  67. Knaapen P, van Campen LM, de Cock CC, et al. Effects of cardiac resynchronization therapy on myocardial perfusion reserve. Circulation 2004;110:646–51.
    Crossref | PubMed
  68. Diaz-Infante E, Mont L, Leal J, et al. Predictors of lack of response to resynchronization therapy. Am J Cardiol 2005;95:1436–40.
    Crossref | PubMed
  69. Birnie DH, Tang AS. The problem of non-response to cardiac resynchronization therapy. Curr Opin Cardiol 2006;21:20–6.
    Crossref | PubMed
  70. Bleeker GB, Kaandorp TA, Lamb HJ, et al. Effect of posterolateral scar tissue on clinical and echocardiographic improvement after cardiac resynchronization therapy. Circulation 2006;113:969–76.
    Crossref | PubMed
  71. Reuter S, Garrigue S, Barold SS, et al. Comparison of characteristics in responders versus nonresponders with biventricular pacing for drug-resistant congestive heart failure. Am J Cardiol 2002;89:346–50.
    Crossref | PubMed
  72. Lindner O, Vogt J, Kammeier A, et al. Effect of cardiac resynchronization therapy on global and regional oxygen consumption and myocardial blood flow in patients with non-ischaemic and ischaemic cardiomyopathy. Eur Heart J 2005;26:70-6.
    Crossref | PubMed
  73. Gasparini M, Mantica M, Galimberti P, et al. Is the outcome of cardiac resynchronization therapy related to the underlying etiology? Pacing Clin Electrophysiol 2003;26:175–80.
    Crossref | PubMed
  74. Hummel JP, Lindner JR, Belcik JT, et al. Extent of myocardial viability predicts response to biventricular pacing in ischemic cardiomyopathy. Heart Rhythm 2005;2:1211–7.
    Crossref | PubMed
  75. Kass DA, Chen CH, Curry C, et al. Improved left ventricular mechanics from acute VDD pacing in patients with dilated cardiomyopathy and ventricular conduction delay. Circulation 1999;99:1567–73.
    PubMed
  76. Nelson GS, Curry CW, Wyman BT, et al. Predictors of systolic augmentation from left ventricular preexcitation in patients with dilated cardiomyopathy and intraventricular conduction delay. Circulation 2000;101:2703–9.
    PubMed
  77. Curry CW, Nelson GS, Wyman BT, et al. Mechanical dyssynchrony in dilated cardiomyopathy with intraventricular conduction delay as depicted by 3D tagged magnetic resonance imaging. Circulation 2000;101:E2.
    PubMed
  78. Katz AM. Is the failing heart an energy-starved organ? J Card Fail 1996;2:267–72.
    PubMed
  79. Ingwall JS, Kelly RA. Nitric oxide, myocardial oxygen consumption, and ATP synthesis. Circ Res 1998;83:1067–8.
    PubMed
  80. Suga H, Igarashi Y, Yamada O, et al. Mechanical efficiency of the left ventricle as a function of preload, afterload, and contractility. Heart Vessels 1985;1:3–8.
    PubMed
  81. Nelson GS, Berger RD, Fetics BJ, et al. Left ventricular or biventricular pacing improves cardiac function at diminished energy cost in patients with dilated cardiomyopathy and left bundle-branch block. Circulation 2000;102:3053–9.
    PubMed
  82. Duncan AM, Francis DP, Henein MY, et al. Limitation of cardiac output by total isovolumic time during pharmacologic stress in patients with dilated cardiomyopathy: activation-mediated effects of left bundle branch block and coronary artery disease. J Am Coll Cardiol 2003;41:121–8.
    Crossref | PubMed
  83. Xiao HB, Lee CH, Gibson DG. Effect of left bundle branch block on diastolic function in dilated cardiomyopathy. Br Heart J 1991;66:443–7.
    Crossref | PubMed
  84. Grines CL, Bashore TM, Boudoulas H, et al. Functional abnormalities in isolated left bundle branch block. The effect of interventricular asynchrony. Circulation 1989;79:845–53.
    PubMed
  85. Ciampi Q, Cortigiani L, Pratali L, et al. Left bundle branch block negatively affects coronary flow velocity reserve and myocardial contractile reserve in nonischemic dilated cardiomyopathy. J Am Soc Echocardiogr 2016;29:112–8.
    Crossref | PubMed
  86. Vitale GD, deKemp RA, Ruddy TD, et al. Myocardial glucose utilization and optimization of (18)F-FDG PET imaging in patients with non-insulin-dependent diabetes mellitus, coronary artery disease, and left ventricular dysfunction. J Nucl Med 2001;42:1730–6.
    PubMed
  87. Dávila-Román VG, Vedala G, Herrero P, et al. Altered myocardial fatty acid and glucose metabolism in idiopathic dilated cardiomyopathy. J Am Coll Cardiol 2002;40:271–7.
    Crossref | PubMed
  88. Lewandowski ED. Cardiac carbon 13 magnetic resonance spectroscopy: on the horizon or over the rainbow? J Nucl Cardiol 2002;9:419–28.
    PubMed
  89. Ning XH, Zhang J, Liu J, et al. Signaling and expression for mitochondrial membrane proteins during left ventricular remodeling and contractile failure after myocardial infarction. J Am Coll Cardiol 2000;36:282–7.
    Crossref | PubMed
  90. Ono S, Nohara R, Kambara H, et al. Regional myocardial perfusion and glucose metabolism in experimental left bundle branch block. Circulation 1992;85:1125–31.
    PubMed
  91. Doenst T, Goodwin GW, Cedars AM, et al. Load-induced changes in vivo alter substrate fluxes and insulin responsiveness of rat heart in vitro. Metabolism 2001;50:1083–90.
    Crossref | PubMed
  92. Ukkonen H, Beanlands RS, Burwash IG, et al. Effect of cardiac resynchronization on myocardial efficiency and regional oxidative metabolism. Circulation 2003;107:28–31.
    PubMed
  93. Sundell J, Engblom E, Koistinen J, et al. The effects of cardiac resynchronization therapy on left ventricular function, myocardial energetics, and metabolic reserve in patients with dilated cardiomyopathy and heart failure. J Am Coll Cardiol 2004;43:1027–33.
    Crossref | PubMed
  94. Auricchio A, Salo RW. Acute hemodynamic improvement by pacing in patients with severe congestive heart failure. Pacing Clin Electrophysiol 1997;20:313–24.
    PubMed
  95. Niederer SA, Lamata P, Plank G, et al. Analyses of the redistribution of work following cardiac resynchronisation therapy in a patient specific model. PLoS One 2012;7:e43504.
    Crossref | PubMed
  96. Kyriacou A, Pabari PA, Mayet J, et al. Cardiac resynchronization therapy and AV optimization increase myocardial oxygen consumption, but increase cardiac function more than proportionally. Int J Cardiol 2014;171:144–52.
    Crossref | PubMed
  97. Spragg DD, Leclercq C, Loghmani M, et al. Regional alterations in protein expression in the dyssynchronous failing heart. Circulation 2003;108:929–32.
    Crossref | PubMed
  98. Zajac J, Eriksson J, Alehagen U, et al. Mechanical dyssynchrony alters left ventricular flow energetics in failing hearts with LBBB: a 4D flow CMR pilot study. Int J Cardiovasc Imaging 2018;34:587–96.
    Crossref | PubMed
  99. Kakizaki R, Nabeta T, Ishii S, et al. Cardiac resynchronization therapy reduces left ventricular energy loss. Int J Cardiol 2016;221:546–8.
    Crossref | PubMed
  100. Siciliano M, Migliore F, Badano L, et al. Cardiac resynchronization therapy by multipoint pacing improves response of left ventricular mechanics and fluid dynamics: a three-dimensional and particle image velocimetry echo study. Europace 2017;19:1833–40.
    Crossref | PubMed
  101. Cimino S, Palombizio D, Cicogna F, et al. Significant increase of flow kinetic energy in “nonresponders” patients to cardiac resynchronization therapy. Echocardiography 2017;34:709–15.
    Crossref | PubMed
  102. Thompson K, Saab G, Birnie D, et al. Is septal glucose metabolism altered in patients with left bundle branch block and ischemic cardiomyopathy? J Nucl Med 2006;47:1763–8.
    PubMed
  103. Gudbjarnason S, Mathes P, Ravens KG. Functional compartmentation of ATP and creatine phosphate in heart muscle. J Mol Cell Cardiol 1970;1:325–39.
  104. Niazi I, Baker J 2nd, Corbisiero R, et al. Safety and efficacy of multipoint pacing in cardiac resynchronization therapy: the MultiPoint Pacing Trial. JACC Clin Electrophysiol 2017;3:1510–18.
    Crossref | PubMed
  105. Qiu Q, Yang L, Mai JT, et al. Acute Effects of multisite biventricular pacing on dyssynchrony and hemodynamics in canines with heart failure. J Card Fail 2017;23:304–11.
    Crossref | PubMed
  106. Chrysohoou C, Dilaveris P, Antoniou CK, et al. Heart failure study of multipoint pacing effects on ventriculoarterial coupling: rationale and design of the HUMVEE trial. Ann Noninvasive Electrocardiol 2018;23:e12510.
    Crossref | PubMed
  107. Kumar V, Venkataraman R, Aljaroudi W, et al. Implications of left bundle branch block in patient treatment. Am J Cardiol 2013;111:291–300.
    Crossref | PubMed
  108. ter Keurs HE, Zhang YM, Davidoff AW, et al. Damage induced arrhythmias: mechanisms and implications. Can J Physiol Pharmacol 2001;79:73–81.
    PubMed
  109. Leyva F, Foley PW. Is cardiac resynchronisation therapy proarrhythmic? Indian Pacing Electrophysiol J 2008;8:268–80.
    PubMed
  110. Bradfield JS, Shivkumar K. Cardiac resynchronization therapy-induced proarrhythmia: understanding preferential conduction within myocardial scars. Circ Arrhythm Electrophysiol 2014;7:1000–2.
    Crossref | PubMed
  111. Roque C, Trevisi N, Silberbauer J, et al. Electrical storm induced by cardiac resynchronization therapy is determined by pacing on epicardial scar and can be successfully managed by catheter ablation. Circ Arrhythm Electrophysiol 2014;7:1064–9.
    Crossref | PubMed
  112. van der Wall EE, Schalij MJ, van der Laarse A, Bax JJ. Cardiac resynchronization therapy; the importance of evaluating cardiac metabolism. Int J Cardiovasc Imaging 2010;26:293–7.
    Crossref | PubMed
  113. Kitaizumi K, Yukiiri K, Masugata H, et al. Positron emission tomographic demonstration of myocardial oxidative metabolism in a case of left ventricular restoration after cardiac resynchronization therapy. Circ J 2008;72:1900–3.
    Crossref | PubMed
  114. Agnetti G, Kaludercic N, Kane LA, et al. Modulation of mitochondrial proteome and improved mitochondrial function by biventricular pacing of dyssynchronous failing hearts. Circ Cardiovasc Genet 2010; 3:78–87.
    Crossref | PubMed
  115. Nemutlu E, Zhang S, Xu YZ, et al. Cardiac resynchronization therapy induces adaptive metabolic transitions in the metabolomic profile of heart failure. J Card Fail 2015;21:460–9.
    Crossref | PubMed
  116. Diwan A, Dorn GW 2nd. Decompensation of cardiac hypertrophy: cellular mechanisms and novel therapeutic targets. Physiology (Bethesda) 2007;22:56–64.
    Crossref | PubMed
  117. Fournier NC, Rahim M. Control of energy production in the heart: a new function for fatty acid binding protein. Biochemistry 1985;24:2387–96.
    Crossref | PubMed
  118. Paradies G, Petrosillo G, Paradies V, et al. Oxidative stress, mitochondrial bioenergetics, and cardiolipin in aging. Free Radic Biol Med 2010;48:1286–95.
    Crossref | PubMed
  119. Wen JJ, Garg N. Oxidative modification of mitochondrial respiratory complexes in response to the stress of Trypanosoma cruzi infection. Free Radic Biol Med 2004;37:2072–81.
    Crossref | PubMed
  120. Hill BG, Bhatnagar A. Protein S-glutathiolation: redox-sensitive regulation of protein function. J Mol Cell Cardiol 2012;52:559–67.
    Crossref | PubMed
  121. Chen CA, Wang TY, Varadharaj S, et al. S-glutathionylation uncouples eNOS and regulates its cellular and vascular function. Nature 2010;468:1115–8.
    Crossref | PubMed
  122. Mieyal JJ, Gallogly MM, Qanungo S, et al. Molecular mechanisms and clinical implications of reversible protein S-glutathionylation. Antioxid Redox Signal 2008;10:1941–88.
    Crossref | PubMed
  123. Holmgren A, Lu J. Thioredoxin and thioredoxin reductase: current research with special reference to human disease. Biochem Biophys Res Commun 2010;396:120–4.
    Crossref | PubMed
  124. Wang SB, Foster DB, Rucker J, et al. Redox regulation of mitochondrial ATP synthase: implications for cardiac resynchronization therapy. Circ Res 2011;109:750–7.
    Crossref | PubMed
  125. Sugamura K, Keaney JF, Jr. Reactive oxygen species in cardiovascular disease. Free Radic Biol Med 2011;51:978–92.
    Crossref | PubMed
  126. McCarty MF. Practical prevention of cardiac remodeling and atrial fibrillation with full-spectrum antioxidant therapy and ancillary strategies. Med Hypotheses 2010;75:141–7.
    Crossref | PubMed
  127. Zweier JL, Talukder MA. The role of oxidants and free radicals in reperfusion injury. Cardiovasc Res 2006;70:181–90.
    Crossref | PubMed
  128. Foster DB, Van Eyk JE, Marban E, et al. Redox signaling and protein phosphorylation in mitochondria: progress and prospects. J Bioenerg Biomembr 2009;41:159–68.
    Crossref | PubMed
  129. Ying J, Clavreul N, Sethuraman M, et al. Thiol oxidation in signaling and response to stress: detection and quantification of physiological and pathophysiological thiol modifications. Free Radic Biol Med 2007;43:1099–108.
    Crossref | PubMed
  130. Burwell LS, Brookes PS. Mitochondria as a target for the cardioprotective effects of nitric oxide in ischemia-reperfusion injury. Antioxid Redox Signal 2008;10:579–99.
    Crossref | PubMed
  131. Tatsuta T, Model K, Langer T. Formation of membrane-bound ring complexes by prohibitins in mitochondria. Mol Biol Cell 2005;16:248–59.
    Crossref | PubMed
  132. Arnold I, Langer T. Membrane protein degradation by AAA proteases in mitochondria. Biochim Biophys Acta 2002;1592:89–96.
    PubMed
  133. Augustin S, Nolden M, Muller S, et al. Characterization of peptides released from mitochondria: evidence for constant proteolysis and peptide efflux. J Biol Chem 2005;280:2691–9.
    Crossref | PubMed
  134. Barth AS, Aiba T, Halperin V, et al. Cardiac resynchronization therapy corrects dyssynchrony-induced regional gene expression changes on a genomic level. Circ Cardiovasc Genet 2009;2:371–8.
    Crossref | PubMed
  135. Manfra O, Frisk M, Louch WE. Regulation of cardiomyocyte t-tubular structure: opportunities for therapy. Curr Heart Fail Rep 2017;14:167–78.
    Crossref | PubMed
  136. Ibrahim M, Terracciano CM. Reversibility of T-tubule remodelling in heart failure: mechanical load as a dynamic regulator of the T-tubules. Cardiovasc Res 2013;98:225–32.
    Crossref | PubMed
  137. Stachowski MJ, Holewinski RJ, Grote E, et al. Phospho-proteomic analysis of cardiac dyssynchrony and resynchronization therapy. Proteomics 2018;18:e1800079.
    Crossref | PubMed
  138. Ahmad KA, Wang G, Unger G, et al. Protein kinase CK2 – a key suppressor of apoptosis. Adv Enzyme Regul 2008;48:179–87.
    Crossref | PubMed
  139. Litchfield DW. Protein kinase CK2: structure, regulation and role in cellular decisions of life and death. Biochem J 2003;369:1–15.
    Crossref | PubMed
  140. Jaussaud J, Blanc P, Bordachar P, et al. Response to cardiac resynchronization therapy: the muscular metabolic pathway. Cardiol Res Pract 2010;2011:830279.
    Crossref | PubMed
  141. Xiao RP, Zhang SJ, Chakir K, et al. Enhanced G(i) signaling selectively negates beta2-adrenergic receptor (AR) – but not beta1-AR-mediated positive inotropic effect in myocytes from failing rat hearts. Circulation 2003;108:1633–9.
    Crossref | PubMed
  142. Gong H, Adamson DL, Ranu HK, et al. The effect of Gi-protein inactivation on basal, and beta(1)- and beta(2)AR-stimulated contraction of myocytes from transgenic mice overexpressing the beta(2)-adrenoceptor. Br J Pharmacol 2000;131:594–600.
    Crossref | PubMed
  143. Rau T, Nose M, Remmers U, et al. Overexpression of wild-type Galpha(i)-2 suppresses beta-adrenergic signaling in cardiac myocytes. FASEB J 2003;17:523–5.
    Crossref | PubMed
  144. Raake PW, Vinge LE, Gao E, et al. G protein-coupled receptor kinase 2 ablation in cardiac myocytes before or after myocardial infarction prevents heart failure. Circ Res 2008;103:413–22.
    Crossref | PubMed
  145. Chakir K, Depry C, Dimaano VL, et al. Galphas-biased beta2-adrenergic receptor signaling from restoring synchronous contraction in the failing heart. Sci Transl Med 2011;3:100ra88.
    Crossref | PubMed
  146. Kuschel M, Zhou YY, Cheng H, et al. G(i) protein-mediated functional compartmentalization of cardiac beta(2)-adrenergic signaling. J Biol Chem 1999;274:22048–52.
    PubMed
  147. Aiba T, Hesketh GG, Barth AS, et al. Electrophysiological consequences of dyssynchronous heart failure and its restoration by resynchronization therapy. Circulation 2009;119:1220–30.
    Crossref | PubMed
  148. Nishijima Y, Sridhar A, Viatchenko-Karpinski S, et al. Chronic cardiac resynchronization therapy and reverse ventricular remodeling in a model of nonischemic cardiomyopathy. Life Sci 2007;81:1152–9.
    Crossref | PubMed
  149. Cho H, Barth AS, Tomaselli GF. Basic science of cardiac resynchronization therapy: molecular and electrophysiological mechanisms. Circ Arrhythm Electrophysiol 2012;5:594–603.
    Crossref | PubMed
  150. Kirk JA, Chakir K, Lee KH, et al. Pacemaker-induced transient asynchrony suppresses heart failure progression. Sci Transl Med 2015;7:319ra207.
    Crossref | PubMed
  151. Kirk JA, Holewinski RJ, Kooij V, et al. Cardiac resynchronization sensitizes the sarcomere to calcium by reactivating GSK-3beta. J Clin Invest 2014;124:129–38.
    Crossref | PubMed
  152. Vanderheyden M, Mullens W, Delrue L, et al. Myocardial gene expression in heart failure patients treated with cardiac resynchronization therapy responders versus nonresponders. J Am Coll Cardiol 2008;51:129–36.
    Crossref | PubMed
  153. Cheng A, Nguyen TC, Malinowski M, et al. Heterogeneity of left ventricular wall thickening mechanisms. Circulation 2008;118:713–21.
    Crossref | PubMed
  154. LeGrice IJ, Takayama Y, Covell JW. Transverse shear along myocardial cleavage planes provides a mechanism for normal systolic wall thickening. Circ Res 1995;77:182–93.
    PubMed
  155. Marijianowski MM, Teeling P, Mann J, Becker AE. Dilated cardiomyopathy is associated with an increase in the type I/type III collagen ratio: a quantitative assessment. J Am Coll Cardiol 1995;25:1263–72.
    Crossref | PubMed
  156. Constantino J, Hu Y, Trayanova NA. A computational approach to understanding the cardiac electromechanical activation sequence in the normal and failing heart, with translation to the clinical practice of CRT. Prog Biophys Mol Biol 2012;110:372–9.
    Crossref | PubMed
  157. Akar FG, Nass RD, Hahn S, et al. Dynamic changes in conduction velocity and gap junction properties during development of pacing-induced heart failure. Am J Physiol Heart Circ Physiol 2007;293:H1223–30.
    Crossref | PubMed
  158. Akar FG, Spragg DD, Tunin RS, et al. Mechanisms underlying conduction slowing and arrhythmogenesis in nonischemic dilated cardiomyopathy. Circ Res 2004;95:717–25.
    Crossref | PubMed
  159. Zhao G, Qiu Y, Zhang HM, et al. Intercalated discs: cellular adhesion and signaling in heart health and diseases. Heart Fail Rev 2019;24:115–32.
    Crossref | PubMed
  160. Nawata J, Ohno I, Isoyama S, et al. Differential expression of alpha 1, alpha 3 and alpha 5 integrin subunits in acute and chronic stages of myocardial infarction in rats. Cardiovasc Res 1999;43:371–81.
    PubMed
  161. Samarel AM. Focal adhesion signaling in heart failure. Pflugers Arch 2014;466:1101–11.
    Crossref | PubMed
  162. Pardo JV, Siliciano JD, Craig SW. A vinculin-containing cortical lattice in skeletal muscle: transverse lattice elements (“costameres”) mark sites of attachment between myofibrils and sarcolemma. Proc Natl Acad Sci U S A 1983;80:1008–12.
    PubMed
  163. Pardo JV, Siliciano JD, Craig SW. Vinculin is a component of an extensive network of myofibril-sarcolemma attachment regions in cardiac muscle fibers. J Cell Biol 1983;97:1081–8.
    Crossref | PubMed
  164. Samarel AM. Costameres, focal adhesions, and cardiomyocyte mechanotransduction. Am J Physiol Heart Circ Physiol 2005;289:H2291-301.
    Crossref | PubMed
  165. Jaka O, Casas–Fraile L, Lopez de Munain A, et al. Costamere proteins and their involvement in myopathic processes. Expert Rev Mol Med 2015;17:e12.
    Crossref | PubMed
  166. Peter AK, Cheng H, Ross RS, et al. The costamere bridges sarcomeres to the sarcolemma in striated muscle. Prog Pediatr Cardiol 2011;31:83–88.
    Crossref | PubMed
  167. Ervasti JM, Campbell KP. A role for the dystrophin-glycoprotein complex as a transmembrane linker between laminin and actin. J Cell Biol 1993;122:809–23.
    PubMed
  168. Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell 2002;110:673–87.
    PubMed
  169. Luo BH, Carman CV, Springer TA. Structural basis of integrin regulation and signaling. Annu Rev Immunol 2007;25:619–47.
    Crossref | PubMed
  170. Ginsberg MH, Partridge A, Shattil SJ. Integrin regulation. Curr Opin Cell Biol 2005;17:509–16.
    Crossref | PubMed
  171. Zemljic-Harpf AE, Miller JC, Henderson SA, et al. Cardiac-myocyte-specific excision of the vinculin gene disrupts cellular junctions, causing sudden death or dilated cardiomyopathy. Mol Cell Biol 2007;27:7522–37.
    Crossref | PubMed
  172. Sorge M, Brancaccio M. Melusin promotes a protective signal transduction cascade in stressed hearts. Front Mol Biosci 2016;3:53.
    Crossref | PubMed
  173. De Acetis M, Notte A, Accornero F, et al. Cardiac overexpression of melusin protects from dilated cardiomyopathy due to long-standing pressure overload. Circ Res 2005;96:1087–94.
    Crossref | PubMed
  174. Brancaccio M, Fratta L, Notte A, et al. Melusin, a muscle-specific integrin beta1-interacting protein, is required to prevent cardiac failure in response to chronic pressure overload. Nat Med 2003;9:68–75.
    Crossref | PubMed
  175. Brokat S, Thomas J, Herda LR, et al. Altered melusin expression in the hearts of aortic stenosis patients. Eur J Heart Fail 2007;9:568–73.
    Crossref | PubMed
  176. Dullens HF, Schipper ME, van Kuik J, et al. Integrin expression during reverse remodeling in the myocardium of heart failure patients. Cardiovasc Pathol 2012;21:291–8.
    Crossref | PubMed
  177. Pyle WG, Hart MC, Cooper JA, et al. Actin capping protein: an essential element in protein kinase signaling to the myofilaments. Circ Res 2002;90:1299–306.
    PubMed
  178. Gupta MP, Samant SA, Smith SH, et al. HDAC4 and PCAF bind to cardiac sarcomeres and play a role in regulating myofilament contractile activity. J Biol Chem 2008;283:10135–46.
    Crossref | PubMed
  179. Kovacic-Milivojevic B, Roediger F, Almeida EA, et al. Focal adhesion kinase and p130Cas mediate both sarcomeric organization and activation of genes associated with cardiac myocyte hypertrophy. Mol Biol Cell 2001;12:2290–307.
    Crossref | PubMed
  180. Agnetti G, Halperin VL, Kirk JA, et al. Desmin modifications associate with amyloid-like oligomers deposition in heart failure. Cardiovasc Res 2014;102:24–34.
    Crossref | PubMed
  181. Otey CA, Carpen O. Alpha-actinin revisited: a fresh look at an old player. Cell Motil Cytoskeleton 2004;58:104–11.
    Crossref | PubMed
  182. Melo TG, Almeida DS, Meirelles MN, Pereira MC. Disarray of sarcomeric alpha-actinin in cardiomyocytes infected by Trypanosoma cruzi. Parasitology 2006;133:171–8.
    Crossref | PubMed
  183. Lichter JG, Carruth E, Mitchell C, et al. Remodeling of the sarcomeric cytoskeleton in cardiac ventricular myocytes during heart failure and after cardiac resynchronization therapy. J Mol Cell Cardiol 2014;72:186–95.
    Crossref | PubMed
  184. Fletcher DA, Mullins RD. Cell mechanics and the cytoskeleton. Nature 2010;463:485–92.
    Crossref | PubMed
  185. Heggeness MH, Simon M, Singer SJ. Association of mitochondria with microtubules in cultured cells. Proc Natl Acad Sci U S A 1978;75:3863–6.
    PubMed
  186. Engler AJ, Sen S, Sweeney HL, et al. Matrix elasticity directs stem cell lineage specification. Cell 2006;126:677–89.
    Crossref | PubMed
  187. Ali MH, Pearlstein DP, Mathieu CE, et al. Mitochondrial requirement for endothelial responses to cyclic strain: implications for mechanotransduction. Am J Physiol Lung Cell Mol Physiol 2004;287:L486–96.
    Crossref | PubMed
  188. Bartolák-Suki E, Imsirovic J, Parameswaran H, et al. Fluctuation-driven mechanotransduction regulates mitochondrial-network structure and function. Nat Mater 2015;14:1049–57.
    Crossref | PubMed
  189. Lyra-Leite DM, Andres AM, Petersen AP, et al. Mitochondrial function in engineered cardiac tissues is regulated by extracellular matrix elasticity and tissue alignment. Am J Physiol Heart Circ Physiol 2017;313:H757–67.
    Crossref | PubMed
  190. Kirk JA, Kass DA. Cellular and molecular aspects of dyssynchrony and resynchronization. Card Electrophysiol Clin 2015;7:585–97.
    Crossref | PubMed
  191. Hu Y, Gurev V, Constantino J, et al. Optimizing cardiac resynchronization therapy to minimize ATP consumption heterogeneity throughout the left ventricle: a simulation analysis using a canine heart failure model. Heart Rhythm 2014;11:1063–9.
    Crossref | PubMed
  192. Eichhorn EJ, Heesch CM, Barnett JH, et al. Effect of metoprolol on myocardial function and energetics in patients with nonischemic dilated cardiomyopathy: a randomized, double-blind, placebo-controlled study. J Am Coll Cardiol 1994;24:1310–20.
    Crossref | PubMed
  193. Eichhorn EJ, Bedotto JB, Malloy CR, et al. Effect of beta-adrenergic blockade on myocardial function and energetics in congestive heart failure. Improvements in hemodynamic, contractile, and diastolic performance with bucindolol. Circulation 1990;82:473–83.
    PubMed
  194. Saxon LA, Ellenbogen KA. Resynchronization therapy for the treatment of heart failure. Circulation 2003;108:1044–8.
    Crossref | PubMed
  195. Penicka M, Bartunek J, De Bruyne B, et al. Improvement of left ventricular function after cardiac resynchronization therapy is predicted by tissue Doppler imaging echocardiography. Circulation 2004;109:978–83.
    Crossref | PubMed
  196. Bax JJ, Bleeker GB, Marwick TH, et al. Left ventricular dyssynchrony predicts response and prognosis after cardiac resynchronization therapy. J Am Coll Cardiol 2004;44:1834–40.
    Crossref | PubMed
  197. Yu CM, Fung WH, Lin H, et al. Predictors of left ventricular reverse remodeling after cardiac resynchronization therapy for heart failure secondary to idiopathic dilated or ischemic cardiomyopathy. Am J Cardiol 2003;91:684–8.
    Crossref | PubMed
  198. Molhoek SG, van Erven L, Bootsma M, et al. QRS duration and shortening to predict clinical response to cardiac resynchronization therapy in patients with end-stage heart failure. Pacing Clin Electrophysiol 2004;27:308–13.
    Crossref | PubMed
  199. Lecoq G, Leclercq C, Leray E, et al. Clinical and electrocardiographic predictors of a positive response to cardiac resynchronization therapy in advanced heart failure. Eur Heart J 2005;26:1094–100.
    Crossref | PubMed
  200. Davis DR, Krahn AD, Tang AS, et al. Long-term outcome of cardiac resynchronization therapy in patients with severe congestive heart failure. Can J Cardiol 2005;21:413–7.
    PubMed
  201. Buck S, Maass AH, van Veldhuisen DJ, et al. Cardiac resynchronisation therapy and the role of optimal device utilisation. Neth Heart J 2009;17:354–7.
    PubMed
  202. van Hemel NM, Scheffer M. Cardiac resynchronisation therapy in daily practice and loss of confidence in predictive techniques to response. Neth Heart J 2009;17:4–5.
    PubMed
  203. Bleeker GB, Holman ER, Steendijk P, et al. Cardiac resynchronization therapy in patients with a narrow QRS complex. J Am Coll Cardiol 2006;48:2243–50.
    Crossref | PubMed
  204. Warnholtz A, Nickenig G, Schulz E, et al. Increased NADH-oxidase-mediated superoxide production in the early stages of atherosclerosis: evidence for involvement of the renin-angiotensin system. Circulation 1999;99:2027–33.
    Crossref | PubMed
  205. Paolisso G, Gambardella A, Galzerano D, et al. Total-body and myocardial substrate oxidation in congestive heart failure. Metabolism 1994;43:174–9.
    Crossref | PubMed
  206. Lommi J, Kupari M, Yki-Järvinen H. Free fatty acid kinetics and oxidation in congestive heart failure. Am J Cardiol 1998;81:45–50.
    Crossref | PubMed
  207. Zhou YT, Grayburn P, Karim A, et al. Lipotoxic heart disease in obese rats: implications for human obesity. Proc Natl Acad Sci U S A 2000;97:1784–9.
    Crossref | PubMed
  208. Hudsmith LE, Neubauer S. Magnetic resonance spectroscopy in myocardial disease. JACC Cardiovasc Imaging 2009;2:87–96.
    Crossref | PubMed
  209. Abdurrachim D, Prompers JJ. Evaluation of cardiac energetics by non-invasive 31P magnetic resonance spectroscopy. Biochim Biophys Acta Mol Basis Dis 2018;1864:1939–48.
    Crossref | PubMed
  210. Park RC, Little WC, O’Rourke RA. Effect of alteration of left ventricular activation sequence on the left ventricular end-systolic pressure-volume relation in closed-chest dogs. Circ Res 1985;57:706–17.
    PubMed
  211. Wyman BT, Hunter WC, Prinzen FW, et al. Mapping propagation of mechanical activation in the paced heart with MRI tagging. Am J Physiol 1999;276:H881–91.
    Crossref | PubMed
  212. Karbowski M, Spodnik JH, Teranishi M, et al. Opposite effects of microtubule-stabilizing and microtubule-destabilizing drugs on biogenesis of mitochondria in mammalian cells. J Cell Sci 2001;114:281–91.
    PubMed