Metabolic Approaches for Treatment of Dilated Cardiomyopathy: Comparison
Please note this is a comparison between Version 2 by Peter Tang and Version 3 by Peter Tang.

In dilated cardiomyopathy (DCM), where the heart muscle becomes stretched and thin, heart failure (HF) occurs, and the cardiomyocytes suffer from an energetic inefficiency caused by an abnormal cardiac metabolism. Although underappreciated as a potential therapeutic target, the optimal metabolic milieu of a failing heart is still largely unknown and subject to debate. Because glucose naturally has a lower P/O ratio (the ATP yield per oxygen atom), the previous studies using this strategy to increase glucose oxidation have produced some intriguing findings. In reality, the vast majority of small-scale pilot trials using trimetazidine, ranolazine, perhexiline, and etomoxir have demonstrated enhanced left ventricular (LV) function and, in some circumstances, myocardial energetics in chronic ischemic and non-ischemic HF with a reduced ejection fraction (EF).

  • dilated cardiomyopathy
  • heart failure
  • metabolic therapy
  • myocardial energetics
  • SGLT2 inhibitors

1. The Purpose of Metabolic Therapy in Dilated Cardiomyopathy

Dilated cardiomyopathy (DCM) is a primary myocardial disorder characterized by enlarged ventricles with a contractile deficit that leads to reduced ventricular function in the absence of volume or to pressure overload, congenital heart disease (CHD), or significant coronary artery disease (CAD) [1]. Genetic and non-genetic causes underlie its pathogenesis, in which a mixed mechanism is often involved [2]. Its estimated prevalence in the general population ranges from 1:500 to 1:2500 [3][4].
DCM is a recognized cause of systolic heart failure (HF), a condition in which the impaired cardiac pump is unable to fulfil the energetic demand of the body in terms of nutrients and oxygen supply. HF has been defined as the cardiovascular epidemic of the 21st century [5], with an estimated prevalence of about 2–4% in the general adult population; this prevalence is known to increase with age [6][7]. Moreover, due to the prevalence of HF in high-income countries, significant amounts of human and economic resources are employed in this field [8].
Multiple factors, such as hemodynamics, neurohormones, and genetics, participate in progressive heart failure remodeling [9]. The efficiency of the myocardial pump depends mainly on the metabolism of the cardiac cells [10]. However, the hyperactivation of the adrenergic system and the renin–angiotensin–aldosterone system (RAAS), when attempting to sustain hemodynamic failure, contributes to indirect changes in the metabolism of the cardiac and skeletal muscles, worsening their efficiency [11]. In the last decades, better knowledge of the pathophysiological mechanisms of the effects of the neurohormonal axis on the cardiovascular (CV) system has enabled the adoption of drugs that block this detrimental activation. Despite the development of additional molecules targeting different pathological pathways, as outlined by the European and American guidelines on HF [12][13], a specific phenotype-oriented therapy is generally lacking, and the HF prognosis remains poor [14].
In the last decades, there has been a growing interest in the cardiac metabolism; the aim has been to find new potential disease biomarkers and therapeutic targets to improve the HF prognosis [15].
A healthy adult heart produces adenosine triphosphate (ATP) by metabolizing different types of fuels (fatty acids, glucose, lactate, ketones, and amino acids), primarily via oxygen-dependent mitochondrial oxidative phosphorylation and the electron transport chain and, to a lesser extent, the anaerobic glycolytic pathway [16]. Between 60 and 80% of the energy produced by the healthy heart is derived from free fatty acid (FFA) oxidation, despite a metabolic flexibility that allows a shift between different energy substrates to maintain ATP production [16]. However, FFA oxidation is a less efficient source of energy production than glucose oxidation (in terms of the produced ATP per consumed O2 molecules). Of note, the amount of ATP produced per O2 molecules consumed is greater for glucose oxidation compared to that of FFAs. For example, the mitochondrial ATP yield per oxygen atom (P/O ratio) is only 2.33 for long-chain fatty acids, whereas it is 2.50 for ketone bodies and 2.58 for glucose. Consequently, oxidizing glucose results in an increase in cardiac efficiency of up to 30% [17].
The failing heart is considered to be an “engine out of fuel” [18]. A reduction in the mitochondrial oxidative capacity is the first metabolic change characterizing the heart’s deteriorating energy deficiency [19][20]. The ensuing increased glycolytic pathway [21] is not as efficient as mitochondrial phosphorylation and fails to compensate for the status of the energy deficit; this also leads to H+ accumulation in the cytoplasm as glycolysis is uncoupled from the oxidation of pyruvate and lactate [22][23]. Moreover, a highly significant shift in substrate utilization also occurs. A decrease in FFA oxidation has been observed in humans with idiopathic DCM [24], despite evidence from other studies that is not consistent with this finding [25][26]. Furthermore, despite a reduction in FFA oxidation, the failing heart still mainly counts on this substrate for the highest proportion of mitochondrial ATP generation [27].
Changes in substrate utilization in the failing heart have also been observed relative to glucose metabolism, ketone bodies, and long-chain amino acids [15]. More metabolites have been found to be involved in the pathophysiological process of HF; thus, the studies on HF are no longer limited to glucose and FA metabolism [28]. When glucose and lipid metabolism decrease, it is possible to hypothesize that ketone bodies may act as alternative substrates in failing hearts [28]. Due to insulin resistance and other factors, the failing heart has significantly reduced glucose and fatty acid utilization, and ketone bodies are fast-metabolizing small molecular energy substrates that the heart can use to improve cardiac efficiency. In addition, recent research has demonstrated that branched-chain amino acids (BCAAs) also play a significant role in the pathophysiology of end-stage heart failure. The myocardial BCAA metabolism can effectively improve cardiac function and slow the progression of heart failure [28]. Therefore, the metabolic remodeling of small molecular substrates such as ketone bodies and amino acids also plays a significant role in the onset and progression of HF, in addition to the changes in the metabolism of glucose and FAs during HF. However, there have been conflicting results; these results are partially explained by the disparities in the severity of the disease and the presence of other medical conditions, such as metabolic syndrome, in the individuals participating in human clinical studies.
In addition, in systolic heart failure, metabolic changes can occur regardless of weight status. However, the specific metabolic changes seen in obese, normal-weight, and underweight individuals may vary. There may be an increased reliance on fatty acid oxidation as a source of energy for the heart in obese people with heart failure. This can lead to an impaired glucose metabolism and decreased myocardial glucose uptake and oxidation. In addition, metabolic dysregulation in the heart may be exacerbated by insulin resistance and inflammation, which are frequently associated with obesity. As previously mentioned, heart failure can cause metabolic changes, such as an increase in glycolysis as a source of myocardial energy, even in healthy people. In heart failure, the oxidative metabolism is less efficient; so, this metabolic switch is made to make up for it. Heart failure patients who are underweight may experience metabolic changes that are comparable to those seen in people of normal weight. There may still be a shift toward more glycolysis [29]. Implementing strategies for the targeted reduction in this particular fat store in obese individuals is the obvious solution to the problem of preventing adipose tissue inflammation and the metabolic and cardiovascular complications that come with it. The treatment for insulin resistance and obesity still relies heavily on lifestyle changes, such as diet and exercise modifications [29].
Recently, the use of various “omics” technologies, such as metabolomics, has offered a new chance to enhance the understanding of the mechanisms involved in this disease and to find new biomarkers for the prognosis and diagnosis of DCM [30]. A recent comprehensive review on this topic highlighted what the recent literature has provided in terms of the metabolite-based biomarkers which are useful for predicting and diagnosing DSM and for monitoring therapeutic interventions [31]. One main limitation in understanding metabolic pathophysiology from the results of these studies is their limited sample sizes, which prevent the reaching of a definitive conclusion about the practicality of the identified DCM biomarkers for clinical purposes. Moreover, since these studies have mainly been conducted on biofluids such as serum, the alterations in plasma metabolites may represent the impact of the contribution of several organs. To overcome this last issue, a recent analysis by Flam et al. was conducted on myocardial biopsies from patients with end-stage HF; the analysis comprised metabolomics, genome-wide RNA sequencing, and global proteomic assessment [32]. The findings confirmed the significant alterations in the metabolic process of the heart that had been seen previously in HF experiments with animal subjects, including a decline in the utilization of fatty acids and a heightened dependence on the utilization of ketones and carbohydrates. These results aligned with prior studies on human and animal models of HF. However, this particular study provides a novel insight. The previous research on animal models suggested that the decrease in fatty acid oxidation was due to a decline in mitochondrial oxidative function. However, the present research revealed a scarcity of fatty acids or acylcarnitines in the damaged heart, rather than an accumulation, indicating a shortage of fatty acid supply to the heart. As plasma fatty acid levels in HF patients remain unchanged, this scarcity in heart tissue could suggest a potential issue with fatty acid import. The specific cause of this is still unknown. This discovery is significant because it provides a new avenue for the targeting of the fatty acid metabolism in the treatment of HF. It is essential to determine whether similar changes occur during the development of HF and not just as a result of advanced disease. The study found that multiple classes of carbon substrates, including many amino acids, tricarboxylic acid cycle (TCA) metabolites, and glycolytic intermediates, were all reduced in the failing heart samples. This raises the possibility that these changes may reflect a state of malnutrition in end-stage disease and must be ruled out. Another difference between the human and animal studies is that HF patients often receive extensive treatment, which may impact the cardiac metabolism.
A better knowledge of cardiac metabolic adaptations in HF would certainly serve as a starting point from which to highlight new therapeutic targets, for old and new drugs, to exploit in different phenotypes and disease stages (Figure 1).
Figure 1. Myocardial metabolic modulation in dilated cardiomyopathy. This figure illustrates the different therapeutic metabolic targets in dilated cardiomyopathy. Abbreviations: SGLT2i, sodium-glucose cotransporter-2 inhibitors; BCAAs, branched-chain amino acids.

2.1. Betablockers

Without affecting their detrimental chronotropic and inotropic effects, betablockers have the ability to directly alter myocardial energetics [33]. By reducing peripheral lipolysis, this pharmacological class lowers the levels of FFA in the blood and allows a change in the heart’s energy metabolism that increases the use of carbohydrates [34]. Through these metabolic processes, betablockers are responsible for this substrate competition’s decreased myocardial FFA uptake and increased glucose utilization [35][36]. A decrease in FFA delivery and an increase in the availability of arterial glutamate, which is highly advantageous for myocardial tissue as it can serve as both aerobic and anaerobic fuel, making it a particularly versatile substrate [36], are both likely to be responsible for the increase in carbohydrate metabolism in the heart that was observed after a beta-blockade [37][38]. As a result, a beta-blockade may result in higher glucose consumption, which in turn may cause the heart to produce more energy without using up more oxygen. This suggests that in addition to their hemodynamic effects, betablockers may also directly affect the metabolic alterations observed in heart failure. When betablockers are used to treat systolic HF, greater energy efficiency and decreased oxygen consumption are seen. The changes in the way the heart produces energy may be the cause of these changes [39]. In actuality, the phenomenon of heart-rate reduction in HF patients may only be a marker of a greater functional response to betablocker therapy [40]. To lessen the failing heart’s reliance on fatty acids and to overcome the inhibition of myocardial glucose utilization brought on by fatty acids, the primary goal of therapy may be to reduce the plasma levels of FFAs and triacylglycerols. According to two studies, patients with New York Heart Association (NYHA) functional class III HF who took the betablocker carvedilol had lower FFA utilization and higher glucose utilization [41][42]. The reduction in oxygen consumption and the increase in energy effectiveness seen in HF patients after betablocker medication could possibly be explained by a change in the way the heart produces energy. In shifting the body’s energy substrate usage from lipid to glucose oxidation, non-selective betablockers appear to be more successful than selective ones [42]. Non-selective betablockers, on the other hand, appear to worsen insulin resistance, which is already known to be linked to HF and CV disorders [43][44]. However, it does not seem that vasodilators and cardio-selective drugs promote insulin resistance. Carvedilol, in particular, may have beneficial metabolic effects on boosting insulin sensitivity in HF patients [45]. Notably, the former’s greater metabolic effects may be one of the causes of the higher survival rates seen during their usage [46].

2.2. RAAS Inhibitors

The hormone system known as RAAS controls blood pressure and fluid balance. Angiotensin I (AT I) conversion to angiotensin II (AT II) is blocked by RAAS inhibitors, and the AT II receptors at the end of the route are also blocked. Due to its direct ability to cause and sustain ventricular dysfunction through a variety of pathways, AT II is a key participant in the regulation of cardiac energy metabolism [47]. It affects mitochondrial oxidative phosphorylation, particularly fatty acid oxidation [48], and damages cardiomyocyte mitochondria by increasing the formation of reactive oxygen species [49]. Additionally, there is proof that AT II reduces glucose oxidation [50]. Overall, AT II can lower ATP levels by reducing oxidative metabolism [51]. Its antagonism represents an appealing therapeutic strategy in this situation. Studies using the euglycemic insulin clamp technique demonstrated that the positive impact of AT II antagonistic action is exerted on insulin sensitivity. In fact, it has been demonstrated that ACE inhibitors [52] and angiotensin receptor antagonists [53] improve glucose homeostasis and left ventricular performance. The potential routes of action include elevated skeletal muscle blood flow, bradykinin build up, or more effective insulin release. Finally, RAAS inhibitors are able to reduce the atrial wall stress and fibrosis, consequently promoting a progressive reverse remodeling of the enlarged left atrium in dilated cardiomyopathy, with positive outcome effects [54].

2.3. Angiotensin Receptor Neprilysin Inhibitors (ARNI)

After a median follow-up of 27 months, the PARADIGM-HF study demonstrated that sacubitril/valsartan, the first member of a new class of medications known as ARNI, reduced the morbidity and mortality of patients with HF and reduced EF compared with the ACE inhibitor enalapril [55]. Sacubitril/valsartan is thought to provide an extra benefit over the renin–angiotensin blockade alone because it inhibits neprilysin, an endopeptidase that breaks down endogenous vasoactive peptides such natriuretic peptides [56]. Although there were very few patients who developed new-onset diabetes during the course of the PARADIGM-HF trial, sacubitril/valsartan did not lower the pre-specified exploratory outcome of new-onset diabetes, in contrast to enalapril. Despite this, new research suggests that sacubitril/valsartan, regardless of diabetes, may enhance lipid metabolism, insulin sensitivity, and glucose metabolism in individuals with HF [57][58].

2.4. Mineralocorticoid Receptor Antagonists (MRAs)

Aldosterone has negative effects that are mediated via the mineralocorticoid receptor; these effects are blocked by MRAs such as spironolactone and eplerenone. MRAs are therefore effective in treating hypertension, especially resistant hypertension, and in lowering the risk of morbidity and death in HF patients through this pharmacological activity. The “off-target effects” of spironolactone have also been shown to have negative effects on lipid and glucose homeostasis [59]. The blockage of glucocorticoid receptors by spironolactone is thought to be the mechanism by which cortisol blood concentrations are raised. By accelerating lipolysis and gluconeogenesis, the glucocorticoid cortisol raises blood glucose levels. On the other hand, the selective MRA eplerenone has relatively little effect on other steroid receptors. Because of this, it does not impact glucose metabolism, and it lowers serum cortisol levels [59]. It has been confirmed that spironolactone may cause changes in blood glucose levels, whereas eplerenone has no effect on glucose homeostasis, according to a recent systematic review of randomized controlled trials, prospectives, and observational studies evaluating the influence of the various MRAs on the biomarkers of glucose homeostasis in a variety of populations [60][61]. The Eplerenone in Mild Patients Hospitalization and Survival Study in Heart Failure (EMPHASIS-HF) study of participants with chronic HF actually had no effect on new-onset diabetes [62].

2.5. Loop Diuretics

Loop diuretics, such as furosemide, primarily affect the kidneys to increase urine production. However, they may also have an indirect effect on the heart’s metabolism. Hypomagnesemia and hypokalemia can be brought on by loop diuretics. Arrhythmias are more likely to occur in people with these two conditions, which can impair cardiac function by interfering with normal electrical signaling. Additionally, hypocalcemia, which may affect cardiac contractility, can be brought on by these medications. Last but not least, loop diuretics can contribute to all of the metabolic changes associated with RAAS activation (as previously described) by stimulating the RAAS through volume depletion and decreased blood pressure [63].

3. Direct Cardiac Metabolism Modulators

Although the ideal metabolic environment for a failing heart remains highly debated and poorly understood, it may be a good target for future treatments. With the use of medications such as trimetazidine, ranolazine, perhexiline, and etomoxir, the previous research has concentrated on boosting glucose oxidation, which, in comparison to FFA oxidation, has a higher P/O ratio. These medications have been shown to enhance myocardial energetics and LV function in patients with chronic ischemic and non-ischemic HF and low EF. For unknown reasons, these medications have not, however, ever undergone extensive clinical studies [64].
The failing heart in severe non-ischemic cardiomyopathy, on the other hand, may be metabolically adaptive and not oxygen-restricted, according to some preliminary investigations. This challenges the conventional wisdom that the optimal therapeutic goal is to increase glucose oxidation; instead, it supports the alternative position that increasing fatty acid oxidation is a wise course of action [64].

4. The Ketone Bodies Hypothesis

As outlined above, targeting cardiac metabolism by decreasing fatty acid oxidation and promoting glucose oxidation appears to be an interesting approach to the treatment of chronic HF. Several drugs have been investigated in small-scale studies, but large clinical trials are needed to confirm the efficacy of these agents as a part of chronic HF treatment. More recently, a new interest in ketone body metabolism has arisen as their modulation may be of potential benefit to HF patients. Under normal conditions, ketones represent a minimal part of all substrates utilized by the myocardium for energy production. These compounds, however, become critical during periods of stress and fasting since their utilization allows the preservation of glycogen stores. The myocardium is the highest ketone body consumer per unit mass. Ketone body oxidation is also more efficient than fatty acid oxidation in terms of ATP synthesis per molecule of oxygen used [65][66]. In addition, ketone body metabolism exerts anti-oxidant effects since it oxidizes mitochondrial co-enzyme Q and reduces cytosolic [NADP+]/[NADPH+], thereby decreasing free radical production [66]. The resourcefulness of the heart in using ketone bodies in order to satisfy its ATP requirements serves as a tool to spare glucose. However, it remains rather unclear whether their employment is compensative to balance out the negative effects of the failing heart adaptive/maladaptive substrate utilization. In hypertrophied and early-stage failing rat hearts, a reduced capacity to oxidize fatty acids and a shift to ketone oxidation as an alternative metabolic fuel have been observed [67]. Similar data have been found in failing human hearts: patients with reduced LV EF nearly tripled their consumptions of ketones as metabolic substrates compared to patients with preserved EF [27]. A case control study involving patients with chronic dilated non-ischemic cardiomyopathy showed increased amounts of beta-hydroxybutyryl CoA and decreased amounts of myocardial beta-hydroxybutyrate in myocardial tissue, suggesting an increased ketone body metabolism in this setting [68]. Additional studies have shown that circulating ketone bodies in subjects with chronic HF increase proportionally to the intensity of their symptoms, the level of congestion in the venous circulatory system, and the magnitude of neurohormonal and cytokine involvement, as well as the increasing deterioration of left ventricular function [69][70]. In this context, more ketones are produced through hepatic ketogenesis and become a fundamental substrate for energy production in cardiomyocytes [71]. These results clearly indicate that chronic HF determines a ketosis-prone state [69]. Indeed, exhaled acetone levels have been shown to be able to identify HF patients with a predictive value which is somewhat similar to that of brain natriuretic peptide (BNP); moreover, this predictive value is proportional to the NYHA class [70]. It is also known that exhaled breath acetone is increased in HF patients with reduced EF and is associated with higher mortality or heart transplantation [72]. Interestingly, higher serum levels of beta-hydroxybutyrate seem to relate to disease progression and adverse prognosis in arrhythmogenic cardiomyopathy patients, supporting the hypothesis that an enhanced ketone body metabolism may be a standard myocardium response to injuries [73]. According to another study, the cardiomyocytes’ specific loss of succinyl-CoA:3-oxoacid CoA transferase, which is involved in ketone body oxidation, is associated with significantly increased left ventricular volume and a decreased ejection fraction as a response to pressure overload [74]. Overall, these studies confirm the fundamental role of this metabolic pathway, showing that impaired ketone body oxidation may be associated with worsened heart remodeling following pressure overload. In this context, the concept of the therapeutic modulation of ketone metabolism as a potential new target in HF treatment is emerging [74][75][76][77][78].

5. Sodium-Glucose Cotransporter-2 Inhibitors (SGLT2i)

There is a bidirectional link between diabetes mellitus (DM) and HF. Longstanding diabetes causes changes in myocardial metabolism, abnormal calcium signaling, and inflammatory pathways, resulting in structural and functional changes in the myocardium and leading to the development of diabetic cardiomyopathy and the progression of HF [79][80]. Conversely, HF patients without DM are at an increased risk of developing glycemic abnormalities [79]. The shared underlying risk factors and the overlap of the pathophysiological mechanisms play a critical role in the frequent coexistence of DM and HF. As with HF, there is also a strong link between diabetes, coronary artery disease, hypertension, and renal disease. During the last decade, cardiovascular outcome trials have investigated several classes of new glucose-lowering agents, including SGLT2i, which, apart from showing evidence of cardiovascular safety, have also been shown to exert beneficial effects on the cardiovascular outcome [81][82]. Most studies have shown the independence of cardiovascular outcome from glycemic control, indicating mechanisms of action other than those usually postulated to explain the cardiovascular benefits of glucose-lowering therapies [83][84][85][86][87][88]. In fact, the significant beneficial clinical effects observed with SGLT2i use cannot be explained by one single mechanism.

References

  1. Elliott, P.; Andersson, B.; Arbustini, E.; Bilinska, Z.; Cecchi, F.; Charron, P.; Dubourg, O.; Kühl, U.; Maisch, B.; McKenna, W.J.; et al. Classification of the cardiomyopathies: A position statement from the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur. Heart J. 2008, 29, 270–276.
  2. Pinto, Y.M.; Elliott, P.M.; Arbustini, E.; Adler, Y.; Anastasakis, A.; Böhm, M.; Duboc, D.; Gimeno, J.; de Groote, P.; Imazio, M.; et al. Proposal for a revised definition of dilated cardiomyopathy, hypokinetic non-dilated cardiomyopathy, and its implications for clinical practice: A position statement of the ESC working group on myocardial and pericardial diseases. Eur. Heart J. 2016, 37, 1850–1858.
  3. Weintraub, R.G.; Semsarian, C.; Macdonald, P. Dilated cardiomyopathy. Lancet 2017, 390, 400–414.
  4. Reichart, D.; Magnussen, C.; Zeller, T.; Blankenberg, S. Dilated cardiomyopathy: From epidemiologic to genetic phenotypes: A translational review of current literature. J. Intern. Med. 2019, 286, 362–372.
  5. Lüscher, T.F. Heart failure: The cardiovascular epidemic of the 21st century. Eur. Heart J. 2015, 36, 395–397.
  6. Bleumink, G.S.; Knetsch, A.M.; Sturkenboom, M.C.; Straus, S.M.; Hofman, A.; Deckers, J.W.; Witteman, J.C.; Stricker, B.H. Quantifying the heart failure epidemic: Prevalence, incidence rate, lifetime risk and prognosis of heart failure The Rotterdam Study. Eur. Heart J. 2004, 25, 1614–1619.
  7. van Riet, E.E.; Hoes, A.W.; Wagenaar, K.P.; Limburg, A.; Landman, M.A.; Rutten, F.H. Epidemiology of heart failure: The prevalence of heart failure and ventricular dysfunction in older adults over time. A systematic review. Eur. J. Heart Fail. 2016, 18, 242–252.
  8. Urbich, M.; Globe, G.; Pantiri, K.; Heisen, M.; Bennison, C.; Wirtz, H.S.; Di Tanna, G.L. A Systematic Review of Medical Costs Associated with Heart Failure in the USA (2014–2020). Pharmacoeconomics 2020, 38, 1219–1236.
  9. Girerd, N.; Von Hunolstein, J.J.; Pellicori, P.; Bayés-Genís, A.; Jaarsma, T.; Lund, L.H.; Bilbault, P.; Boivin, J.M.; Chouihed, T.; Costa, J.; et al. Therapeutic inertia in the pharmacological management of heart failure with reduced ejection fraction. ESC Heart Fail. 2022, 9, 2063–2069.
  10. Lopaschuk, G.D.; Ussher, J.R.; Folmes, C.D.; Jaswal, J.S.; Stanley, W.C. Myocardial fatty acid metabolism in health and disease. Physiol. Rev. 2010, 90, 207–258.
  11. Ingwall, J.S. Energy metabolism in heart failure and remodelling. Cardiovasc. Res. 2009, 81, 412–419.
  12. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Böhm, M.; Burri, H.; Butler, J.; Čelutkienė, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 2021, 42, 3599–3726.
  13. Heidenreich, P.A.; Bozkurt, B.; Aguilar, D.; Allen, L.A.; Byun, J.J.; Colvin, M.M.; Deswal, A.; Drazner, M.H.; Dunlay, S.M.; Evers, L.R.; et al. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation 2022, 145, e895–e1032.
  14. Jones, N.R.; Roalfe, A.K.; Adoki, I.; Hobbs, F.D.R.; Taylor, C.J. Survival of patients with chronic heart failure in the community: A systematic review and meta-analysis. Eur. J. Heart Fail. 2019, 21, 1306–1325.
  15. Lopaschuk, G.D.; Karwi, Q.G.; Tian, R.; Wende, A.R.; Abel, E.D. Cardiac Energy Metabolism in Heart Failure. Circ. Res. 2021, 128, 1487–1513.
  16. Stanley, W.C.; Recchia, F.A.; Lopaschuk, G.D. Myocardial substrate metabolism in the normal and failing heart. Physiol. Rev. 2005, 85, 1093–1129.
  17. Karwi, Q.G.; Uddin, G.M.; Ho, K.L.; Lopaschuk, G.D. Loss of Metabolic Flexibility in the Failing Heart. Front. Cardiovasc. Med. 2018, 5, 68.
  18. Neubauer, S. The failing heart--an engine out of fuel. N. Engl. J. Med. 2007, 356, 1140–1151.
  19. Zhou, B.; Tian, R. Mitochondrial dysfunction in pathophysiology of heart failure. J. Clin. Investig. 2018, 128, 3716–3726.
  20. Tian, R.; Colucci, W.S.; Arany, Z.; Bachschmid, M.M.; Ballinger, S.W.; Boudina, S.; Bruce, J.E.; Busija, D.W.; Dikalov, S.; Dorn, G.W., II; et al. Unlocking the Secrets of Mitochondria in the Cardiovascular System: Path to a Cure in Heart Failure—A Report. from the 2018 National Heart, Lung, and Blood Institute Workshop. Circulation 2019, 140, 1205–1216.
  21. Allard, M.F.; Schönekess, B.O.; Henning, S.L.; English, D.R.; Lopaschuk, G.D. Contribution of oxidative metabolism and glycolysis to ATP production in hypertrophied hearts. Am. J. Physiol. 1994, 267 Pt 2, H742–H750.
  22. Doenst, T.; Nguyen, T.D.; Abel, E.D. Cardiac metabolism in heart failure: Implications beyond ATP production. Circ. Res. 2013, 113, 709–724.
  23. Despa, S.; Bers, D.M. Na⁺ transport in the normal and failing heart—Remember the balance. J. Mol. Cell. Cardiol. 2013, 61, 2–10.
  24. Barger, P.M.; Kelly, D.P. Fatty acid utilization in the hypertrophied and failing heart: Molecular regulatory mechanisms. Am. J. Med. Sci. 1999, 318, 36–42.
  25. Funada, J.; Betts, T.R.; Hodson, L.; Humphreys, S.M.; Timperley, J.; Frayn, K.N.; Karpe, F. Substrate utilization by the failing human heart by direct quantification using arterio-venous blood sampling. PLoS ONE 2009, 4, e7533.
  26. Voros, G.; Ector, J.; Garweg, C.; Droogne, W.; Van Cleemput, J.; Peersman, N.; Vermeersch, P.; Janssens, S. Increased Cardiac Uptake of Ketone Bodies and Free Fatty Acids in Human Heart Failure and Hypertrophic Left Ventricular Remodeling. Circ. Heart Fail. 2018, 11, e004953.
  27. Murashige, D.; Jang, C.; Neinast, M.; Edwards, J.J.; Cowan, A.; Hyman, M.C.; Rabinowitz, J.D.; Frankel, D.S.; Arany, Z. Comprehensive quantification of fuel use by the failing and nonfailing human heart. Science 2020, 370, 364–368.
  28. Chen, L.; Song, J.; Hu, S. Metabolic remodeling of substrate utilization during heart failure progression. Heart Fail. Rev. 2019, 24, 143–154.
  29. Larsen, T.S.; Jansen, K.M. Impact of Obesity-Related Inflammation on Cardiac Metabolism and Function. J. Lipid Atheroscler. 2021, 10, 8–23.
  30. Raghow, R. An ‘Omics’ Perspective on Cardiomyopathies and Heart Failure. Trends Mol. Med. 2016, 22, 813–827.
  31. Ampong, I. Metabolic and Metabolomics Insights into Dilated Cardiomyopathy. Ann. Nutr. Metab. 2022, 78, 147–155.
  32. Flam, E.; Jang, C.; Murashige, D.; Yang, Y.; Morley, M.P.; Jung, S.; Kantner, D.S.; Pepper, H.; Bedi, K.C., Jr.; Brandimarto, J.; et al. Integrated landscape of cardiac metabolism in end-stage human nonischemic dilated cardiomyopathy. Nat. Cardiovasc. Res. 2022, 1, 817–829.
  33. Thompson, D.S.; Naqvi, N.; Juul, S.M.; Coltart, D.J.; Jenkins, B.S.; Webb-Peploe, M.M. Haemodynamic and metabolic effects of atenolol in patients with angina pectoris. Br. Heart J. 1980, 43, 668–679.
  34. Day, J.L. The metabolic consequences of adrenergic blockade: A review. Metabolism 1975, 24, 987–996.
  35. Lech, J.J.; Jesmok, G.J.; Calvert, D.N. Effects of drugs and hormones on lipolysis in heart. Fed. Proc. 1977, 36, 2000–2008.
  36. Simonsen, S.; Ihlen, H.; Kjekshus, J.K. Haemodynamic and metabolic effects of timolol (Blocadren) on ischaemic myocardium. Acta Med. Scand. 1983, 213, 393–398.
  37. Nielsen, T.T.; Bagger, J.P.; Thomassen, A. Improved myocardial lactate extraction after propranolol in coronary artery disease: Effected by peripheral glutamate and free fatty acid metabolism. Br. Heart J. 1986, 55, 140–147.
  38. Pisarenko, O.I.; Solomatina, E.S.; Studneva, I.M.; Ivanov, V.E.; Kapelko, V.I.; Smirnov, V.N. Protective effect of glutamic acid on cardiac function and metabolism during cardioplegia and reperfusion. Basic. Res. Cardiol. 1983, 78, 534–543.
  39. Spoladore, R.; Fragasso, G.; Perseghin, G.; De Cobelli, F.; Esposito, A.; Maranta, F.; Calori, G.; Locatelli, M.; Lattuada, G.; Scifo, P.; et al. Beneficial effects of beta-blockers on left ventricular function and cellular energy reserve in patients with heart failure. Fundam. Clin. Pharmacol. 2013, 27, 455–464.
  40. Fragasso, G.; Salerno, A.; Margonato, A. Heart rate reduction is probably not the main beneficial mechanism by which beta blockade improves outcome in patients with systolic chronic heart failure. Am. J. Cardiol. 2008, 102, 506–507.
  41. Al-Hesayen, A.; Azevedo, E.R.; Floras, J.S.; Hollingshead, S.; Lopaschuk, G.D.; Parker, J.D. Selective versus nonselective beta-adrenergic receptor blockade in chronic heart failure: Differential effects on myocardial energy substrate utilization. Eur. J. Heart Fail. 2005, 7, 618–623.
  42. Podbregar, M.; Voga, G. Effect of selective and nonselective beta-blockers on resting energy production rate and total body substrate utilization in chronic heart failure. J. Card. Fail. 2002, 8, 369–378.
  43. Reneland, R.; Alvarez, E.; Andersson, P.E.; Haenni, A.; Byberg, L.; Lithell, H. Induction of insulin resistance by beta-blockade but not ACE-inhibition: Long-term treatment with atenolol or trandolapril. J. Hum. Hypertens. 2000, 14, 175–180.
  44. Paolisso, G.; De Riu, S.; Marrazzo, G.; Verza, M.; Varricchio, M.; D’Onofrio, F. Insulin resistance and hyperinsulinemia in patients with chronic congestive heart failure. Metabolism 1991, 40, 972–977.
  45. Ferrua, S.; Bobbio, M.; Catalano, E.; Grassi, G.; Massobrio, N.; Pinach, S.; Rossi, C.; Veglio, M.; Trevi, G.P. Does carvedilol impair insulin sensitivity in heart failure patients without diabetes? J. Card. Fail. 2005, 11, 590–594.
  46. Poole-Wilson, P.A.; Swedberg, K.; Cleland, J.G.; Di Lenarda, A.; Hanrath, P.; Komajda, M.; Lubsen, J.; Lutiger, B.; Metra, M.; Remme, W.J. Carvedilol or Metoprolol European Trial Investigators. Comparison of carvedilol and metoprolol on clinical outcomes in patients with chronic heart failure in the Carvedilol or Metoprolol European Trial (COMET): Randomised controlled trial. Lancet 2003, 362, 7–13.
  47. Mori, J.; Basu, R.; McLean, B.A.; Das, S.K.; Zhang, L.; Patel, V.B.; Wagg, C.S.; Kassiri, Z.; Lopaschuk, G.D.; Oudit, G.Y. Agonist-induced hypertrophy and diastolic dysfunction are associated with selective reduction in glucose oxidation: A metabolic contribution to heart failure with normal ejection fraction. Circ. Heart Fail. 2012, 5, 493–503.
  48. Dai, D.F.; Johnson, S.C.; Villarin, J.J.; Chin, M.T.; Nieves-Cintrón, M.; Chen, T.; Marcinek, D.J.; Dorn, G.W., 2nd; Kang, Y.J.; Prolla, T.A.; et al. Mitochondrial oxidative stress mediates angiotensin II-induced cardiac hypertrophy and Galphaq overexpression-induced heart failure. Circ. Res. 2011, 108, 837–846.
  49. Pellieux, C.; Aasum, E.; Larsen, T.S.; Montessuit, C.; Papageorgiou, I.; Pedrazzini, T.; Lerch, R. Overexpression of angiotensinogen in the myocardium induces downregulation of the fatty acid oxidation pathway. J. Mol. Cell. Cardiol. 2006, 41, 459–466.
  50. Mori, J.; Alrob, O.A.; Wagg, C.S.; Harris, R.A.; Lopaschuk, G.D.; Oudit., G.Y. ANG II causes insulin resistance and induces cardiac metabolic switch and inefficiency: A critical role of PDK4. Am. J. Physiol. Heart Circ. Physiol. 2013, 304, H1103–H1113.
  51. Fillmore, N.; Mori, J.; Lopaschuk, G.D. Mitochondrial fatty acid oxidation alterations in heart failure, ischaemic heart disease and diabetic cardiomyopathy. Br. J. Pharmacol. 2014, 171, 2080–2090.
  52. Vermes, E.; Ducharme, A.; Bourassa, M.G.; Lessard, M.; White, M.; Tardif, J.C. Enalapril reduces the incidence of diabetes in patients with chronic heart failure: Insight from the Studies of Left Ventricular Dysfunction (SOLVD). Circulation 2003, 107, 1291–1296.
  53. Yusuf, S.; Ostergren, J.B.; Gerstein, H.C.; Pfeffer, M.A.; Swedberg, K.; Granger, C.B.; Olofsson, B.; Probstfield, J.; McMurray, J.V. Candesartan in Heart Failure-Assessment of Reduction in Mortality and Morbidity Program Investigators. Effects of candesartan on the development of a new diagnosis of diabetes mellitus in patients with heart failure. Circulation 2005, 112, 48–53.
  54. Nuzzi, V.; Raafs, A.; Manca, P.; Henkens, M.T.; Gregorio, C.; Boscutti, A.; Verdonschot, J.; Hazebroek, M.; Knackstedt, C.; Merlo, M.; et al. Left Atrial Reverse Remodeling in Dilated Cardiomyopathy. J. Am. Soc. Echocardiogr. 2023, 36, 154–162.
  55. McMurray, J.J.; Packer, M.; Desai, A.S.; Gong, J.; Lefkowitz, M.P.; Rizkala, A.R.; Rouleau, J.L.; Shi, V.C.; Solomon, S.D.; Swedberg, K.; et al. PARADIGM-HF Investigators and Committees. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N. Engl. J. Med. 2014, 371, 993–1004.
  56. Bayes-Genis, A.; Barallat, J.; Richards, A.M. A Test in Context: Neprilysin: Function, Inhibition, and Biomarker. J. Am. Coll. Cardiol. 2016, 68, 639–653.
  57. Seferovic, J.P.; Claggett, B.; Seidelmann, S.B.; Seely, E.W.; Packer, M.; Zile, M.R.; Rouleau, J.L.; Swedberg, K.; Lefkowitz, M.; Shi, V.C.; et al. Effect of sacubitril/valsartan versus enalapril on glycaemic control in patients with heart failure and diabetes: A post-hoc analysis from the PARADIGM-HF trial. Lancet Diabetes Endocrinol. 2017, 5, 333–340.
  58. Armentaro, G.; D’Arrigo, G.; Miceli, S.; Cassano, V.; Perticone, M.; Maio, R.; Marra, A.M.; Arturi, F.; Cittadini, A.; Tripepi, G.; et al. Long Term Metabolic Effects of Sacubitril/Valsartan in Non-Diabetic and Diabetic Patients with Heart Failure Reduced Ejection Fraction: A Real Life Study. Front. Physiol. 2022, 13, 897109.
  59. Falch, D.K.; Schreiner, A. The effect of spironolactone on lipid, glucose and uric acid levels in blood during long-term administration to hypertensives. Acta Med. Scand. 1983, 213, 27–30.
  60. Yamaji, M.; Tsutamoto, T.; Kawahara, C.; Nishiyama, K.; Yamamoto, T.; Fujii, M.; Horie, M. Effect of eplerenone versus spironolactone on cortisol and hemoglobin A₁(c) levels in patients with chronic heart failure. Am. Heart J. 2010, 160, 915–921.
  61. Korol, S.; Mottet, F.; Perreault, S.; Baker, W.L.; White, M.; de Denus, S. A systematic review and meta-analysis of the impact of mineralocorticoid receptor antagonists on glucose homeostasis. Medicine 2017, 96, e8719.
  62. Preiss, D.; van Veldhuisen, D.J.; Sattar, N.; Krum, H.; Swedberg, K.; Shi, H.; Vincent, J.; Pocock, S.J.; Pitt, B.; Zannad, F.; et al. Eplerenone and new-onset diabetes in patients with mild heart failure: Results from the Eplerenone in Mild Patients Hospitalization and Survival Study in Heart Failure (EMPHASIS-HF). Eur. J. Heart Fail. 2012, 14, 909–915.
  63. Fragasso, G.; Margonato, A.; Spoladore, R.; Lopashuck, G.D. Metabolic effects of cardiovascular drugs. Trends Cardiovasc. Med. 2019, 29, 176–187.
  64. Fragasso, G. Deranged Cardiac Metabolism and the Pathogenesis of Heart Failure. Card. Fail. Rev. 2016, 2, 8–13.
  65. Puchalska, P.; Crawford, P.A. Multi-dimensional Roles of Ketone Bodies in Fuel Metabolism, Signaling, and Therapeutics. Cell. Metab. 2017, 25, 262–284.
  66. Veech, R.L. The therapeutic implications of ketone bodies: The effects of ketone bodies in pathological conditions: Ketosis, ketogenic diet, redox states, insulin resistance, and mitochondrial metabolism. Prostaglandins Leukot. Essent. Fatty Acids. 2004, 70, 309–319.
  67. Aubert, G.; Martin, O.J.; Horton, J.L.; Lai, L.; Vega, R.B.; Leone, T.C.; Koves, T.; Gardell, S.J.; Krüger, M.; Hoppel, C.L.; et al. The Failing Heart Relies on Ketone Bodies as a Fuel. Circulation 2016, 133, 698–705.
  68. Bedi, K.C., Jr.; Snyder, N.W.; Brandimarto, J.; Aziz, M.; Mesaros, C.; Worth, A.J.; Wang, L.L.; Javaheri, A.; Blair, I.A.; Margulies, K.B. Evidence for Intramyocardial Disruption of Lipid Metabolism and Increased Myocardial Ketone Utilization in Advanced Human Heart Failure. Circulation 2016, 133, 706–716.
  69. Lommi, J.; Kupari, M.; Koskinen, P.; Näveri, H.; Leinonen, H.; Pulkki, K.; Härkönen, M. Blood ketone bodies in congestive heart failure. J. Am. Coll. Cardiol. 1996, 28, 665–672.
  70. Marcondes-Braga, F.G.; Gutz, I.G.R.; Batista, G.L.; Saldiva, P.H.N.; Ayub-Ferreira, S.M.; Issa, V.S.; Mangini, S.; Bocchi, E.A.; Bacal, F. Exhaled acetone as a new biomaker of heart failure severity. Chest 2012, 142, 457–466.
  71. Schugar, R.C.; Moll, A.R.; André d’Avignon, D.; Weinheimer, C.J.; Kovacs, A.; Crawford, P.A. Cardiomyocyte-specific deficiency of ketone body metabolism promotes accelerated pathological remodeling. Mol. Metab. 2014, 3, 754–769.
  72. Marcondes-Braga, F.G.; Batista, G.L.; Gutz, I.G.; Saldiva, P.H.; Mangini, S.; Issa, V.S.; Ayub-Ferreira, S.M.; Bocchi, E.A.; Pereira, A.C.; Bacal, F. Impact of Exhaled Breath Acetone in the Prognosis of Patients with Heart Failure with Reduced Ejection Fraction (HFrEF). One Year of Clinical Follow-up. PLoS ONE 2016, 11, e0168790.
  73. Song, J.P.; Chen, L.; Chen, X.; Ren, J.; Zhang, N.N.; Tirasawasdichai, T.; Hu, Z.L.; Hua, W.; Hu, Y.R.; Tang, H.R.; et al. Elevated plasma β-hydroxybutyrate predicts adverse outcomes and disease progression in patients with arrhythmogenic cardiomyopathy. Sci. Transl. Med. 2020, 12, eaay8329.
  74. Horton, J.L.; Davidson, M.T.; Kurishima, C.; Vega, R.B.; Powers, J.C.; Matsuura, T.R.; Petucci, C.; Lewandowski, E.D.; Crawford, P.A.; Muoio, D.M.; et al. The failing heart utilizes 3-hydroxybutyrate as a metabolic stress defense. JCI Insight 2019, 4, e124079.
  75. Nielsen, R.; Møller, N.; Gormsen, L.C.; Tolbod, L.P.; Hansson, N.H.; Sorensen, J.; Harms, H.J.; Frøkiær, J.; Eiskjaer, H.; Jespersen, N.R.; et al. Cardiovascular Effects of Treatment with the Ketone Body 3-Hydroxybutyrate in Chronic Heart Failure Patients. Circulation 2019, 139, 2129–2141.
  76. Yurista, S.R.; Silljé, H.H.W.; Oberdorf-Maass, S.U.; Schouten, E.M.; Pavez Giani, M.G.; Hillebrands, J.L.; van Goor, H.; van Veldhuisen, D.J.; de Boer, R.A.; Westenbrink, B.D. Sodium-glucose co-transporter 2 inhibition with empagliflozin improves cardiac function in non-diabetic rats with left ventricular dysfunction after myocardial infarction. Eur. J. Heart Fail. 2019, 21, 862–873.
  77. Yurista, S.R.; Chong, C.R.; Badimon, J.J.; Kelly, D.P.; de Boer, R.A.; Westenbrink, B.D. Therapeutic Potential of Ketone Bodies for Patients with Cardiovascular Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2021, 77, 1660–1669.
  78. Monzo, L.; Sedlacek, K.; Hromanikova, K.; Tomanova, L.; Borlaug, B.A.; Jabor, A.; Kautzner, J.; Melenovsky, V. Myocardial ketone body utilization in patients with heart failure: The impact of oral ketone ester. Metabolism 2021, 115, 154452.
  79. Oktay, A.A.; Aktürk, H.K.; Paul, T.K.; O’Keefe, J.H.; Ventura, H.O.; Koch, C.A.; Lavie, C.J.; Feingold, K.R.; Anawalt, B.; Boyce, A.; et al. Diabetes, Cardiomyopathy, and Heart Failure. In Endotext ; MDText.com, Inc.: South Dartmouth, MA, USA, 2000.
  80. Li, N.; Zhou, H. SGLT2 Inhibitors: A Novel Player in the Treatment and Prevention of Diabetic Cardiomyopathy. Drug. Des. Dev. Ther. 2020, 14, 4775–4788.
  81. Ahmad, Y.; Madhavan, M.V.; Stone, G.W.; Francis, D.P.; Makkar, R.; Bhatt, D.L.; Howard, J.P. Sodium-glucose cotransporter 2 inhibitors in patients with heart failure: A systematic review and meta-analysis of randomized trials. Eur. Heart J. Qual. Care Clin. Outcomes 2022, 8, 383–390.
  82. Zannad, F.; Ferreira, J.P.; Pocock, S.J.; Anker, S.D.; Butler, J.; Filippatos, G.; Brueckmann, M.; Ofstad, A.P.; Pfarr, E.; Jamal, W.; et al. SGLT2 inhibitors in patients with heart failure with reduced ejection fraction: A meta-analysis of the EMPEROR-Reduced and DAPA-HF trials. Lancet 2020, 396, 819–829.
  83. Giugliano, D.; Longo, M.; Scappaticcio, L.; Bellastella, G.; Maiorino, M.I.; Esposito, K. SGLT-2 inhibitors and cardiorenal outcomes in patients with or without type 2 diabetes: A meta-analysis of 11 CVOTs. Cardiovasc. Diabetol. 2021, 20, 236.
  84. Inzucchi, S.E.; Kosiborod, M.; Fitchett, D.; Wanner, C.; Hehnke, U.; Kaspers, S.; George, J.T.; Zinman, B. Improvement in Cardiovascular Outcomes with Empagliflozin Is Independent of Glycemic Control. Circulation 2018, 138, 1904–1907.
  85. Petrie, M.C.; Verma, S.; Docherty, K.F.; Inzucchi, S.E.; Anand, I.; Belohlávek, J.; Böhm, M.; Chiang, C.E.; Chopra, V.K.; de Boer, R.A.; et al. Effect of Dapagliflozin on Worsening Heart Failure and Cardiovascular Death in Patients with Heart Failure with and Without Diabetes. JAMA 2020, 323, 1353–1368.
  86. Williams, D.M.; Evans, M. Are SGLT-2 Inhibitors the Future of Heart Failure Treatment? The EMPEROR-Preserved and EMPEROR-Reduced Trials. Diabetes Ther. 2020, 11, 1925–1934.
  87. Zinman, B.; Wanner, C.; Lachin, J.M.; Fitchett, D.; Bluhmki, E.; Hantel, S.; Mattheus, M.; Devins, T.; Johansen, O.E.; Woerle, H.J.; et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N. Engl. J. Med. 2015, 373, 2117–2128.
  88. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Zannad, F. Effects of Sodium-Glucose Cotransporter 2 Inhibitors for the Treatment of Patients with Heart Failure: Proposal of a Novel Mechanism of Action. JAMA Cardiol. 2017, 2, 1025–1029.
More
Video Production Service