Acessibilidade / Reportar erro

Driving force of deteriorated cellular environment in heart failure: Metabolic remodeling

Abstract

Heart Failure (HF) has been one of the leading causes of death worldwide. Though its latent mechanism and therapeutic manipulation are updated and developed ceaselessly, there remain great gaps in the cognition of heart failure. High morbidity and readmission rates among HF patients are waiting to be addressed. Recent studies have found that myocardial energy metabolism was closely related to heart failure, in which substrate utilization, as well as intermediate metabolism disorders, insulin resistance, oxidative stress, and mitochondrial dysfunction, might underlie systolic dysfunction and progression of HF. This article centers on the changes and counteraction of cardiac energy metabolism in the failing heart. Therefore, targeting impaired energy provision is of great potential in the treatment of HF. And shifting the objective from traditional neurohormones to improving the cellular environment is expected to further optimize the management of HF.

Keywords
Heart failure; Physiological metabolism; Metabolic remodeling; Deteriorated cellular environment; Pharmacological agents; Stem cells

Highlights

  • Improving the cellular environment is expected to further optimize the management of HF.

  • Metabolic remodeling is the driving force of deteriorated cellular environment in HF.

  • Targeting impaired energy provision is of great potential in the treatment of HF.

Highlights

  • Improving the cellular environment is expected to further optimize the management of HF.

  • Metabolic remodeling is the driving force of deteriorated cellular environment in HF.

  • Targeting impaired energy provision is of great potential in the treatment of HF.

Introduction

HF is characterized by impaired ventricular filling and ejection function, which is the ultimate destination of numerous cardiovascular diseases [1]1 Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JG, Coats AJ, et al.; Authors/Task Force Members; document reviewers. 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 J Heart Fail. 2016;18(8):891-975.. It is estimated that the prevalence of HF in adults was about 1%‒2%, with over 13.7 million people suffering from HF worldwide. Thus, HF has brought about severe public health issues and financial burdens [22 Groenewegen A, Rutten FH, Mosterd A, Hoes AW Epidemiology of heart failure. Eur J Heart Fail. 2020;22(8):1342-56.,33 Hao G, Wang X, Chen Z, Zhang L, Zhang Y, Wei B, et al.; China Hypertension Survey Investigators. Prevalence of heart failure and left ventricular dysfunction in China: the China Hypertension Survey, 2012-2015. Eur J Heart Fail. 2019;21(11):1329-37.]. With the aging of the population, there is an upward trend in the incidence of chronic diseases at an earlier age, such as hypertension, Diabetes Mellitus (DM), and obesity, and the age of onset tends to be younger. The prompt precaution and management of cardiovascular diseases prolong the lifetime of patients, which leads to the great number of HF patients [44 GBD 2017 Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2018;392(10159):1789-858.,55 Heart Failure Group of Chinses Society of Cardiology of Chinses Medical Association, Chinses Heart Failure Association of Chinses Medical Doctor Association, Editorial Board of Chinses Journar of Cardioloy. 2018 China Guidelines for the diagnosis and treatment of heart failure. Chin J Cardiol. 2018; 2018(10) [in Chinese].]. In addition, the high death and all-cause hospitalization rates remain the greatest threat to HF patients. The research reported that the readmission rates within 30 days amounted to 24.8%, and 1-year, 5-year, and 10-year survival rates were 86.5%, 56.7% and 34.9%, respectively [66 Dharmarajan K, Hsieh AF, Lin Z, Bueno H, Ross JS, Horwitz LI, et al. Diagnoses and timing of 30-day readmissions after hospitalization for heart failure, acute myocardial infarction, or pneumonia. JAMA. 2013;309(4):355-63.,77 Jones NR, Roalfe AK, Adoki I, Hobbs FDR, Taylor CJ Survival of patients with chronic heart failure in the community: a systematic review and meta-analysis. Eur J Heart Fail. 2019;21(11):1306-25.]. The abnormal structure and function lead to disorders in blood circulation, appearing low cardiac output and fluid retention whose severity is consistent with symptoms and signs of HF. There is a general belief that myocardial remodeling is an adaptive change to hemodynamic abnormality. And the activation of the sympathetic nervous system as well as inflammatory response play a pivotal role that aggravates the development of ventricular remodeling [8]8 Burchfield JS, Xie M, Hill JA Pathological ventricular remodeling: mechanisms: part 1 of 2. Circulation. 2013;128(4):388-400.. Recent studies show that marked alterations of energy metabolism in the failing heart were going on. The concomitant insulin resistance, lipotoxicity, oxidative stress, and imbalance of calcium homeostasis worsen the cellular environment and drive the progression of the failing heart. Above all, targeting cardiac metabolism to optimize the cellular environment provides new possibilities for the treatment and management of HF.

The physiological role of energy metabolism in the normal heart

The heart is a high-energy-consuming organ, accounting for approximately 12% oxygen consumption of the whole body [9]9 Stanley WC, Recchia FA, Lopaschuk GD Myocardial substrate metabolism in the normal and failing heart. Physiol Rev. 2005;85(3):1093-129.. And the heart never fails to utilize a range of substrates at hand for the sake of acclimatizing the changing environment. But very often, Fatty Acid Oxidation (FAO), meeting about 60% of the cardiac demand, is the main energy provision, and the rest suppliers include glucose, Ketone Bodies (KBs) and amino acids [10]10 Glatz JFC, Nabben M, Young ME, Schulze PC, Taegtmeyer H, Luiken JJFP Re-balancing cellular energy substrate metabolism to mend the failing heart. Biochim Biophys Acta Mol Basis Dis. 2020;1866(5):165579.. The fatty acid requires two transmembrane movements before oxidation in the mitochondria, and the membrane proteins involved in this process include Fatty-Acid Translocase (FAT/CD36), plasma membrane-associated Fatty-Acid Binding Protein (FABPpm), and Fatty-Acid Transport Proteins (FATP) [11]11 Glatz JFC, Luiken JJFP Dynamic role of the transmembrane glycoprotein CD36 (SR-B2) in cellular fatty acid uptake and utilization. J Lipid Res. 2018;59(7):1084-93.. Among them, it was observed in CD36(-/-) mice that fatty acid uptake mediated by CD36 accounted for 70% of total intake [12]12 Habets DD, Coumans WA, Voshol PJ, den Boer MA, Febbraio M, Bonen A, et al. AMPK-mediated increase in myocardial long-chain fatty acid uptake critically depends on sarcolemmal CD36. Biochem Biophys Res Commun. 2007;355(1):204-10.. Peroxisome-Proliferator-Activated Receptor (PPAR), a ligand-activated nuclear transcription factor, positively regulates FAO involving uptake (FAT/CD36), storage (FABP), and Transport (CPT-1) in cardiomyocytes [1313 Kalinowska A, Górski J, Harasim E, Harasiuk D, Bonen A, Chabowski A Differential effects of chronic, in vivo, PPAR's stimulation on the myocardial subcellular redistribution of FAT/CD36 and FABPpm. FEBS Lett. 2009;583(15):2527-34.,1414 Smeets PJ, Teunissen BE, Willemsen PH, van Nieuwenhoven FA, Brouns AE, Janssen BJ, et al. Cardiac hypertrophy is enhanced in PPAR alpha-/- mice in response to chronic pressure overload. Cardiovasc Res. 2008;78(1):79-89.]. A recent study found that fatty acids excited the acetylation of CREB-binding protein-dependent Ovarian-Tumor-Domain-containing Deubiquitinase 3 (OTUD3) which adjusted relative genes referring to metabolism and oxidative phosphorylation by stabilizing PPARδ [15]15 Zhou N, Qi H, Liu J, Zhang G, Liu J, Liu N, et al. Deubiquitinase OTUD3 regulates metabolism homeostasis in response to nutritional stresses. Cell Metab. 2022;34(7):1023-1041.e8..

The uptake of cardiac glucose is determined by the concentration of glucose, the number and intrinsic activity of Glucose Transporter 4 (GLUT4) [16]16 Shao D, Tian R Glucose transporters in cardiac metabolism and hypertrophy. Compr Physiol. 2015;6(1):331-51.. Glucose is phosphorylated to Glucose 6-Phosphate (G6P) by hexokinase, and glycolysis is the preferred destination for G6P [17]17 Badolia R, Ramadurai DKA, Abel ED, Ferrin P, Taleb I, Shankar TS, et al. The role of nonglycolytic glucose metabolism in myocardial recovery upon mechanical unloading and circulatory support in chronic heart failure. Circulation. 2020;142(3):259-74.. The coupling between glycolysis and ion transport ensures the optimal transmission of energy between ion channels and transporters in cellular activities, which play a vital role in maintaining the systolic function, though glycolysis does not contribute much in terms of energy production [18]18 Dhar-Chowdhury P, Malester B, Rajacic P, Coetzee WA The regulation of ion channels and transporters by glycolytically derived ATP. Cell Mol Life Sci. 2007;64(23):3069-83.. Glucose and fatty acids regulate each other in a competitive way to maintain metabolic homeostasis, namely the Randle cycle. It is generally believed that pyruvate dehydrogenase complex and acetyl-CoA carboxylase are connected with the regulation of the Acetyl-CoA which is the coincidence and competition point of metabolism [19]19 Park S, Jeon JH, Min BK, Ha CM, Thoudam T, Park BY, Lee IK Role of the pyruvate dehydrogenase complex in metabolic remodeling: differential pyruvate dehydrogenase complex functions in metabolism. Diabetes Metab J. 2018;42(4):270-81..

KBs include Acetylacetic Acid (AcAc), β-Hydroxybutyrate (β-HB), and acetone. KBs provide only about 5% needs of the whole body, and the contribution of KBs can rise up to 20% during fasting [20]20 Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, Smith A, et al. Nutritional ketosis alters fuel preference and thereby endurance performance in athletes. Cell Metab. 2016;24(2):256-68.. Ketone bodies, the intermediate of β-oxidation, are mainly synthesized in the mitochondria of the liver via 3-Hydroxy-3-Methylglutaryl-CoA (HMG-CoA) ketogenesis pathway. Ketone bodies are metabolized extrahepatic as a transport form of acetyl-CoA, catalyzing the exchange of CoA between succinic acid and AcAc. Besides, KBs also act as a signal transducer factor to regulate oxidative stress and the post-translational modification of protein [21]21 Puchalska P, Crawford PA Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 2017;25(2):262-84. (Fig. 1).

Fig. 1
The energy metabolism in the normal heart. Fatty acid oxidation occupies center stage in energy provision, and the rest suppliers include glucose and ketone bodies. KBs can only be metabolized outside the liver as the liver is short of crucial enzyme SCOT. AcAc, Acetylacetic Acid; BDH1, 3-Hydroxy-n-Butyrate Dehydrogenase; β-OHB, Beta-Hydroxybutyrate; CPT1/2, Carnitine Palmitoyltransferase-1/2; FACS, Fatty Acyl CoA Synthetase; FAT, Fatty Acid Translocase (also known as CD36); FFA, Free Fatty Acid; GLUT1/4, Glucose Transporter 1/4; G6P, Glucose 6-Phosphate; HBP, Hexosamine Biosynthesis Pathway; HMGCL, 3-Hydroxy-3-Methylglutaryl-CoA Lyase; HMG-CoA, 3-Hydroxy-3-Methylglutaryl-CoA; HMGCS2, 3-Hydroxy-3-Methylglutaryl-CoA Synthetase 2; MCT, Medium-Chain Triacylglycerol; MPC, Mitochondrial Pyruvate Carrier; PFK, 6-Phosphofructokinase; PPP, Pentose Phosphate Pathway; SCOT, Succinyl CoA: 3-oxoacid-CoA-Transferase; TAG, Triacylglyceride; TCA cycle, Tricarboxylic Acid; VLDL, Very Low-Density Lipoprotein.

The pathologic change of energy in the failing heart

The process of HF is accompanied by poor energy production and metabolic remodeling. Metabolic remodeling involves increased neurohormonal stimulation and impaired calcium processing that exacerbate energy expenditure. These changes disrupt the balance between energy supply and demand, challenging the cell environment and promoting cardiomyopathy.

Disorders of cardiac metabolism in the failing heart

Variations in the utilization of the fatty acid

Metabolic changes seem complex in failing hearts, not only depend on the severity and type of HF, but also highly correlated with comorbidities, such as obesity and Type 2 Diabetes (T2D). Although the exact metabolic changes and substrate preferences in HF remain controversial, current studies believe that FAO is unchanged or slightly increased in the early stage of HF and significantly decreased in later (Tables 1 and 2). Studies have shown that there was impaired microcirculation in obesity and diabetes. And Advanced Glycation Endproducts (AGEs) activate inflammatory signals and mediate cell apoptosis and fibrosis [22]22 Falcão-Pires I, Hamdani N, Borbély A, Gavina C, Schalkwijk CG, van der Velden J, et al. Diabetes mellitus worsens diastolic left ventricular dysfunction in aortic stenosis through altered myocardial structure and cardiomyocyte stiffness. Circulation. 2011;124(10):1151-9.. Meanwhile, molecular variations such as transcription, post-translational modifications, and mitochondrial biogenesis contribute to the diversity of pathological energy metabolism in HF [23]23 Lopaschuk GD, Ussher JR, Folmes CD, Jaswal JS, Stanley WC Myocardial fatty acid metabolism in health and disease. Physiol Rev. 2010;90(1):207-58.. PPAR plays a central role in regulating FA metabolism. There were declined FAO and UCP3 expression followed by heart failure in PPARα-deficient mouse models [24]24 Cole MA, Abd Jamil AH, Heather LC, Murray AJ, Sutton ER, Slingo M, et al. On the pivotal role of PPARα in adaptation of the heart to hypoxia and why fat in the diet increases hypoxic injury. FASEB J. 2016;30(8):2684-97.. On the other hand, overexpression of PPARαbrings about cardiac dysfunction on account of the mismatch between FA uptake and utilization [25]25 Finck BN, Lehman JJ, Leone TC, Welch MJ, Bennett MJ, Kovacs A, et al. The cardiac phenotype induced by PPARalpha overexpression mimics that caused by diabetes mellitus. J Clin Invest. 2002;109(1):121-30.. Estrogen-related Receptors (ERRs), orphan nuclear receptors, take part in the regulation of genes referring to mitochondrial energy transduction, systolic function, and ion transport. A study found that ERRα/γ knockdown resulted in the arrest of mitochondrial maturation, activation of fibroblast-related genes, and eventually developed heart failure [26]26 Sakamoto T, Matsuura TR, Wan S, Ryba DM, Kim JU, Won KJ, et al. A critical role for estrogen-related receptor signaling in cardiac maturation. Circ Res. 2020;126(12):1685-702.. Interestingly, overexpression of ERRγ in mice also showed signs of cardiac hypertrophy and fibrosis in a GATA4-driven manner [27]27 Lasheras J, Pardo R, Velilla M, Poncelas M, Salvatella N, Simó R, et al. Cardiac-specific overexpression of ERRγ in mice induces severe heart dysfunction and early lethality. Int J Mol Sci. 2021;22(15):8047.. However, the relationship between cardiac function and PPAR as well as ERR expression in the failing heart has not been specifically studied.

Table 1
Study relevant to cardiac metabolism changes in human heart failure.
Table 2
Study relevant to cardiac metabolism changes in rats models of heart failure.

Uncoupling between glucose oxidase and glycolysis

Increased glucose uptake and glycolysis are conspicuous marks of a failing heart, but glucose oxidation is not synchronized with the increase in glycolysis [2828 Allard MF, Schönekess BO, Henning SL, English DR, Lopaschuk GD. Contribution of oxidative metabolism and glycolysis to ATP production in hypertrophied hearts. Am J Physiol. 1994;267(2 Pt 2):H742-50.,2929 Nascimben L, Ingwall JS, Lorell BH, Pinz I, Schultz V, Tornheim K, et al. Mechanisms for increased glycolysis in the hypertrophied rat heart. Hypertension. 2004;44(5):662-7.]. When the heart is faced with overloaded pressure, the expression of PFK1 and Fructose 2,6 Bisphosphate (F2,6BP) are boosted which contributes to the uptrend flow through glycolysis in the failing heart [30]30 Wang J, Xu J, Wang Q, Brainard RE, Watson LJ, Jones SP, et al. Reduced cardiac fructose 2,6 bisphosphate increases hypertrophy and decreases glycolysis following aortic constriction. PLoS One. 2013;8(1):e53951.. On the other side, the impaired liveness of the rate-limiting enzyme of glucose oxidation in the failing heart may aggravate the mismatch. The analysis of metabolomics, gene transcripts, and proteomics of tissues from the left ventricle revealed that mRNA expressions of PDH, MCT1, and pyruvate/alanine aminotransferase were reduced in HF. The possible mechanism is that hyperacetylation induced by HF and the hyperacetylation of PDH may inhibit their activity [31]31 Gupte AA, Hamilton DJ, Cordero-Reyes AM, Youker KA, Yin Z, Estep JD, et al. Mechanical unloading promotes myocardial energy recovery in human heart failure. Circ Cardiovasc Genet. 2014;7(3):266-76.. The reduction of myocardial glucose oxidation precedes the onset of diastolic dysfunction in hypertrophy mice which further verifies the adaptive role of reduced glucose oxidation in the development of heart failure [32]32 Zhang L, Jaswal JS, Ussher JR, Sankaralingam S, Wagg C, Zaugg M, et al. Cardiac insulin-resistance and decreased mitochondrial energy production precede the development of systolic heart failure after pressure-overload hypertrophy. Circ Heart Fail. 2013;6(5):1039-48..

Increased contribution of ketone bodies

Emerging evidence suggested that under the condition of low cardiac output, increased lipolysis in HF patients augmented the availability of ketone bodies, and overexcitation of the sympathetic nervous system promoted the generation and utilization of KBs (Tables 1 and 2). In HF rat models, the expression of BDH1, catalyzing ketone body oxidation, was elevated. And the same phenomenon was observed in patients with end-stage HF [3333 Aubert G, Martin OJ, Horton JL, Lai L, Vega RB, Leone TC, et al. The failing heart relies on ketone bodies as a fuel. Circulation. 2016;133(8):698-705.,3434 Bedi KC Jr, Snyder NW, Brandimarto J, Aziz M, Mesaros C, Worth AJ, et al. Evidence for intramyocardial disruption of lipid metabolism and increased myocardial ketone utilization in advanced human heart failure. Circulation. 2016;133(8):706-16.]. It is generally accepted that KBs are more efficient superfuels than other substrates [35]35 Ferrannini E, Mark M, Mayoux E CV protection in the EMPA-REG OUTCOME trial: A "thrifty substrate" hypothesis. Diabetes Care. 2016;39(7):1108-14.. In fact, recent studies have challenged the notion that ketone bodies are fuel-saving for the failing heart. Research indicated that the additional reducing equivalent accompanied by ketone bodies oxidation didn't match the production of ATP. The findings suggested that elevated ketone oxidation in the failing heart didn't do good to cardiac efficiency though KBs prompt ATP synthesis [3636 Ho KL, Karwi QG, Wagg C, Zhang L, Vo K, Altamimi T, et al. Ketones can become the major fuel source for the heart but do not increase cardiac efficiency. Cardiovasc Res. 2021;117(4):1178-87.,3737 Ho KL, Zhang L, Wagg C, Al Batran R, Gopal K, Levasseur J, et al. Increased ketone body oxidation provides additional energy for the failing heart without improving cardiac efficiency. Cardiovasc Res. 2019;115(11):1606-616.]. Therefore, the role of ketone body utilization in the development of HF is waiting to be further confirmed.

Dysfunction of mitochondria in the failing heart

Mitochondria are in charge of the homeostasis of energy metabolism. The latest study found that the expression of Dual-Specificity Tyrosine-Regulated Kinase 1B (DYRK1B) which mediated cardiac hypertrophy and fibrosis by damaging mitochondrial bioenergetics was upregulated in HF. DYRK1B increases its phosphorylation and nuclear accumulation by directly binding to STAT3, leading to the down-regulation of PGC-1α level and subsequent cardiac insufficiency [38]38 Zhuang L, Jia K, Chen C, Li Z, Zhao J, Hu J, et al. DYRK1B-STAT3 drives cardiac hypertrophy and heart failure by impairing mitochondrial bioenergetics. Circulation. 2022;145(11):829-46.. Mitochondrial function is controlled by multiple post-translational modifications. In animal models and human failing hearts, acetylation of mitochondrial proteins was significantly increased which resulted in an impaired mitochondrial respiratory chain [39]39 Horton JL, Martin OJ, Lai L, Riley NM, Richards AL, Vega RB, et al. Mitochondrial protein hyperacetylation in the failing heart. JCI Insight. 2016;2(1):e84897.. The reduced protein deacetylation is the latent mechanism. A study found that the expression of mitochondrial deacetylase SIRT3 was decreased and proved that Mir-195 down-regulated SIRT3 expression by directly targeting the direct 3′-untranslated region [40]40 Zhang X, Ji R, Liao X, Castillero E, Kennel PJ, Brunjes DL, et al. MicroRNA-195 regulates metabolism in failing myocardium via alterations in sirtuin 3 expression and mitochondrial protein acetylation. Circulation. 2018;137(19):2052-2067.. Conversely, the activity of deacetylation relies on the availability of NAD+. Hyperacetylation inhibits the activity of the malate-aspartate shuttle which is the limitation of the transport of NADH from cytoplasm to mitochondria, and then disrupts the cytoplasmic REDOX state of the failing heart [41]41 Lee CF, Chavez JD, Garcia-Menendez L, Choi Y, Roe ND, Chiao YA, et al. Normalization of NAD+ redox balance as a therapy for heart failure. Circulation. 2016;134(12):883-94..

Counteraction of metabolic remodeling ‒ deteriorated cellular environment

Insulin resistance

Failing hearts exhibit significant metabolic remodeling, and there is a debate about whether these alterations contribute to the development of heart failure. A study involving 2623 patients found that there were increased wall thickness and LV mass with the deterioration of glucose intolerance among patients [42]42 Rutter MK, Parise H, Benjamin EJ, Levy D, Larson MG, Meigs JB, et al. Impact of glucose intolerance and insulin resistance on cardiac structure and function: sex-related differences in the Framingham Heart Study. Circulation. 2003;107(3):448-54.. Normally, insulin reduces mitochondrial FA uptake by increasing the activity of acetyl-CoA carboxylase. Impaired mitochondrial β-oxidation and boosted FAO rate are accompanied by changes in cardiac insulin signaling, including the activation of the proximal insulin signaling pathway IRS/Akt [32]32 Zhang L, Jaswal JS, Ussher JR, Sankaralingam S, Wagg C, Zaugg M, et al. Cardiac insulin-resistance and decreased mitochondrial energy production precede the development of systolic heart failure after pressure-overload hypertrophy. Circ Heart Fail. 2013;6(5):1039-48.. This alteration contributes to insulin resistance characterized by reduced glucose oxidation and impaired inhibition of FAO. Recent studies found that IRS-1/Akt1 was activated in the failing heart and the deficiency of IRS1 exerted a protective effect in HF mice while IRS-2 acted the opposite [43]43 Riehle C, Weatherford ET, Wende AR, Jaishy BP, Seei AW, McCarty NS, et al. Insulin receptor substrates differentially exacerbate insulin-mediated left ventricular remodeling. JCI Insight. 2020;5(6):e134920.. In a post-MI mouse model, it was found that the injured myocardium promoted the degradation of Insulin Receptor Substrate 1 (IRS1) by upregulating MIR128-3p [44]44 Ruiz-Velasco A, Zi M, Hille SS, Azam T, Kaur N, Jiang J, et al. Targeting mir128-3p alleviates myocardial insulin resistance and prevents ischemia-induced heart failure. Elife. 2020;9:e54298.. However, scholars proposed that insulin resistance provided protection for myocardial fuel overload in obesity or diabetes [45]45 Taegtmeyer H, Beauloye C, Harmancey R, Hue L Insulin resistance protects the heart from fuel overload in dysregulated metabolic states. Am J Physiol Heart Circ Physiol. 2013;305(12):H1693-7..

Lipotoxicity

In heart failure, the chaos of uptake and utilization of FA gives rise to the accumulation of lipid intermediates, such as ceramide and diacylglycerol. The study has found that the overexpression of GSK3α boosted the uptake of FFA in the failing heart leading to fibrosis and cardiac hypertrophy [46]46 Nakamura M, Liu T, Husain S, Zhai P, Warren JS, Hsu CP, et al. Glycogen synthase kinase-3α promotes fatty acid uptake and lipotoxic cardiomyopathy. Cell Metab. 2019;29(5):1119-1134.e12.. Similarly, increased FFA uptake was found to be accompanied by impaired left ventricular filling function and atrial enlargement in FATP1+/+ mice [47]47 Chiu HC, Kovacs A, Blanton RM, Han X, Courtois M, Weinheimer CJ, et al. Transgenic expression of fatty acid transport protein 1 in the heart causes lipotoxic cardiomyopathy. Circ Res. 2005;96(2):225-33.. Under the circumstances, endoplasmic reticulum stress makes negative impact on cardiomyocytes mediated by pressure-driven lipid accumulation, which is correlated with the expression of the Very Low-Density Lipoprotein Receptor (VLDLR). Lipotoxicity, a byproduct of this process, may cause myocardial apoptosis, insulin resistance and systolic dysfunction [48]48 Okada K, Minamino T, Tsukamoto Y, Liao Y, Tsukamoto O, Takashima S, et al. Prolonged endoplasmic reticulum stress in hypertrophic and failing heart after aortic constriction: possible contribution of endoplasmic reticulum stress to cardiac myocyte apoptosis. Circulation. 2004;110(6):705-12.. This point of view was confirmed by reduced ischemia-induced endoplasmic reticulum stress and cardiac apoptosis among VLDLR−/− mice and mice treated with antibodies specific for VLDLR [49]49 Perman JC, Boström P, Lindbom M, Lidberg U, StÅhlman M, Hägg D, et al. The VLDL receptor promotes lipotoxicity and increases mortality in mice following an acute myocardial infarction. J Clin Invest. 2011;121(7):2625-40.. The study has proved that the expression of Mst1 elevated in rats with lipid-induced heart injury. In this study, lipotoxicity induced the expression of Forkhead box O3 (FoxO3) by promoting the binding of FoxO3 to the Mst1 promoter, and deficiency of Mst1 gene ameliorated apoptosis and inflammation [50]50 Xiong Z, Li Y, Zhao Z, Zhang Y, Man W, Lin J, et al. Mst1 knockdown alleviates cardiac lipotoxicity and inhibits the development of diabetic cardiomyopathy in db/db mice. Biochim Biophys Acta Mol Basis Dis. 2020;1866(8):165806..

Oxidative stress

The extra glucose entered pathways, mainly Pentose Phosphate Pathway (PPP), and Hexosamine Biosynthesis Pathway (HBP). Glucose 6-Phosphate Dehydrogenase (G6PD), the rate-limiting enzyme of PPP, is essential to maintain the REDOX state of cardiomyocytes. And elevated expression and activity of G6PD are observed in both humans and canines [5151 Gupte RS, Vijay V, Marks B, Levine RJ, Sabbah HN, Wolin MS, et al. Upregulation of glucose-6-phosphate dehydrogenase and NAD(P)H oxidase activity increases oxidative stress in failing human heart. J Card Fail. 2007;13(6):497-506.,5252 Gupte SA, Levine RJ, Gupte RS, Young ME, Lionetti V, Labinskyy V, et al. Glucose-6-phosphate dehydrogenase-derived NADPH fuels superoxide production in the failing heart. J Mol Cell Cardiol. 2006;41(2):340-9.]. Studies showed that G6PD deficiency might contribute to cardiac dysfunction by boosting susceptibility to free radical damage and impairing intracellular ion transport [53]53 Jain M, Brenner DA, Cui L, Lim CC, Wang B, Pimentel DR, et al. Glucose-6-phosphate dehydrogenase modulates cytosolic redox status and contractile phenotype in adult cardiomyocytes. Circ Res. 2003;93(2):e9-16.. As an adaptive response, HBP mediates the O-linked-β-N-Acetylglucosaminylation (O-GlcNAcylation) of proteins via glutamine-fructose-6-phosphate while the integration of various cellular signals, including intracellular and intracellular stress and nutrient levels [5454 Champattanachai V, Marchase RB, Chatham JC Glucosamine protects neonatal cardiomyocytes from ischemia-reperfusion injury via increased protein-associated O-GlcNAc. Am J Physiol Cell Physiol. 2007;292(1):C178-87.,5555 Liu J, Pang Y, Chang T, Bounelis P, Chatham JC, Marchase RB Increased hexosamine biosynthesis and protein O-GlcNAc levels associated with myocardial protection against calcium paradox and ischemia. J Mol Cell Cardiol. 2006;40(2):303-12.]. Glutamine-Fructose-6-phosphate Transaminase (GFAT) is a key enzyme in HBP, and studies found that GFAT1 was the target of X-box binding protein 1 (Xbp1s), a pivotal transcription factor of Unfolded Protein Reaction (UPR). The overexpression of XBP1s in cardiomyocytes gives rise to elevated activity of HBP and O-GlcNAcylation [56]56 Wang ZV, Deng Y, Gao N, Pedrozo Z, Li DL, Morales CR, et al. Spliced X-box binding protein 1 couples the unfolded protein response to hexosamine biosynthetic pathway. Cell. 2014;156(6):1179-92.. Besides, oxidative stress also disrupts the Nitric Oxide (NO); Soluble Guanylate Cyclase (sGC); Cyclic Guanosine Monophosphate (cGMP) pathway, causing elevated cGMP levels in cells, further promoting the development of HF [57]57 Kolijn D, Pabel S, Tian Y, Lódi M, Herwig M, Carrizzo A, et al. Empagliflozin improves endothelial and cardiomyocyte function in human heart failure with preserved ejection fraction via reduced pro-inflammatory-oxidative pathways and protein kinase Gα oxidation. Cardiovasc Res. 2021;117(2):495-507..

Calcium dyshomeostasis

Ca2+, governing EC coupling, is the bridge of the communication between cardiac electrical activity and excitation-contraction coupling. In HF, Ca2+ leaks through the channel RyR2. Current research has identified several potential factors for Ca2+ leakage, including the conformational change of RyR2 induced by hyperphosphorylation [5858 Lehnart SE, Mongillo M, Bellinger A, Lindegger N, Chen BX, Hsueh W, et al. Leaky Ca2+ release channel/ryanodine receptor 2 causes seizures and sudden cardiac death in mice. J Clin Invest. 2008;118(6):2230-45.,5959 Uchinoumi H, Yang Y, Oda T, Li N, Alsina KM, Puglisi JL, et al. CaMKII-dependent phosphorylation of RyR2 promotes targetable pathological RyR2 conformational shift. J Mol Cell Cardiol. 2016;98:62-72.]. In addition, abnormal mitochondrial aggregation and structural reorganization may impair the transportation of Ca2+ signaling, and then give rise to ROS accumulation [60]60 Pinali C, Bennett H, Davenport JB, Trafford AW, Kitmitto A Three-dimensional reconstruction of cardiac sarcoplasmic reticulum reveals a continuous network linking transverse-tubules: this organization is perturbed in heart failure. Circ Res. 2013;113(11):1219-30.. ROS, a by-product of mitochondrial respiration, has a negative impact on the failing heart related to the activation of multiple signaling pathways and apoptosis. Studies confirmed that Calcium/Calmodulin-dependent protein Kinase II (CaMKII) took a vital part in the development of HF that promoted mPTP opening and apoptosis by mediating the current of the inner membrane Mitochondrial Ca2+ Uniporter (MCU) [6161 Vila-Petroff M, Salas MA, Said M, Valverde CA, Sapia L, Portiansky E, et al. CaMKII inhibition protects against necrosis and apoptosis in irreversible ischemia-reperfusion injury. Cardiovasc Res. 2007;73(4):689-98.,6262 Zhang T, Maier LS, Dalton ND, Miyamoto S, Ross J Jr, Bers DM, et al. The deltaC isoform of CaMKII is activated in cardiac hypertrophy and induces dilated cardiomyopathy and heart failure. Circ Res. 2003;92(8):912-9.]. Latest research indicated that ROS-mediated oxidation induced the changes in mitochondrial permeability which led to cellular damage and apoptosis by activating RIP3, an upstream kinase of CaMKII [63]63 Zhang T, Zhang Y, Cui M, Jin L, Wang Y, Lv F, et al. CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress-induced myocardial necroptosis. Nat Med. 2016;22(2):175-82.. To a certain degree, cardiac function relies on the persistent supply of ATP and the balance between ROS production and clearance. In HF and other cardiac diseases, EC uncoupling and impaired mitochondrial Ca2+ dynamics are widespread that deteriorate the cellular environment.

Redistribution of receptors on cardiomyocytes

Heart failure causes significant changes in hormone receptors, which typically manifests as the failing heart escaping from the supervision of adrenergic. And the downregulation of β1 receptor is the most significant [64]64 Bristow MR Changes in myocardial and vascular receptors in heart failure. J Am Coll Cardiol. 1993;22(4 Suppl A):61A-71A.. Myocardial membrane biopsy of HF patients indicated that the density of β-AR gradually decreased as the increased severity of heart failure [65]65 Fowler MB, Laser JA, Hopkins GL, Minobe W, Bristow MR Assessment of the beta-adrenergic receptor pathway in the intact failing human heart: progressive receptor down-regulation and subsensitivity to agonist response. Circulation. 1986;74(6):1290-302.. Meanwhile, the ratio of β1:β2-AR decreased from 80:20 to 60:40, and other changes included increased β-AR uncoupling and G protein activity [66]66 Lamba S, Abraham WT Alterations in adrenergic receptor signaling in heart failure. Heart Fail Rev. 2000;5(1):7-16.. β-AR receptors are dominant in the human heart, among which β3-AR is closely related to glucose and lipid metabolism. It is suggested that β3-AR confronts the myocardial contraction through NO/cGMP pathway. Under physiological conditions, the level of β3-AR in myocardial tissue is extremely low (only 3%), and the expression of β3-AR is significantly upregulated in failing hearts and shows a myocardial protective effect [6767 Bristow MR, Ginsburg R, Umans V, Fowler M, Minobe W, Rasmussen R, et al. Beta 1- and beta 2-adrenergic-receptor subpopulations in nonfailing and failing human ventricular myocardium: coupling of both receptor subtypes to muscle contraction and selective beta 1-receptor down-regulation in heart failure. Circ Res. 1986;59(3):297-309.,6868 Trappanese DM, Liu Y, McCormick RC, Cannavo A, Nanayakkara G, Baskharoun MM, et al. Chronic β1-adrenergic blockade enhances myocardial β3-adrenergic coupling with nitric oxide-cGMP signaling in a canine model of chronic volume overload: new insight into mechanisms of cardiac benefit with selective β1-blocker therapy. Basic Res Cardiol. 2015;110(1):456.]. Clinical studies have shown that mirabegrone (the β3-AR receptor agonist) improved insulin sensitivity and decreases plasma hemoglobin A1c levels in obese patients, and the expression of PGC1, transcription factor A, and cyclooxygenase IV was increased. In addition, Mirabelone can increase the level of Nonesterified Fatty Acids (NEFA) in plasma and adipose tissue. And it is widely believed that a high level of NEFA contributes to ectopic lipid accumulation and the inhibition of insulin receptor signaling in skeletal muscle [69]69 Finlin BS, Memetimin H, Zhu B, Confides AL, Vekaria HJ, El Khouli RH, et al. The β3-adrenergic receptor agonist mirabegron improves glucose homeostasis in obese humans. J Clin Invest. 2020;130(5):2319-31.. β3-AR agonist improved LVEDP and end-systolic pressure-volume relation slope, reducing ventricular muscle stiffness and inhibiting Smad2/3, phospho-Smad2/3, and TGF-β1 expression in CHF mice [70]70 Kamiya M, Asai K, Maejima Y, Shirakabe A, Murai K, Noma S, et al. β3-Adrenergic receptor agonist prevents diastolic dysfunction in an angiotensin II-induced cardiomyopathy mouse model. J Pharmacol Exp Ther. 2021;376(3):473-481.. Overexpression of β3-AR relieved cardiomyocyte hypertrophy, delaying the progression of heart failure, reducing mitochondrial fragmentation, and maintaining mitochondrial ridge integrity and dynamic homeostasis in Aortic stenosis mice [71]71 Pun-García A, Clemente-Moragón A, Villena-Gutierrez R, Gómez M, Sanz-Rosa D, Díaz-Guerra A, et al. Beta-3 adrenergic receptor overexpression reverses aortic stenosis-induced heart failure and restores balanced mitochondrial dynamics. Basic Res Cardiol. 2022;117(1):62.. Intracellular Ca2+ is a major factor affecting arrhythmia during heart failure. β3-AR agonists can reduce the incidence of ventricular arrhythmia in CHF rabbits, and inhibit INCX, Ca2+ transient, SR Ca2+ load and leakage [72]72 Li H, Liu Y, Huang H, Tang Y, Yang B, Huang C Activation of β3-adrenergic receptor inhibits ventricular arrhythmia in heart failure through calcium handling. Tohoku J Exp Med. 2010;222(3):167-74.. Whereas, the absence of β3-AR receptor caused left ventricular diastolic dysfunction in mice [73]73 Yang W, Wei X, Su X, Shen Y, Jin W, Fang Y. Depletion of β3-adrenergic receptor induces left ventricular diastolic dysfunction via potential regulation of energy metabolism and cardiac contraction. Gene. 2019;697:1-10.. PPAR is involved in the regulation of blood cholesterol and glucose concentration, among which PPARα/δ is the dominant subtype in the heart tissue. In HF, increased adrenergic tension activates signal transduction molecules such as HIF, mTOR, and NF-κB, thereby reversely inhibiting the expression and activity of PPARα/δ, which results in a shift in the energy metabolic profile of the failing heart to the infantile pattern [74]74 Montaigne D, Butruille L, Staels B PPAR control of metabolism and cardiovascular functions. Nat Rev Cardiol. 2021;18(12):809-823.. P2 purinergic receptors are key molecules for ATP. The expression of P2X4/7 is the highest in the human sinoatrial node. In MI-induced HF rats, the mRNA expression of P2X4 was elevated in the sinus node. And the highly expressed of P2X4 receptor increased the formation of S-nitrosylation, cGMP, and NO in hyperlipidemia mice [7575 Musa H, Tellez JO, Chandler NJ, Greener ID, Maczewski M, Mackiewicz U, et al. P2 purinergic receptor mRNA in rat and human sinoatrial node and other heart regions. Naunyn Schmiedebergs Arch Pharmacol. 2009;379(6):541-9.,7676 Yang SM, Liu J, Li CX Intermedin protects against myocardial ischemia-reperfusion injury in hyperlipidemia rats. Genet Mol Res. 2014;13(4):8309-19.]. The expression of P2Y6 receptor was elevated in TAC mice and induced fibrosis through the activation of Galpha 12/13 [77]77 Nishida M, Sato Y, Uemura A, Narita Y, Tozaki-Saitoh H, Nakaya M, et al. P2Y6 receptor-Galpha12/13 signalling in cardiomyocytes triggers pressure overload-induced cardiac fibrosis. EMBO J. 2008;27(23):3104-15.. However, the relationship between changes in receptors of cardiomyocytes and heart failure has not been specifically studied.

Metabolic changes in different types of heart failure

In heart failure, the pathophysiological differences of different types of HF make the uptake and utilization of energy substrates present different metabolic profiles. A study collected the coronary blood samples of 15 patients with HFrEF. The results indicated that compared with the control group, the absorption of FFA and glucose in HFrEF patients had not increased, while the plasma β-hydroxybutyrate and acetylacetate levels were slightly elevated, and the concentration of multiple acylcarnitines increased. In terms of energy contribution, the relative contributions of FFA and glucose decreased, while the relative contributions of ketone bodies increased. And the contribution of beta-hydroxybutyric acid and acetoacetic acid on the ATP production increased from 6.8% in the control group to 24% in the HFrEF group [78]78 Voros G, Ector J, Garweg C, Droogne W, Van Cleemput J, Peersman N, et al. Increased cardiac uptake of ketone bodies and free fatty acids in human heart failure and hypertrophic left ventricular remodeling. Circ Heart Fail. 2018;11(12):e004953.. Another study included 46 patients with decreased ejection fraction (LVEF: 39.59 ± 10.94%; NYHA class ≥ II) and showed that the concentration of 3-hydroxybutyrate, acetone and succinate were significantly elevated with the increase of cardiac energy consumption [79]79 Du Z, Shen A, Huang Y, Su L, Lai W, Wang P, et al. 1H-NMR-based metabolic analysis of human serum reveals novel markers of myocardial energy expenditure in heart failure patients. PLoS One. 2014;9(2):e88102.. 82 serum samples were quantitatively detected by LC-MS/MS and 1H-NMR. The results showed that compared with the non-HF group, the serum concentrations of acylcarnitine, carnitine, creatinine, betaine, and amino acids in HFpEF patients were increased, while the levels of phosphatidylcholine, lysophosphatidylcholine, and sphingomyelin were decreased. However, the levels of medium and long-chain acylcarnitine and ketone bodies in HFpEF patients were higher than those with HFrEF [80]80 Zordoky BN, Sung MM, Ezekowitz J, Mandal R, Han B, Bjorndahl TC, Alberta HEART. Metabolomic fingerprint of heart failure with preserved ejection fraction. PLoS One. 2015;10(5):e0124844.. Cardiac magnetic resonance was applied to detect the myocardial fat content in HFpEF (n = 163), HFrEF (n = 34) and the control group, and the results showed that the cardiac fat content in the HFpEF group was more than twice that of the control group. On the contrary, the HFrEF group showed a downward trend [81]81 Wu CK, Lee JK, Hsu JC, Su MM, Wu YF, Lin TT, et al. Myocardial adipose deposition and the development of heart failure with preserved ejection fraction. Eur J Heart Fail. 2020;22(3):445-54.. In addition, most studies support the idea that under pressure overload the utilization of glucose increases. Positron emission tomography showed that there were increased glucose uptake and utilization, and decreased FFA among patients with cardiomyopathy [82]82 Dávila-Román VG, Vedala G, Herrero P, de las Fuentes L, Rogers JG, Kelly DP, et al. Altered myocardial fatty acid and glucose metabolism in idiopathic dilated cardiomyopathy. J Am Coll Cardiol. 2002;40(2):271-7.. Basic studies have shown that the glucose uptake and utilization increased on the first day after TAC operation in HF mice, and showed an elevated trend with time [83]83 Doenst T, Pytel G, Schrepper A, Amorim P, Färber G, Shingu Y, et al. Decreased rates of substrate oxidation ex vivo predict the onset of heart failure and contractile dysfunction in rats with pressure overload. Cardiovasc Res. 2010;86(3):461-70.. The changes in energy metabolism in a failing heart are complex, and the severity, type, stage, and comorbidities of heart failure will have a profound impact on the utilization of heart energy.

Treatments

Pharmacological agents for improving the cellular metabolic environment

Altering fatty acid oxidation

Whether the reduction in FAO during HF is either protective or unadapted determines administration of medication. Based on what boosted FAO is strongly correlated with the increase of ROS, inhibition of FAO may exert a great cardioprotective effect by inhibiting intracellular oxidative stress levels, preventing the accumulation of toxic lipid intermediates, and maintaining cellular environmental homeostasis [84]84 Schönfeld P, Wojtczak L Fatty acids decrease mitochondrial generation of reactive oxygen species at the reverse electron transport but increase it at the forward transport. Biochim Biophys Acta. 2007;1767(8):1032-40.. Trimetazidine and ranolazine, optimizing substrates of myocardial metabolism, are inhibitors of 3-Ketoacyl coenzyme A Thiolase (3-KAT) which catalyzes the last step of β-oxidation. Trimetazidine indirectly stimulates the activity of PDH and raises the utilization of glucose by selectively inhibiting FAO [85]85 Marzilli M, Vinereanu D, Lopaschuk G, Chen Y, Dalal JJ, Danchin N, et al. Trimetazidine in cardiovascular medicine. Int J Cardiol. 2019;293:39-44.. In mouse models with HF induced by pressure-overloading, trimetazidine activated AMPK in a dose-dependent manner to enhance glucose uptake as well as transformation of metabolic substrates and ameliorate insulin resistance [86]86 Shu H, Hang W, Peng Y, Nie J, Wu L, Zhang W, et al. Trimetazidine attenuates heart failure by improving myocardial metabolism via AMPK. Front Pharmacol. 2021;12:707399.. And trimetazidine decreases the accumulation of H+ and lactic acid in the cytoplasm, in turn, avoids calcium overload and other adverse cardiac events [87]87 Shu H, Peng Y, Hang W, Zhou N, Wang DW. Trimetazidine in heart failure. Front Pharmacol. 2020;11:569132.. Renolazine alleviates myocardial hypertrophy and fibrosis by optimizing myocardial energy metabolism and alleviating Na+-dependent calcium overload [88]88 Nie J, Duan Q, He M, Li X, Wang B, Zhou C, et al. Ranolazine prevents pressure overload-induced cardiac hypertrophy and heart failure by restoring aberrant Na+ and Ca2+ handling. J Cell Physiol. 2019;234(7):11587-601.. β-blockers are the cornerstone drugs in the treatment of heart failure, which can shift myocardial substrate utilization from FFA to glucose oxidation, thereby reducing myocardial oxygen consumption and improving myocardial efficiency. In 26 patients with moderate to severe heart failure treated with carvedilol for 6 months, the lipid oxidation rate decreased significantly (2.4±1.4 to 1.5±0.9 mg m2/kg min) [89]89 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(6):369-78.. Glucose oxidation rate increased (2.6±1.4 to 4.4±1.6 mg m2/kg min). Basic experiments showed that metoprolol alleviated cardiac dysfunction, reducing palmitate oxidation rate, stimulating glucose oxidation, and increasing tissue ATP levels in diabetic rats [90]90 Sharma V, Dhillon P, Wambolt R, Parsons H, Brownsey R, Allard MF, et al. Metoprolol improves cardiac function and modulates cardiac metabolism in the streptozotocin-diabetic rat. Am J Physiol Heart Circ Physiol. 2008, 294(4):H1609-20.. While recent studies have shown that II/III generation β-blockers have little effect on substrate oxidation among HFrEF patients [91]91 Ribeiro PAB, Normandin E, Meyer P, Juneau M, White M, Nigam A, et al. Beta-blocker type effect on substrate oxidation during HIIE in heart failure patients: pilot data. Arq Bras Cardiol. 2019;112(3):304-308..

Besides, reducing cardiac FAO through the modification of PPARs is a latent therapeutic approach. Fibrates, the PPARs modulators, decrease β-oxidation by reducing FFA levels and increasing the utilization by other tissues. Studies confirmed that pemafibrate induced the expression of Lipoprotein Lipase (LPL) in mice by activating PPARα to reduce TG and lessen the secretion of LDL-C. At the same time, based on the concept of easing the burden on kidneys, pemafibrate is mainly metabolized through the liver, and excretion through urine only accounts for 14.5%. Thus, pemafibrate can be safely applied in patients with chronic kidney disease [92]92 Pradhan AD, Paynter NP, Everett BM, Glynn RJ, Amarenco P, Elam M, et al. Rationale and design of the pemafibrate to reduce cardiovascular outcomes by reducing triglycerides in patients with diabetes (PROMINENT) study. Am Heart J. 2018;206:80-93.. The accumulation of ROS and toxic lipid metabolites caused by FFA is a vital agent for the deteriorated cellular environment. However, there is a potential risk of reducing fatty acid oxidation, which would further reduce the energy supply of ATP. Therefore, the management of heart failure through inhibiting fatty acid oxidation needs to be carefully considered.

Modulating glucose oxidation

Insulin signaling is an important metabolic pathway in the cellular environment. In heart failure, neurohumoral and cytokine imbalances and cellular oxidative stress induce insulin resistance, resulting in increased fatty acid flux into cardiomyocytes. Since glucose is a more efficient substrate, the switch in cardiometabolic metabolism from glucose utilization to fatty acid oxidation may reduce cardiac efficiency. In the severe stage of heart failure, ATP level declines sharply and the inhibition of FAO may further damage cardiac function. Thus, direct stimulation of glucose oxidation may be a better option. Metformin was verified to have a relation with reduced HF mortality as well as readmission in a meta-analysis, with a 22% lower all-cause mortality for patients taking metformin than for those not [93]93 Crowley MJ, Diamantidis CJ, McDuffie JR, Cameron CB, Stanifer JW, Mock CK, et al. Clinical outcomes of metformin use in populations with chronic kidney disease, congestive heart failure, or chronic liver disease: a systematic review. Ann Intern Med. 2017;166(3):191-200.. Studies found that Metformin promoted the uptake as well as utilization of glucose and lowered FFA to avoid the lipotoxicity followed by FA accumulation. Other beneficial effects include improving the function of vascular endothelial cells and fighting against oxidative stress [94]94 Mather KJ, Verma S, Anderson TJ Improved endothelial function with metformin in type 2 diabetes mellitus. J Am Coll Cardiol. 2001;37(5):1344-50.. A recent experiment revealed the direct molecular target of metformin. Metformin-bound PEN2 forms a complex with ATP6AP1, a subunit of the v-ATPase8, which induces the inhibition of v-ATPase and the activation of AMPK [95]95 Ma T, Tian X, Zhang B, Li M, Wang Y, Yang C, et al. Low-dose metformin targets the lysosomal AMPK pathway through PEN2. Nature. 2022;603(7899):159-165.. The process enhances that metformin performs its beneficial role without substantial side effects.

2021 ESC Guidelines clearly brought Sodium-Glucose cotransporter-2 inhibitors (SGLT2i) as the primary drug for the treatment of heart failure with reduced ejection fraction (HFrEF). Its latent beneficial effects on the heart are as follows: Firstly, SGLT2i, acting like osmotic diuretics, improves ventricular load conditions, optimizes volume management, and reduces cardiac energy consumption by boosting sodium and glucose excretion [96]96 Garcia-Ropero A, Santos-Gallego CG, Zafar MU, Badimon JJ Metabolism of the failing heart and the impact of SGLT2 inhibitors. Expert Opin Drug Metab Toxicol. 2019;15(4):275-85.. Secondly, SGLT2i may transform the utilization of myocardial substrates and then boost the production and storage of ATP in mitochondria on account of promoting the decomposition of FA and lifting the level of KBs [97]97 Santos-Gallego CG, Requena-Ibanez JA, San Antonio R, Ishikawa K, Watanabe S, Picatoste B, et al. Empagliflozin ameliorates adverse left ventricular remodeling in nondiabetic heart failure by enhancing myocardial energetics. J Am Coll Cardiol. 2019;73(15):1931-44.. Thirdly, the newest research found that SGLT2i presented cardioprotective effects by regulating excessive autophagy of myocardium, directly suppressing the activity of the Na+/H+ Exchanger 1 (NHE1) in the cardiomyocytes [98]98 Jiang K, Xu Y, Wang D, Chen F, Tu Z, Qian J, et al. Cardioprotective mechanism of SGLT2 inhibitor against myocardial infarction is through reduction of autosis. Protein Cell. 2022;13(5):336-59.. A recent trial involving 4744 patients with HFrEF confirmed that the application of dapagliflozin lowered the risk of worsening heart failure or death from cardiovascular causes more than those who received a placebo, regardless of the presence or absence of DM [99]99 McMurray JJV, DeMets DL, Inzucchi SE, Køber L, Kosiborod MN, Langkilde AM, et al.; DAPA-HF committees and investigators. A trial to evaluate the effect of the sodium-glucose co-transporter 2 inhibitor dapagliflozin on morbidity and mortality in patients with heart failure and reduced left ventricular ejection fraction (DAPA-HF). Eur J Heart Fail. 2019;21(5):665-75.. And studies have verified that facilitating cardiac ketone body oxidation was the protective way against the failing heart. The application of ketone ester in a post-MI rat might improve cardiac function and ameliorate cardiac remodeling by reprogramming the genetic expression involved in KBs utilization [100]100 Yurista SR, Matsuura TR, Silljé HHW, Nijholt KT, McDaid KS, Shewale SV, et al. Ketone ester treatment improves cardiac function and reduces pathologic remodeling in preclinical models of heart failure. Circ Heart Fail. 2021;14(1):e007684.. However, it seems unable to maintain high circulating KBs level in the long term, whereas the emergence of SGLT2i, increasing ketone bodies to support TCA circulation, overcomes this problem. And other studies showed that increased circulating ketone bodies following the administration of SGLT2i may relieve inflammation in the failing heart by attenuating NLRP3 inflammasome activation [101]101 Kim SR, Lee SG, Kim SH, Kim JH, Choi E, Cho W, et al. SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nat Commun. 2020;11(1):2127.. It is worth noting that in the cellular environment, the advantages and disadvantages of a single metabolic substrate are not absolute. And the ability to maintain metabolic flexibility and boost ATP production plays an equally vital role in the diseases.

Improving mitochondrial dysfunction

Mitochondrial dysfunction has a close relation with oxidative stress during the development of HF, and attempts targeting mitochondrial ROS have shown advantages in the treatment of HF. Coenzyme Q, a part of the electron transport chain, is involved in electron transfer in ETC to regulate substrate oxidation and thereby exert antioxidant effects by removing excess ROS. Thus Coenzyme Q10 (CoQ10) supplementation has turned into a safe and effective option [102]102 Ayer A, Macdonald P, Stocker R CoQ₁₀ function and role in heart failure and ischemic heart disease. Annu Rev Nutr. 2015;35:175-213.. A meta-analysis incorporating 14 RCT experiments confirmed that the application of CoQ10 reduced mortality and improved the exercise capacity of patients with HF [103]103 Lei L, Liu Y Efficacy of coenzyme Q10 in patients with cardiac failure: a meta-analysis of clinical trials. BMC Cardiovasc Disord. 2017;17(1):196.. NAD+/NADH, at a relatively low-level amount patients with HF, is another prospective approach to restore the metabolic balance in the failing myocardium [104]104 Diguet N, Trammell SAJ, Tannous C, Deloux R, Piquereau J, Mougenot N, et al. Nicotinamide riboside preserves cardiac function in a mouse model of dilated cardiomyopathy. Circulation. 2018;137(21):2256-73.. NAD+, the electron donor, participates in glycolysis, TCA cycle and oxidative phosphorylation while acting as a signal transduction molecule to regulate acetylation of mitochondrial proteins [105]105 Hong W, Mo F, Zhang Z, Huang M, Wei X Nicotinamide mononucleotide: a promising molecule for therapy of diverse diseases by targeting NAD+ metabolism. Front Cell Dev Biol. 2020;8:246.. In a murine model with HFpEF, NAD+ repletion has been demonstrated to improve the mitochondrial function as well as exercise capacity and ameliorate the HFpEF phenotype [106]106 Tong D, Schiattarella GG, Jiang N, Altamirano F, Szweda PA, Elnwasany A, et al. NAD+ repletion reverses heart failure with preserved ejection fraction. Circ Res. 2021;128(11):1629-41.. This experiment supports the positive side of elevated NAD+ levels in the failing heart, but the efficacy and safety of exogenous NAD+ supplement therapy for HF need to be verified in clinical trials (Table 3).

Table 3
Pharmacological therapies targeting cardiac metabolism in heart failure.

Stem cells targeting cytothesis

Injured cardiomyocytes enter the new cell cycle, and some endogenous cardiac stem cells may participate in the process of regeneration and repairment by regulating the secretion of cytokines. In the last decade, several types of stem cells have been applied to repair damaged cardiomyocytes in pre-clinical and clinical trials. For now, the hypotheses about latent mechanisms include promoting cardiomyocyte regeneration, angiopoiesis, reducing apoptosis, and intervening in ventricular remodeling by paracrine [107107 Gnecchi M, He H, Liang OD, Melo LG, Morello F, Mu H, et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med. 2005;11(4):367-8.,108108 Mirotsou M, Jayawardena TM, Schmeckpeper J, Gnecchi M, Dzau VJ Paracrine mechanisms of stem cell reparative and regenerative actions in the heart. J Mol Cell Cardiol. 2011;50(2):280-9]. Studies found that injection of bone marrow Mesenchymal Stem Cells (MSCs) significantly elevated left ventricular ejection fraction, lessened end-systolic volume, and increased 6-minute walking distance without cardiotoxicity [109]109 Bartunek J, Behfar A, Dolatabadi D, Vanderheyden M, Ostojic M, Dens J, et al. Cardiopoietic stem cell therapy in heart failure: the C-CURE (Cardiopoietic stem Cell therapy in heart failURE) multicenter randomized trial with lineage-specified biologics. J Am Coll Cardiol. 2013;61(23):2329-38.. In a study involving 65 patients, MSCs infusion increased the expression of hepatocyte growth factors such as myogenesis, cell migration, and immune adjustment with improvements in the New York Heart Association class and the Minnesota Heart Failure Living Questionnaire under the standard treatment of heart failure [110]110 Bartolucci J, Verdugo FJ, González PL, Larrea RE, Abarzua E, Goset C, et al. Safety and efficacy of the intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure: a phase 1/2 randomized controlled trial (RIMECARD trial [Randomized clinical trial of intravenous infusion umbilical cord mesenchymal stem cells on cardiopathy]). Circ Res. 2017;121(10):1192-204.. Ixmyelocel-T consists of a mixture of cells including macrophages, granulocytes, monocytes, lymphocytes, and MSCs. In a study, the application of ixmyelocel-T in symptomatic HF patients realized a 37% reduction in adverse cardiovascular events compared with the control group [111]111 Patel AN, Henry TD, Quyyumi AA, Schaer GL, Anderson RD, Toma C, ixCELL-DCM Investigators. Ixmyelocel-T for patients with ischaemic heart failure: a prospective randomised double-blind trial. Lancet. 2016;387(10036):2412-21..

Whereas, dysregulation of glucose metabolism may interfere with the positive effect of stem cells on heart failure. Among diabetic patients, CD26/DPP-4 on CD34+ cells failed to increase, suggesting that abnormal glucose metabolism and diabetes might impair the responsiveness of stem cells [112]112 Fadini GP, Albiero M, Vigili de Kreutzenberg S, Boscaro E, Cappellari R, Marescotti M, et al. Diabetes impairs stem cell and proangiogenic cell mobilization in humans. Diabetes Care. 2013;36(4):943-9.. In another research, CD34+ stem cells failed to take effect among patients with diabetes, while putting up great responsiveness in patients with insulin resistance, implying that the effect of stem cell therapy might be regulated by glucose metabolism [113]113 Vrtovec B, Sever M, Jensterle M, Poglajen G, Janez A, Kravos N, et al. Efficacy of CD34+ stem cell therapy in nonischemic dilated cardiomyopathy is absent in patients with diabetes but preserved in patients with insulin resistance. Stem Cell Transl Med. 2016;5(5):632-8.. Therefore, it will be a new challenge to explore the factors that intervening in the efficacy of stem cells in the treatment of heart failure.

Conclusion

A sound heart can make adaptive adjustments to cope with environmental changes according to the availability of the substrate. In heart failure, transcription of key enzymes involved in cardiac metabolism, REDOX, and changes in signal transduction give rise to cardiac disturbance of energy metabolism and impairment of metabolic flexibility. And put up with a harder challenge for survival. Hence, A better and more thorough understanding of the role and regulatory mechanisms of cardiac metabolism remodeling in HF may pave the way for the resolution of HF. Existing studies have proved that regulation of substrate utilization, oxidative phosphorylation, mitochondrial function, and cytothesis improved cardiac function and the quality of patients’ life. Taking all these factors into account, shifting the emphasis of pharmacotherapy from neurohormonal therapy to metabolic regulation is expected to be a key point in reducing the high readmission rate of HF. For decades to come, research ought to focus on clarifying the efficacy and safety of improving the cellular metabolic environment and seek systematic and feasible therapeutic regimens.

  • Funding
    This work was supported by the Young Talent Lifting Project of China Association of Chinese Medicine (No. CACM-2018-QNRC2-B04).

All manuscripts must conform to specific study guidelines

It is not applicable.

Statement

The present manuscript needs more than 50 references.

Research involving human participants and/or animals

The research involved no human participants and/or animals.

Informed consent

All authors agree to publish the article.

References

  • 1
    Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JG, Coats AJ, et al.; Authors/Task Force Members; document reviewers. 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 J Heart Fail. 2016;18(8):891-975.
  • 2
    Groenewegen A, Rutten FH, Mosterd A, Hoes AW Epidemiology of heart failure. Eur J Heart Fail. 2020;22(8):1342-56.
  • 3
    Hao G, Wang X, Chen Z, Zhang L, Zhang Y, Wei B, et al.; China Hypertension Survey Investigators. Prevalence of heart failure and left ventricular dysfunction in China: the China Hypertension Survey, 2012-2015. Eur J Heart Fail. 2019;21(11):1329-37.
  • 4
    GBD 2017 Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2018;392(10159):1789-858.
  • 5
    Heart Failure Group of Chinses Society of Cardiology of Chinses Medical Association, Chinses Heart Failure Association of Chinses Medical Doctor Association, Editorial Board of Chinses Journar of Cardioloy. 2018 China Guidelines for the diagnosis and treatment of heart failure. Chin J Cardiol. 2018; 2018(10) [in Chinese].
  • 6
    Dharmarajan K, Hsieh AF, Lin Z, Bueno H, Ross JS, Horwitz LI, et al. Diagnoses and timing of 30-day readmissions after hospitalization for heart failure, acute myocardial infarction, or pneumonia. JAMA. 2013;309(4):355-63.
  • 7
    Jones NR, Roalfe AK, Adoki I, Hobbs FDR, Taylor CJ Survival of patients with chronic heart failure in the community: a systematic review and meta-analysis. Eur J Heart Fail. 2019;21(11):1306-25.
  • 8
    Burchfield JS, Xie M, Hill JA Pathological ventricular remodeling: mechanisms: part 1 of 2. Circulation. 2013;128(4):388-400.
  • 9
    Stanley WC, Recchia FA, Lopaschuk GD Myocardial substrate metabolism in the normal and failing heart. Physiol Rev. 2005;85(3):1093-129.
  • 10
    Glatz JFC, Nabben M, Young ME, Schulze PC, Taegtmeyer H, Luiken JJFP Re-balancing cellular energy substrate metabolism to mend the failing heart. Biochim Biophys Acta Mol Basis Dis. 2020;1866(5):165579.
  • 11
    Glatz JFC, Luiken JJFP Dynamic role of the transmembrane glycoprotein CD36 (SR-B2) in cellular fatty acid uptake and utilization. J Lipid Res. 2018;59(7):1084-93.
  • 12
    Habets DD, Coumans WA, Voshol PJ, den Boer MA, Febbraio M, Bonen A, et al. AMPK-mediated increase in myocardial long-chain fatty acid uptake critically depends on sarcolemmal CD36. Biochem Biophys Res Commun. 2007;355(1):204-10.
  • 13
    Kalinowska A, Górski J, Harasim E, Harasiuk D, Bonen A, Chabowski A Differential effects of chronic, in vivo, PPAR's stimulation on the myocardial subcellular redistribution of FAT/CD36 and FABPpm. FEBS Lett. 2009;583(15):2527-34.
  • 14
    Smeets PJ, Teunissen BE, Willemsen PH, van Nieuwenhoven FA, Brouns AE, Janssen BJ, et al. Cardiac hypertrophy is enhanced in PPAR alpha-/- mice in response to chronic pressure overload. Cardiovasc Res. 2008;78(1):79-89.
  • 15
    Zhou N, Qi H, Liu J, Zhang G, Liu J, Liu N, et al. Deubiquitinase OTUD3 regulates metabolism homeostasis in response to nutritional stresses. Cell Metab. 2022;34(7):1023-1041.e8.
  • 16
    Shao D, Tian R Glucose transporters in cardiac metabolism and hypertrophy. Compr Physiol. 2015;6(1):331-51.
  • 17
    Badolia R, Ramadurai DKA, Abel ED, Ferrin P, Taleb I, Shankar TS, et al. The role of nonglycolytic glucose metabolism in myocardial recovery upon mechanical unloading and circulatory support in chronic heart failure. Circulation. 2020;142(3):259-74.
  • 18
    Dhar-Chowdhury P, Malester B, Rajacic P, Coetzee WA The regulation of ion channels and transporters by glycolytically derived ATP. Cell Mol Life Sci. 2007;64(23):3069-83.
  • 19
    Park S, Jeon JH, Min BK, Ha CM, Thoudam T, Park BY, Lee IK Role of the pyruvate dehydrogenase complex in metabolic remodeling: differential pyruvate dehydrogenase complex functions in metabolism. Diabetes Metab J. 2018;42(4):270-81.
  • 20
    Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, Smith A, et al. Nutritional ketosis alters fuel preference and thereby endurance performance in athletes. Cell Metab. 2016;24(2):256-68.
  • 21
    Puchalska P, Crawford PA Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 2017;25(2):262-84.
  • 22
    Falcão-Pires I, Hamdani N, Borbély A, Gavina C, Schalkwijk CG, van der Velden J, et al. Diabetes mellitus worsens diastolic left ventricular dysfunction in aortic stenosis through altered myocardial structure and cardiomyocyte stiffness. Circulation. 2011;124(10):1151-9.
  • 23
    Lopaschuk GD, Ussher JR, Folmes CD, Jaswal JS, Stanley WC Myocardial fatty acid metabolism in health and disease. Physiol Rev. 2010;90(1):207-58.
  • 24
    Cole MA, Abd Jamil AH, Heather LC, Murray AJ, Sutton ER, Slingo M, et al. On the pivotal role of PPARα in adaptation of the heart to hypoxia and why fat in the diet increases hypoxic injury. FASEB J. 2016;30(8):2684-97.
  • 25
    Finck BN, Lehman JJ, Leone TC, Welch MJ, Bennett MJ, Kovacs A, et al. The cardiac phenotype induced by PPARalpha overexpression mimics that caused by diabetes mellitus. J Clin Invest. 2002;109(1):121-30.
  • 26
    Sakamoto T, Matsuura TR, Wan S, Ryba DM, Kim JU, Won KJ, et al. A critical role for estrogen-related receptor signaling in cardiac maturation. Circ Res. 2020;126(12):1685-702.
  • 27
    Lasheras J, Pardo R, Velilla M, Poncelas M, Salvatella N, Simó R, et al. Cardiac-specific overexpression of ERRγ in mice induces severe heart dysfunction and early lethality. Int J Mol Sci. 2021;22(15):8047.
  • 28
    Allard MF, Schönekess BO, Henning SL, English DR, Lopaschuk GD. Contribution of oxidative metabolism and glycolysis to ATP production in hypertrophied hearts. Am J Physiol. 1994;267(2 Pt 2):H742-50.
  • 29
    Nascimben L, Ingwall JS, Lorell BH, Pinz I, Schultz V, Tornheim K, et al. Mechanisms for increased glycolysis in the hypertrophied rat heart. Hypertension. 2004;44(5):662-7.
  • 30
    Wang J, Xu J, Wang Q, Brainard RE, Watson LJ, Jones SP, et al. Reduced cardiac fructose 2,6 bisphosphate increases hypertrophy and decreases glycolysis following aortic constriction. PLoS One. 2013;8(1):e53951.
  • 31
    Gupte AA, Hamilton DJ, Cordero-Reyes AM, Youker KA, Yin Z, Estep JD, et al. Mechanical unloading promotes myocardial energy recovery in human heart failure. Circ Cardiovasc Genet. 2014;7(3):266-76.
  • 32
    Zhang L, Jaswal JS, Ussher JR, Sankaralingam S, Wagg C, Zaugg M, et al. Cardiac insulin-resistance and decreased mitochondrial energy production precede the development of systolic heart failure after pressure-overload hypertrophy. Circ Heart Fail. 2013;6(5):1039-48.
  • 33
    Aubert G, Martin OJ, Horton JL, Lai L, Vega RB, Leone TC, et al. The failing heart relies on ketone bodies as a fuel. Circulation. 2016;133(8):698-705.
  • 34
    Bedi KC Jr, Snyder NW, Brandimarto J, Aziz M, Mesaros C, Worth AJ, et al. Evidence for intramyocardial disruption of lipid metabolism and increased myocardial ketone utilization in advanced human heart failure. Circulation. 2016;133(8):706-16.
  • 35
    Ferrannini E, Mark M, Mayoux E CV protection in the EMPA-REG OUTCOME trial: A "thrifty substrate" hypothesis. Diabetes Care. 2016;39(7):1108-14.
  • 36
    Ho KL, Karwi QG, Wagg C, Zhang L, Vo K, Altamimi T, et al. Ketones can become the major fuel source for the heart but do not increase cardiac efficiency. Cardiovasc Res. 2021;117(4):1178-87.
  • 37
    Ho KL, Zhang L, Wagg C, Al Batran R, Gopal K, Levasseur J, et al. Increased ketone body oxidation provides additional energy for the failing heart without improving cardiac efficiency. Cardiovasc Res. 2019;115(11):1606-616.
  • 38
    Zhuang L, Jia K, Chen C, Li Z, Zhao J, Hu J, et al. DYRK1B-STAT3 drives cardiac hypertrophy and heart failure by impairing mitochondrial bioenergetics. Circulation. 2022;145(11):829-46.
  • 39
    Horton JL, Martin OJ, Lai L, Riley NM, Richards AL, Vega RB, et al. Mitochondrial protein hyperacetylation in the failing heart. JCI Insight. 2016;2(1):e84897.
  • 40
    Zhang X, Ji R, Liao X, Castillero E, Kennel PJ, Brunjes DL, et al. MicroRNA-195 regulates metabolism in failing myocardium via alterations in sirtuin 3 expression and mitochondrial protein acetylation. Circulation. 2018;137(19):2052-2067.
  • 41
    Lee CF, Chavez JD, Garcia-Menendez L, Choi Y, Roe ND, Chiao YA, et al. Normalization of NAD+ redox balance as a therapy for heart failure. Circulation. 2016;134(12):883-94.
  • 42
    Rutter MK, Parise H, Benjamin EJ, Levy D, Larson MG, Meigs JB, et al. Impact of glucose intolerance and insulin resistance on cardiac structure and function: sex-related differences in the Framingham Heart Study. Circulation. 2003;107(3):448-54.
  • 43
    Riehle C, Weatherford ET, Wende AR, Jaishy BP, Seei AW, McCarty NS, et al. Insulin receptor substrates differentially exacerbate insulin-mediated left ventricular remodeling. JCI Insight. 2020;5(6):e134920.
  • 44
    Ruiz-Velasco A, Zi M, Hille SS, Azam T, Kaur N, Jiang J, et al. Targeting mir128-3p alleviates myocardial insulin resistance and prevents ischemia-induced heart failure. Elife. 2020;9:e54298.
  • 45
    Taegtmeyer H, Beauloye C, Harmancey R, Hue L Insulin resistance protects the heart from fuel overload in dysregulated metabolic states. Am J Physiol Heart Circ Physiol. 2013;305(12):H1693-7.
  • 46
    Nakamura M, Liu T, Husain S, Zhai P, Warren JS, Hsu CP, et al. Glycogen synthase kinase-3α promotes fatty acid uptake and lipotoxic cardiomyopathy. Cell Metab. 2019;29(5):1119-1134.e12.
  • 47
    Chiu HC, Kovacs A, Blanton RM, Han X, Courtois M, Weinheimer CJ, et al. Transgenic expression of fatty acid transport protein 1 in the heart causes lipotoxic cardiomyopathy. Circ Res. 2005;96(2):225-33.
  • 48
    Okada K, Minamino T, Tsukamoto Y, Liao Y, Tsukamoto O, Takashima S, et al. Prolonged endoplasmic reticulum stress in hypertrophic and failing heart after aortic constriction: possible contribution of endoplasmic reticulum stress to cardiac myocyte apoptosis. Circulation. 2004;110(6):705-12.
  • 49
    Perman JC, Boström P, Lindbom M, Lidberg U, StÅhlman M, Hägg D, et al. The VLDL receptor promotes lipotoxicity and increases mortality in mice following an acute myocardial infarction. J Clin Invest. 2011;121(7):2625-40.
  • 50
    Xiong Z, Li Y, Zhao Z, Zhang Y, Man W, Lin J, et al. Mst1 knockdown alleviates cardiac lipotoxicity and inhibits the development of diabetic cardiomyopathy in db/db mice. Biochim Biophys Acta Mol Basis Dis. 2020;1866(8):165806.
  • 51
    Gupte RS, Vijay V, Marks B, Levine RJ, Sabbah HN, Wolin MS, et al. Upregulation of glucose-6-phosphate dehydrogenase and NAD(P)H oxidase activity increases oxidative stress in failing human heart. J Card Fail. 2007;13(6):497-506.
  • 52
    Gupte SA, Levine RJ, Gupte RS, Young ME, Lionetti V, Labinskyy V, et al. Glucose-6-phosphate dehydrogenase-derived NADPH fuels superoxide production in the failing heart. J Mol Cell Cardiol. 2006;41(2):340-9.
  • 53
    Jain M, Brenner DA, Cui L, Lim CC, Wang B, Pimentel DR, et al. Glucose-6-phosphate dehydrogenase modulates cytosolic redox status and contractile phenotype in adult cardiomyocytes. Circ Res. 2003;93(2):e9-16.
  • 54
    Champattanachai V, Marchase RB, Chatham JC Glucosamine protects neonatal cardiomyocytes from ischemia-reperfusion injury via increased protein-associated O-GlcNAc. Am J Physiol Cell Physiol. 2007;292(1):C178-87.
  • 55
    Liu J, Pang Y, Chang T, Bounelis P, Chatham JC, Marchase RB Increased hexosamine biosynthesis and protein O-GlcNAc levels associated with myocardial protection against calcium paradox and ischemia. J Mol Cell Cardiol. 2006;40(2):303-12.
  • 56
    Wang ZV, Deng Y, Gao N, Pedrozo Z, Li DL, Morales CR, et al. Spliced X-box binding protein 1 couples the unfolded protein response to hexosamine biosynthetic pathway. Cell. 2014;156(6):1179-92.
  • 57
    Kolijn D, Pabel S, Tian Y, Lódi M, Herwig M, Carrizzo A, et al. Empagliflozin improves endothelial and cardiomyocyte function in human heart failure with preserved ejection fraction via reduced pro-inflammatory-oxidative pathways and protein kinase Gα oxidation. Cardiovasc Res. 2021;117(2):495-507.
  • 58
    Lehnart SE, Mongillo M, Bellinger A, Lindegger N, Chen BX, Hsueh W, et al. Leaky Ca2+ release channel/ryanodine receptor 2 causes seizures and sudden cardiac death in mice. J Clin Invest. 2008;118(6):2230-45.
  • 59
    Uchinoumi H, Yang Y, Oda T, Li N, Alsina KM, Puglisi JL, et al. CaMKII-dependent phosphorylation of RyR2 promotes targetable pathological RyR2 conformational shift. J Mol Cell Cardiol. 2016;98:62-72.
  • 60
    Pinali C, Bennett H, Davenport JB, Trafford AW, Kitmitto A Three-dimensional reconstruction of cardiac sarcoplasmic reticulum reveals a continuous network linking transverse-tubules: this organization is perturbed in heart failure. Circ Res. 2013;113(11):1219-30.
  • 61
    Vila-Petroff M, Salas MA, Said M, Valverde CA, Sapia L, Portiansky E, et al. CaMKII inhibition protects against necrosis and apoptosis in irreversible ischemia-reperfusion injury. Cardiovasc Res. 2007;73(4):689-98.
  • 62
    Zhang T, Maier LS, Dalton ND, Miyamoto S, Ross J Jr, Bers DM, et al. The deltaC isoform of CaMKII is activated in cardiac hypertrophy and induces dilated cardiomyopathy and heart failure. Circ Res. 2003;92(8):912-9.
  • 63
    Zhang T, Zhang Y, Cui M, Jin L, Wang Y, Lv F, et al. CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress-induced myocardial necroptosis. Nat Med. 2016;22(2):175-82.
  • 64
    Bristow MR Changes in myocardial and vascular receptors in heart failure. J Am Coll Cardiol. 1993;22(4 Suppl A):61A-71A.
  • 65
    Fowler MB, Laser JA, Hopkins GL, Minobe W, Bristow MR Assessment of the beta-adrenergic receptor pathway in the intact failing human heart: progressive receptor down-regulation and subsensitivity to agonist response. Circulation. 1986;74(6):1290-302.
  • 66
    Lamba S, Abraham WT Alterations in adrenergic receptor signaling in heart failure. Heart Fail Rev. 2000;5(1):7-16.
  • 67
    Bristow MR, Ginsburg R, Umans V, Fowler M, Minobe W, Rasmussen R, et al. Beta 1- and beta 2-adrenergic-receptor subpopulations in nonfailing and failing human ventricular myocardium: coupling of both receptor subtypes to muscle contraction and selective beta 1-receptor down-regulation in heart failure. Circ Res. 1986;59(3):297-309.
  • 68
    Trappanese DM, Liu Y, McCormick RC, Cannavo A, Nanayakkara G, Baskharoun MM, et al. Chronic β1-adrenergic blockade enhances myocardial β3-adrenergic coupling with nitric oxide-cGMP signaling in a canine model of chronic volume overload: new insight into mechanisms of cardiac benefit with selective β1-blocker therapy. Basic Res Cardiol. 2015;110(1):456.
  • 69
    Finlin BS, Memetimin H, Zhu B, Confides AL, Vekaria HJ, El Khouli RH, et al. The β3-adrenergic receptor agonist mirabegron improves glucose homeostasis in obese humans. J Clin Invest. 2020;130(5):2319-31.
  • 70
    Kamiya M, Asai K, Maejima Y, Shirakabe A, Murai K, Noma S, et al. β3-Adrenergic receptor agonist prevents diastolic dysfunction in an angiotensin II-induced cardiomyopathy mouse model. J Pharmacol Exp Ther. 2021;376(3):473-481.
  • 71
    Pun-García A, Clemente-Moragón A, Villena-Gutierrez R, Gómez M, Sanz-Rosa D, Díaz-Guerra A, et al. Beta-3 adrenergic receptor overexpression reverses aortic stenosis-induced heart failure and restores balanced mitochondrial dynamics. Basic Res Cardiol. 2022;117(1):62.
  • 72
    Li H, Liu Y, Huang H, Tang Y, Yang B, Huang C Activation of β3-adrenergic receptor inhibits ventricular arrhythmia in heart failure through calcium handling. Tohoku J Exp Med. 2010;222(3):167-74.
  • 73
    Yang W, Wei X, Su X, Shen Y, Jin W, Fang Y. Depletion of β3-adrenergic receptor induces left ventricular diastolic dysfunction via potential regulation of energy metabolism and cardiac contraction. Gene. 2019;697:1-10.
  • 74
    Montaigne D, Butruille L, Staels B PPAR control of metabolism and cardiovascular functions. Nat Rev Cardiol. 2021;18(12):809-823.
  • 75
    Musa H, Tellez JO, Chandler NJ, Greener ID, Maczewski M, Mackiewicz U, et al. P2 purinergic receptor mRNA in rat and human sinoatrial node and other heart regions. Naunyn Schmiedebergs Arch Pharmacol. 2009;379(6):541-9.
  • 76
    Yang SM, Liu J, Li CX Intermedin protects against myocardial ischemia-reperfusion injury in hyperlipidemia rats. Genet Mol Res. 2014;13(4):8309-19.
  • 77
    Nishida M, Sato Y, Uemura A, Narita Y, Tozaki-Saitoh H, Nakaya M, et al. P2Y6 receptor-Galpha12/13 signalling in cardiomyocytes triggers pressure overload-induced cardiac fibrosis. EMBO J. 2008;27(23):3104-15.
  • 78
    Voros G, Ector J, Garweg C, Droogne W, Van Cleemput J, Peersman N, et al. Increased cardiac uptake of ketone bodies and free fatty acids in human heart failure and hypertrophic left ventricular remodeling. Circ Heart Fail. 2018;11(12):e004953.
  • 79
    Du Z, Shen A, Huang Y, Su L, Lai W, Wang P, et al. 1H-NMR-based metabolic analysis of human serum reveals novel markers of myocardial energy expenditure in heart failure patients. PLoS One. 2014;9(2):e88102.
  • 80
    Zordoky BN, Sung MM, Ezekowitz J, Mandal R, Han B, Bjorndahl TC, Alberta HEART. Metabolomic fingerprint of heart failure with preserved ejection fraction. PLoS One. 2015;10(5):e0124844.
  • 81
    Wu CK, Lee JK, Hsu JC, Su MM, Wu YF, Lin TT, et al. Myocardial adipose deposition and the development of heart failure with preserved ejection fraction. Eur J Heart Fail. 2020;22(3):445-54.
  • 82
    Dávila-Román VG, Vedala G, Herrero P, de las Fuentes L, Rogers JG, Kelly DP, et al. Altered myocardial fatty acid and glucose metabolism in idiopathic dilated cardiomyopathy. J Am Coll Cardiol. 2002;40(2):271-7.
  • 83
    Doenst T, Pytel G, Schrepper A, Amorim P, Färber G, Shingu Y, et al. Decreased rates of substrate oxidation ex vivo predict the onset of heart failure and contractile dysfunction in rats with pressure overload. Cardiovasc Res. 2010;86(3):461-70.
  • 84
    Schönfeld P, Wojtczak L Fatty acids decrease mitochondrial generation of reactive oxygen species at the reverse electron transport but increase it at the forward transport. Biochim Biophys Acta. 2007;1767(8):1032-40.
  • 85
    Marzilli M, Vinereanu D, Lopaschuk G, Chen Y, Dalal JJ, Danchin N, et al. Trimetazidine in cardiovascular medicine. Int J Cardiol. 2019;293:39-44.
  • 86
    Shu H, Hang W, Peng Y, Nie J, Wu L, Zhang W, et al. Trimetazidine attenuates heart failure by improving myocardial metabolism via AMPK. Front Pharmacol. 2021;12:707399.
  • 87
    Shu H, Peng Y, Hang W, Zhou N, Wang DW. Trimetazidine in heart failure. Front Pharmacol. 2020;11:569132.
  • 88
    Nie J, Duan Q, He M, Li X, Wang B, Zhou C, et al. Ranolazine prevents pressure overload-induced cardiac hypertrophy and heart failure by restoring aberrant Na+ and Ca2+ handling. J Cell Physiol. 2019;234(7):11587-601.
  • 89
    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(6):369-78.
  • 90
    Sharma V, Dhillon P, Wambolt R, Parsons H, Brownsey R, Allard MF, et al. Metoprolol improves cardiac function and modulates cardiac metabolism in the streptozotocin-diabetic rat. Am J Physiol Heart Circ Physiol. 2008, 294(4):H1609-20.
  • 91
    Ribeiro PAB, Normandin E, Meyer P, Juneau M, White M, Nigam A, et al. Beta-blocker type effect on substrate oxidation during HIIE in heart failure patients: pilot data. Arq Bras Cardiol. 2019;112(3):304-308.
  • 92
    Pradhan AD, Paynter NP, Everett BM, Glynn RJ, Amarenco P, Elam M, et al. Rationale and design of the pemafibrate to reduce cardiovascular outcomes by reducing triglycerides in patients with diabetes (PROMINENT) study. Am Heart J. 2018;206:80-93.
  • 93
    Crowley MJ, Diamantidis CJ, McDuffie JR, Cameron CB, Stanifer JW, Mock CK, et al. Clinical outcomes of metformin use in populations with chronic kidney disease, congestive heart failure, or chronic liver disease: a systematic review. Ann Intern Med. 2017;166(3):191-200.
  • 94
    Mather KJ, Verma S, Anderson TJ Improved endothelial function with metformin in type 2 diabetes mellitus. J Am Coll Cardiol. 2001;37(5):1344-50.
  • 95
    Ma T, Tian X, Zhang B, Li M, Wang Y, Yang C, et al. Low-dose metformin targets the lysosomal AMPK pathway through PEN2. Nature. 2022;603(7899):159-165.
  • 96
    Garcia-Ropero A, Santos-Gallego CG, Zafar MU, Badimon JJ Metabolism of the failing heart and the impact of SGLT2 inhibitors. Expert Opin Drug Metab Toxicol. 2019;15(4):275-85.
  • 97
    Santos-Gallego CG, Requena-Ibanez JA, San Antonio R, Ishikawa K, Watanabe S, Picatoste B, et al. Empagliflozin ameliorates adverse left ventricular remodeling in nondiabetic heart failure by enhancing myocardial energetics. J Am Coll Cardiol. 2019;73(15):1931-44.
  • 98
    Jiang K, Xu Y, Wang D, Chen F, Tu Z, Qian J, et al. Cardioprotective mechanism of SGLT2 inhibitor against myocardial infarction is through reduction of autosis. Protein Cell. 2022;13(5):336-59.
  • 99
    McMurray JJV, DeMets DL, Inzucchi SE, Køber L, Kosiborod MN, Langkilde AM, et al.; DAPA-HF committees and investigators. A trial to evaluate the effect of the sodium-glucose co-transporter 2 inhibitor dapagliflozin on morbidity and mortality in patients with heart failure and reduced left ventricular ejection fraction (DAPA-HF). Eur J Heart Fail. 2019;21(5):665-75.
  • 100
    Yurista SR, Matsuura TR, Silljé HHW, Nijholt KT, McDaid KS, Shewale SV, et al. Ketone ester treatment improves cardiac function and reduces pathologic remodeling in preclinical models of heart failure. Circ Heart Fail. 2021;14(1):e007684.
  • 101
    Kim SR, Lee SG, Kim SH, Kim JH, Choi E, Cho W, et al. SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nat Commun. 2020;11(1):2127.
  • 102
    Ayer A, Macdonald P, Stocker R CoQ₁₀ function and role in heart failure and ischemic heart disease. Annu Rev Nutr. 2015;35:175-213.
  • 103
    Lei L, Liu Y Efficacy of coenzyme Q10 in patients with cardiac failure: a meta-analysis of clinical trials. BMC Cardiovasc Disord. 2017;17(1):196.
  • 104
    Diguet N, Trammell SAJ, Tannous C, Deloux R, Piquereau J, Mougenot N, et al. Nicotinamide riboside preserves cardiac function in a mouse model of dilated cardiomyopathy. Circulation. 2018;137(21):2256-73.
  • 105
    Hong W, Mo F, Zhang Z, Huang M, Wei X Nicotinamide mononucleotide: a promising molecule for therapy of diverse diseases by targeting NAD+ metabolism. Front Cell Dev Biol. 2020;8:246.
  • 106
    Tong D, Schiattarella GG, Jiang N, Altamirano F, Szweda PA, Elnwasany A, et al. NAD+ repletion reverses heart failure with preserved ejection fraction. Circ Res. 2021;128(11):1629-41.
  • 107
    Gnecchi M, He H, Liang OD, Melo LG, Morello F, Mu H, et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med. 2005;11(4):367-8.
  • 108
    Mirotsou M, Jayawardena TM, Schmeckpeper J, Gnecchi M, Dzau VJ Paracrine mechanisms of stem cell reparative and regenerative actions in the heart. J Mol Cell Cardiol. 2011;50(2):280-9
  • 109
    Bartunek J, Behfar A, Dolatabadi D, Vanderheyden M, Ostojic M, Dens J, et al. Cardiopoietic stem cell therapy in heart failure: the C-CURE (Cardiopoietic stem Cell therapy in heart failURE) multicenter randomized trial with lineage-specified biologics. J Am Coll Cardiol. 2013;61(23):2329-38.
  • 110
    Bartolucci J, Verdugo FJ, González PL, Larrea RE, Abarzua E, Goset C, et al. Safety and efficacy of the intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure: a phase 1/2 randomized controlled trial (RIMECARD trial [Randomized clinical trial of intravenous infusion umbilical cord mesenchymal stem cells on cardiopathy]). Circ Res. 2017;121(10):1192-204.
  • 111
    Patel AN, Henry TD, Quyyumi AA, Schaer GL, Anderson RD, Toma C, ixCELL-DCM Investigators. Ixmyelocel-T for patients with ischaemic heart failure: a prospective randomised double-blind trial. Lancet. 2016;387(10036):2412-21.
  • 112
    Fadini GP, Albiero M, Vigili de Kreutzenberg S, Boscaro E, Cappellari R, Marescotti M, et al. Diabetes impairs stem cell and proangiogenic cell mobilization in humans. Diabetes Care. 2013;36(4):943-9.
  • 113
    Vrtovec B, Sever M, Jensterle M, Poglajen G, Janez A, Kravos N, et al. Efficacy of CD34+ stem cell therapy in nonischemic dilated cardiomyopathy is absent in patients with diabetes but preserved in patients with insulin resistance. Stem Cell Transl Med. 2016;5(5):632-8.
  • 114
    Tuunanen H, Engblom E, Naum A, Någren K, Hesse B, Airaksinen KE, et al. Free fatty acid depletion acutely decreases cardiac work and efficiency in cardiomyopathic heart failure. Circulation. 2006;114(20):2130-7.
  • 115
    Taylor M, Wallhaus TR, Degrado TR, Russell DC, Stanko P, Nickles RJ, 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(1):55-62.
  • 116
    Tuunanen H, Engblom E, Naum A, Scheinin M, Någren K, Airaksinen J, et al. Decreased myocardial free fatty acid uptake in patients with idiopathic dilated cardiomyopathy: evidence of relationship with insulin resistance and left ventricular dysfunction. J Card Fail. 2006;12(8):644-52.
  • 117
    Weitzel LB, Ambardekar AV, Brieke A, Cleveland JC, Serkova NJ, Wischmeyer PE, et al. Left ventricular assist device effects on metabolic substrates in the failing heart. PLoS One. 2013;8(4):e60292.
  • 118
    Yokokawa T, Yoshihisa A, Kanno Y, Abe S, Misaka T, Yamada S, et al. Circulating acetoacetate is associated with poor prognosis in heart failure patients. Int J Cardiol Heart Vasc. 2019;25:100432.
  • 119
    Rosenblatt-Velin N, Montessuit C, Papageorgiou I, Terrand J, Lerch R Postinfarction heart failure in rats is associated with upregulation of GLUT-1 and downregulation of genes of fatty acid metabolism. Cardiovasc Res. 2001;52(3):407-16.
  • 120
    Kato T, Niizuma S, Inuzuka Y, Kawashima T, Okuda J, Tamaki Y, et al. Analysis of metabolic remodeling in compensated left ventricular hypertrophy and heart failure. Circ Heart Fail. 2010;3(3):420-30.
  • 121
    Byrne NJ, Levasseur J, Sung MM, Masson G, Boisvenue J, Young ME, et al. Normalization of cardiac substrate utilization and left ventricular hypertrophy precede functional recovery in heart failure regression. Cardiovasc Res. 2016;110(2):249-57.
  • 122
    Sung MM, Byrne NJ, Robertson IM, Kim TT, Samokhvalov V, Levasseur J, et al. Resveratrol improves exercise performance and skeletal muscle oxidative capacity in heart failure. Am J Physiol Heart Circ Physiol. 2017;312(4):H842-H853.
  • 123
    Masoud WG, Ussher JR, Wang W, Jaswal JS, Wagg CS, Dyck JR, et al. Failing mouse hearts utilize energy inefficiently and benefit from improved coupling of glycolysis and glucose oxidation. Cardiovasc Res. 2014;101(1):30-8.
  • 124
    Sankaralingam S, Abo Alrob O, Zhang L, Jaswal JS, Wagg CS, Fukushima A, et al. Lowering body weight in obese mice with diastolic heart failure improves cardiac insulin sensitivity and function: implications for the obesity paradox. Diabetes. 2015;64(5):1643-57.
  • 125
    Zhabyeyev P, Gandhi M, Mori J, Basu R, Kassiri Z, Clanachan A, et al. Pressure-overload-induced heart failure induces a selective reduction in glucose oxidation at physiological afterload. Cardiovasc Res. 2013;97(4):676-85.
  • 126
    Fillmore N, Levasseur JL, Fukushima A, Wagg CS, Wang W, Dyck JRB, et al. Uncoupling of glycolysis from glucose oxidation accompanies the development of heart failure with preserved ejection fraction. Mol Med. 2018;24(1):3.
  • 127
    Nagao M, Toh R, Irino Y, Mori T, Nakajima H, Hara T, et al. β-Hydroxybutyrate elevation as a compensatory response against oxidative stress in cardiomyocytes. Biochem Biophys Res Commun. 2016;475(4):322-8.
  • 128
    Gao D, Ning N, Niu X, Hao G, Meng Z Trimetazidine: a meta-analysis of randomised controlled trials in heart failure. Heart. 2011;97(4):278-86.
  • 129
    Fragasso G, Salerno A, Lattuada G, Cuko A, Calori G, Scollo A, et al. Effect of partial inhibition of fatty acid oxidation by trimetazidine on whole body energy metabolism in patients with chronic heart failure. Heart. 2011;97(18):1495-500.
  • 130
    Maier LS, Layug B, Karwatowska-Prokopczuk E, Belardinelli L, Lee S, Sander J, et al. RAnoLazIne for the treatment of diastolic heart failure in patients with preserved ejection fraction: the RALI-DHF proof-of-concept study. JACC Heart Fail. 2013;1(2):115-22.
  • 131
    Murray GL, Colombo J Ranolazine preserves and improves left ventricular ejection fraction and autonomic measures when added to guideline-driven therapy in chronic heart failure. Heart Int. 2014;9(2):66-73.
  • 132
    Hennuyer N, Duplan I, Paquet C, Vanhoutte J, Woitrain E, Touche V, et al. The novel selective PPARα modulator (SPPARMα) pemafibrate improves dyslipidemia, enhances reverse cholesterol transport and decreases inflammation and atherosclerosis. Atherosclerosis. 2016;249:200-8.
  • 133
    Eurich DT, Weir DL, Majumdar SR, Tsuyuki RT, Johnson JA, Tjosvold L, et al. Comparative safety and effectiveness of metformin in patients with diabetes mellitus and heart failure: systematic review of observational studies involving 34,000 patients. Circ Heart Fail. 2013;6(3):395-402.
  • 134
    Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, et al.; EMPEROR-reduced trial investigators. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med. 2020;383(15):1413-24.
  • 135
    Berg DD, Jhund PS, Docherty KF, Murphy SA, Verma S, Inzucchi SE, et al. Time to clinical benefit of dapagliflozin and significance of prior heart failure hospitalization in patients with heart failure with reduced ejection fraction. JAMA Cardiol. 2021;6(5):499-507.
  • 136
    Bhatt DL, Szarek M, Steg PG, Cannon CP, Leiter LA, McGuire DK, et al.; SOLOIST-WHF trial investigators. sotagliflozin in patients with diabetes and recent worsening heart failure. N Engl J Med. 2021;384(2):117-28.
  • 137
    Mortensen SA, Rosenfeldt F, Kumar A, Dolliner P, Filipiak KJ, Pella D, et al.; Q-SYMBIO study investigators. The effect of coenzyme Q10 on morbidity and mortality in chronic heart failure: results from Q-SYMBIO: a randomized double-blind trial. JACC Heart Fail. 2014;2(6):641-9.
  • 138
    Zhou B, Wang DD, Qiu Y, Airhart S, Liu Y, Stempien-Otero A, O'Brien KD, Tian R Boosting NAD level suppresses inflammatory activation of PBMCs in heart failure. J Clin Invest. 2020;130(11):6054-63.

Publication Dates

  • Publication in this collection
    01 Sept 2023
  • Date of issue
    2023

History

  • Received
    07 Jan 2023
  • Reviewed
    15 July 2023
  • Accepted
    18 July 2023
Faculdade de Medicina / USP Rua Dr Ovídio Pires de Campos, 225 - 6 and., 05403-010 São Paulo SP - Brazil, Tel.: (55 11) 2661-6235 - São Paulo - SP - Brazil
E-mail: clinics@hc.fm.usp.br