Skip to main content

The role of myocardial energy metabolism perturbations in diabetic cardiomyopathy: from the perspective of novel protein post-translational modifications

Abstract

Diabetic cardiomyopathy (DbCM), a significant chronic complication of diabetes, manifests as myocardial hypertrophy, fibrosis, and other pathological alterations that substantially impact cardiac function and elevate the risk of cardiovascular diseases and patient mortality. Myocardial energy metabolism disturbances in DbCM, encompassing glucose, fatty acid, ketone body and lactate metabolism, are crucial factors that contribute to the progression of DbCM. In recent years, novel protein post-translational modifications (PTMs) such as lactylation, β-hydroxybutyrylation, and succinylation have been demonstrated to be intimately associated with the myocardial energy metabolism process, and in conjunction with acetylation, they participate in the regulation of protein activity and gene expression activity in cardiomyocytes. This review examines the epigenetic pathogenesis of DbCM, primarily focusing on myocardial energy metabolism perturbations and novel PTMs associated with them. It provides a detailed analysis of the mechanisms of these novel PTMs in DbCM to enhance the understanding of DbCM pathophysiology and establish a theoretical foundation for the development of new treatment strategies for DbCM.

Introduction

Diabetic cardiomyopathy (DbCM) is a significant chronic complication of diabetes, primarily characterized by myocardial hypertrophy [1], interstitial fibrosis [2], structural changes in sarcomeres [3], and coronary microangiopathy [4]. These alterations can lead to changes in cardiac cavity volume, reduced cardiac compliance, and impaired cardiac function [5]. Moreover, DbCM has a causal relationship with other cardiovascular diseases, such as atherosclerosis, hypertension, myocardial infarction, and heart failure [6]. This interaction not only complicates treatment strategies for DbCM but also greatly increases patient mortality and poses a serious threat to the overall health of patients.

The pathogenesis of DbCM is complex, and disturbance of myocardial energy substrate metabolism [7], involving glucose, fatty acids, and ketone bodies, is one of the main driving forces for the pathogenesis and development of DbCM [8]. First, disturbance of myocardial energy metabolism can directly affect the contractile and diastolic functions of cardiomyocytes. In addition, metabolic intermediates produced during metabolic disorders may act as downstream signaling molecules to activate or inhibit specific signaling pathways that influence cardiomyocyte proliferation, apoptosis, and fibrosis. Finally, metabolic disorders may also regulate the pathophysiological processes of DbCM through epigenetic mechanisms [9].

Epigenetics is a pivotal field in biology that investigates heritable alterations in biological phenotypes that occur without changes to the DNA sequence [10]. Among its various components, protein post-translational modifications (PTMs) are of particular significance. PTMs refer to the acylation modifications of proteins on amino acid residues, such as lysine, after translation. This process is dynamically regulated by specific writing enzymes and erasing enzymes. PTMs influence protein structure, stability, and function. On the one hand, PTMs on histones regulate the stability of histone binding to DNA, thereby affecting gene transcriptional activation in the relevant promoter region. In contrast, PTMs on non-histones regulate enzyme activity, functional localization, and protein–protein interactions [11].

The roles of previously discovered PTMs such as acetylation (Kac) [12] in DbCM have been extensively investigated. In recent years, the gradual application of high-precision mass spectrometry (MS) and the continuous development of specific antibodies have led to the identification and characterization of various novel PTMs [Fig. 1]. However, our understanding of their roles in biological regulation is limited. The formation of certain novel PTMs requires specific energy metabolic intermediates as acyl donors [13]. For instance, lactylation (Kla) uses lactate produced during glycolysis as a substrate [14], β-hydroxybutyrylation (Kbhb) employs the ketone body β-hydroxybutyrate (β-HB) as a substrate [15], and succinylation (Ksucc) relies on succinyl-CoA in the mitochondrial tricarboxylic acid (TCA) cycle as a substrate [16]. Recent studies have shown that these novel PTMs are involved in the metabolic regulation and expression of certain key genes in cardiomyocytes, thereby directly or indirectly influencing the occurrence and progression of DbCM [17].

Fig. 1
figure 1

The discovery process and molecular structure of PTMs related to metabolic disturbance in DbCM. With the development of acyl antibodies and continuous improvement in the accuracy of MS detection, many PTMs related to metabolic disturbance of DbCM cardiomyocytes have been discovered in recent years, and the red part of their molecular structure is the corresponding acyl modification group

This review aimed to summarize the epigenetic effects of myocardial energy metabolism disorders in DbCM, focusing on the regulatory mechanism from the perspective of novel PTMs. First, we briefly analyzed the changes in the energy substrate metabolism observed in the DbCM myocardium. We then outline the relationship between DbCM and Kac. Finally, we describe in detail the formation process of new PTMs that are closely related to energy metabolism, as well as their roles and mechanisms in DbCM. This review provides a crucial theoretical basis for the elucidation of the epigenetic pathogenesis of DbCM and the search for new therapeutic targets, and has great significance for strengthening the prevention and treatment of DbCM.

Disturbance of energy metabolism in DbCM

The heart has the highest oxygen consumption in the body [18] and demonstrates remarkable flexibility in selecting substrates for energy metabolism in healthy adults [19]. Under normal circumstances, adenosine triphosphate (ATP) required by the myocardium is primarily derived from the β-oxidation of fatty acids and aerobic oxidation of glucose [20]. Approximately 60–90% of the energy supply is comes from fatty acids, while the remaining 10–40% is derived from glucose oxidation, with the contributions of amino acids and ketone bodies being relatively minor. However, in the context of DbCM, the metabolic processes of cardiomyocytes involving energy substrates such as glucose, fatty acids, and ketone bodies undergo significant alterations [21]. These changes adversely affect the overall function of the heart through complex mechanisms.

Glucose metabolism

Clinical studies have demonstrated that myocardial glucose metabolism is significantly impaired in patients with type 2 diabetes [22]. A study utilizing advanced hyperpolarized magnetic resonance spectroscopy revealed a marked reduction in the ability of pyruvate, produced through cytoplasmic glucose metabolism in cardiomyocytes to enter the mitochondria for TCA cycle metabolism [23]. Furthermore, diabetic patients frequently exhibit insulin resistance, leading to the downregulation of the glucose transporter GLUT4 on the surface of cardiomyocytes, consequently diminishing glucose uptake and utilization [24, 25]. In the context of diabetes, increased PPARα activity in cardiomyocytes may further inhibit oxidative glucose utilization by promoting Pdk4 transcription [26, 27]. This decline in glucose utilization not only compromises the energy supply efficiency of cardiomyocytes, but may also precipitate a series of metabolic abnormalities, such as heightened hyperglycemic toxicity, which can lead to further impairment of cardiomyocyte function.

Fatty acid metabolism

Cardiomyocytes of diabetic patients partially compensate for their impaired glucose metabolism by significantly enhancing fatty acid metabolism [22]. This compensatory mechanism operates effectively because of the markedly increased expression levels of membrane transporters, such as CD36, in cardiomyocytes, which facilitates the uptake of free fatty acids by the heart [28]. In the context of DbCM, abnormal activation of the transcription factor PPARα further induces the simultaneous upregulation of multiple downstream genes associated with fatty acid uptake, oxidation, and storage, thereby effectively coordinating the enhancement of myocardial fatty acid metabolism [26, 29]. Concurrently, elevated levels of fatty acid metabolic intermediates may directly inhibit the activity of glucose metabolic enzymes through allosteric regulation or covalent modification, resulting in decreased glucose utilization. This shift in the metabolic pattern triggers the gradual accumulation of fatty acids in cardiomyocytes, ultimately leading to increased lipid toxicity and further damage to the structure and function of cardiomyocytes [30]. Additionally, because the oxidation of fatty acids requires greater oxygen consumption, this process can increase the oxygen demand of cardiomyocytes, potentially resulting in a relatively hypoxic state of the myocardium.

Ketone body metabolism

Ketone bodies include acetoacetic acid, β-HB, and acetone, and the total amount of β-HB in human serum accounts for approximately 70% of all ketone bodies [31]. Ketone bodies exhibit greater efficiency in ATP production than glucose and fatty acid oxidation, while requiring relatively little oxygen. Thus, they are considered potential clean energy sources in the context of diabetes. However, research on ketone body metabolism in the diabetic heart is still insufficient, with only a limited number of conflicting experimental results. For instance, some clinical studies and animal experiments have indicated that the level of β-HB in the myocardium is elevated in cases of diabetic cardiomyopathy. Concurrently, the expression and activity of the regulatory enzyme BDH1, which catalyzes the production of β-HB, are significantly reduced [32]. Furthermore, in a study using a rat model of diabetic cardiomyopathy, an increased utilization of acetoacetic acid, another ketone body, was observed in the myocardium, a phenomenon closely linked to heightened activity of the acetoacetic acid-metabolizing enzyme SCOT [33, 34]. Recent studies have demonstrated that ketone bodies can inhibit the activation and release of the NLRP3 inflammasome in human monocytes, protect vascular endothelial function in mice with heart failure, and mitigate oxidative stress damage in sepsis-associated cardiomyopathy [35,36,37]. Therefore, ketone bodies may also contribute to delaying the progression of DbCM through these mechanisms.

Lactate metabolism

Lactate metabolism plays a crucial role in energy acquisition of cardiomyocytes. During the glycolysis of cardiomyocytes, glucose is transported to cells by glucose transporters to convert into two molecules of pyruvate. On the one hand, pyruvate in the cytoplasm can be transported to the mitochondria via mitochondrial pyruvate carriers, which are then converted to acetyl-CoA by pyruvate dehydrogenase and enter the TCA cycle. On the other hand, pyruvate can also be converted to lactate by lactate dehydrogenase (LDH) in the cytoplasm.

In diabetic hyperglycemia, the process of cardiomyocyte glycolysis is accelerated, but due to mitochondrial dysfunction, especially the activity of pyruvate carrier (MPC) is decreased. As a result, pyruvate cannot effectively enter mitochondria for oxidative phosphorylation [38], which promotes more conversion of pyruvate to lactate. In addition, lactate utilization by DbCM cardiomyocytes decreased. This change in substrate preference also resulted in increased lactate levels in cardiomyocytes [39]. In addition, the expression of monocarboxylate transporter 4 (MCT4) in cardiomyocytes is upregulated, which leads to increased lactate outflow, disrupts the lactate-pyruvate balance in cardiomyocytes, and triggers oxidative stress and inflammation, further aggravating myocardial damage [40].

In conclusion, DbCM is closely associated with multiple alterations in the metabolism of glucose, fatty acids, ketone bodies and lactate within cardiomyocytes, which collectively affect cardiac function through complex interactions. These metabolic changes not only diminish energy supply efficiency, but also initiate a series of pathological processes, including oxidative stress, inflammatory responses, and calcium homeostasis imbalances [41], ultimately resulting in cardiomyocyte damage and apoptosis. Long-term chronic hyperglycemia can induce oxidative stress, resulting in increased production of reactive oxygen species (ROS) that damage cardiomyocytes and promote myocardial fibrosis and heart failure [42]. Additionally, prolonged hyperglycemia can stimulate the immune system to generate a persistent inflammatory response, contributing to cardiomyocyte hypertrophy, fibrosis, and diastolic dysfunction. Furthermore, enhanced fatty acid oxidation may lead to mitochondrial dysfunction in cardiomyocytes, particularly through abnormal expression of the mitochondrial calcium homeostasis protein (MCU). This abnormality results in reduced mitochondrial calcium uptake, further exacerbating mitochondrial dysfunction in cardiomyocytes [43]. This cascade of events may lead to severe heart failure and other adverse outcomes. The disruption of cardiomyocyte energy metabolism in DbCM presents a potential therapeutic target. Research has demonstrated that empagliflozin can enhance the rate of glucose oxidation in the myocardium of db/db mice [44]. Additionally, other studies indicate that empagliflozin can decrease the expression of the fatty acid β-oxidation regulatory enzyme CPT1b in diabetic rat cardiomyocytes, consequently diminishing fatty acid utilization and ameliorating the energy metabolism disorder in cardiomyocytes [45].

Furthermore, in the context of DbCM, the protein PTMs formed by energy metabolism intermediates in cardiomyocytes may also exhibit corresponding changes, such as Kac, Kla, Kbhb, and Ksucc, which contribute to the emergence of epigenetic phenomena like “metabolic memory” among these modifications [46]. The relationship between Kac and DbCM, which was initially discovered, has been extensively studied. A systematic summary and in-depth exploration of the regulatory roles of other novel PTMs in the onset and progression of DbCM, along with the implementation of corresponding intervention strategies, holds significant practical importance for the prevention and treatment of DbCM.

Overview of the relationship between acetylation and DbCM

Acetyl-CoA, a key acyl group donor, plays an important role in the formation of Kac [47]. Acetyl-CoA is also a hub of energy metabolism in cardiomyocytes. In the oxidative metabolism of myocardial fatty acids, acetyl-CoA is mainly produced during the β-oxidation of fatty acids and then enters the TCA cycle [48]. In glucose metabolism, pyruvate, the end product of the glycolysis pathway, can be converted into acetyl-CoA after entering the mitochondria and further participates in the TCA cycle to provide energy for cardiomyocytes. In addition, in ketone body metabolism, acetoacetic acid can also be converted into acetyl-CoA, and then participate in the regulation of energy metabolism. Three types of enzymes are involved in the regulatory mechanism of Kac: writing enzymes (GCN5, etc.) [49, 50], erasing enzymes (HDAC1-5, SIRT1-7, etc.), and the corresponding reading enzymes (YEATS, etc.) [51, 52].

In the context of diabetes, the disruption of energy metabolism in cardiomyocytes results in an imbalance between the production and utilization of acetyl-CoA. This imbalance influences the pathogenesis of DbCM by regulating Kac levels of both histone and non-histone proteins. For instance, an experiment conducted in streptozotocin (STZ)-induced type 1 diabetic mice demonstrated that enhancing p53 deacetylation modifications can protect cardiomyocytes from damage caused by high glucose levels through a mechanism involving the inhibition of ferroptosis [53]. Another animal experiment demonstrated reduced Kac of heart histone H3 at lysine 9 and lysine 23 in db/db mice with renal failure, which contributes to cardiomyocyte hypertrophy in mice with type 2 diabetes [54]. Further findings revealed a significant role of specific Kac-modifying regulatory enzymes in DbCM. Lanfang et al. demonstrated through related experiments that jugular vein injection of an adenovirus overexpressing SIRT3 could markedly reduce the Kac level of p53 and the expression of TIGAR in the heart tissue of diabetic db/db mice, thereby effectively improving the pathological condition of DbCM [55]. Other researchers have reported that HDAC4 knockout induces heart failure in mouse models of type 1 and type 2 diabetes [56].

In summary, Kac modification and its regulatory enzymes play an indispensable role in the metabolic perturbation of cardiomyocytes in DbCM, and influence the occurrence and development of DbCM through a complex regulatory network. Future studies will further reveal the specific details of these regulatory mechanisms, and provide new ideas and targets for the prevention and treatment of DbCM.

Novel PTMs in DbCM

Lactylation

Kla was discovered in 2019, and lactate produced by glycolysis served as an acyl donor [57]. Lactate is an alternative energy source that sustains the energy supply of the heart in the DbCM state. During glycolysis, glucose is metabolized to pyruvate through a series of enzymatic reactions, resulting in the production of NADH and lactate [58]. Studies have demonstrated that the concentration of lactate in the peripheral blood of patients with DbCM is significantly elevated, which is closely associated with cardiac diastolic dysfunction [59, 60]. Under the influence of acyl-CoA synthetases such as ACSS2, lactate in the myocardium utilizes acetyl-CoA to form lactoyl-CoA. Subsequently, lactoyl-CoA undergoes Kla modification through the action of specific acyl-writing enzymes, thereby participating in the negative regulation of myocardial function.

Enzymes such as P300/CBP, GCN5, and HBO1 catalyze the covalent attachment of lactoyl-CoA to specific lysine residues on proteins, leading to Kla modifications [61,62,63]. In contrast, erasing enzymes, including HDAC1-3 and SIRT1-3, are responsible for removing Kla from proteins [64, 65], thereby restoring them to their original state and facilitating the precise regulation of the modification level. These writing and erasing enzymes not only contribute to the formation of Kla, but also exhibit the ability to catalyze other acylation modifications, underscoring their versatility in protein PTMs.

Kla plays a critical role as a bridge between glucose metabolism and epigenetic regulation. Recent studies have revealed the biological significance of Kla in DbCM. In DbCM mice, the abnormal upregulation of MCT4 induces excessive lactate excretion in cardiomyocytes, leading to lactoacylation of histone H4K12 in macrophages, which results in a series of inflammatory damages to cardiomyocytes. Thus, Kla may be associated with a detrimental microenvironment in cardiomyocytes [40]. In addition, the results of in vitro experiments indicated that exogenous lactate can promote pathological hypertrophy of the heart by enhancing the expression levels of histone H3K18 lactylation modification in cardiomyocytes. The use of glycolysis and LDH inhibitors to reduce histone lactoacylation modification levels in cardiomyocytes effectively inhibits the progression of cardiac hypertrophy. This finding may offer a novel targeted intervention strategy for reversing ventricular remodeling in DbCM [66]. Patients with DbCM experience a gradual deterioration of heart function, which may ultimately result in heart failure. A new study employed protein-modifying omics to characterize Kla in the hearts of mice suffering from heart failure. Under normal physiological conditions, the tail of the α-myosin heavy chain (α-MHC) binds to myosin, forming a thick muscle filament [67]. Stability of this thick muscle filament is crucial for maintaining the normal structure and contractile function of the heart. These findings indicate that in the context of heart failure, the level of Kla at the K1897 site of α-MHC was significantly reduced. This alteration disrupts the stability of thick muscle filaments and exacerbates the symptoms of heart failure [68]. The decreased level of α-MHC K1897 Kla was primarily attributed to the excessive outflow and depletion of lactate in cardiomyocytes during heart failure. However, the relationship between this phenomenon and the expression and localization of the corresponding lactate transporter receptors in the myocardial membrane requires further investigation. Consequently, it is essential to observe the dynamic changes in lactate levels and lactoacylation modification in cardiomyocytes throughout the progression of diabetic cardiomyopathy to heart failure, as this will enhance our understanding of the regulatory role of lactoacylation modification in the advancement of DbCM.

β-Hydroxybutyrylation

β-HB synthesis primarily occurs in the liver, although the kidneys and intestines also produce small amounts of β-HB [69]. However, it remains unclear whether the heart can synthesize ketone bodies. In the pathological state of diabetes, fatty acids are converted to acetyl-CoA through β-oxidation, which is further converted to β-HB by the liver ketone body synthetase system [70]. Subsequently, β-HB is transported to the cardiomyocytes via the bloodstream. Within cardiomyocytes, β-HB can be oxidized and phosphorylated through the TCA cycle, releasing energy to meet myocardial energy demands. Notably, β-HB not only serves as an additional energy source for cardiac muscle, but also functions as a potent antioxidant [71], mitigating damage to heart muscle cells caused by oxidative stress. In addition, β-HB has been implicated in PTMs of the DbCM heart, especially Kbhb, with the CoA-active form of β-HB as a precursor, which has recently attracted attention.

The regulatory mechanism of Kbhb is complex and intricate, and involves the synergistic action of multiple enzymes. Acyltransferase P300/CBP functions as a writing enzyme, catalyzing the covalent binding of β-hydroxybutyryl-CoA to lysine residues to form Kbhb [72]. This process is crucial for the regulation of various biological activities, including gene expression, cellular metabolism, and signal transduction. Concurrently, histone deacetylases HDAC1 and HDAC2 serve as erasing enzymes responsible for removing Kbhb modifications, thereby maintaining homeostasis of intracellular modification levels [73]. The identification of these regulatory enzymes associated with Kbhb modification offers new insights for a deeper understanding of its biological functions.

Research on Kbhb in DbCM is still in its early stages of development. A recent proteomic study employing liquid chromatography-tandem mass spectrometry (LC–MS/MS) successfully identified differences in the expression of Kbhb modifications in the heart tissue of DbCM rats induced by a high-glycemic, high-fat diet combined with STZ. Quantitative analysis revealed that 284 Kbhb modification sites were upregulated, whereas 52 were downregulated among 47 proteins in the hearts of DbCM rats. Bioinformatic analysis further suggested that Kbhb-modified proteins are involved in various energy metabolic pathways, including the TCA cycle, oxidative phosphorylation, and propionic acid metabolism. Thus, Kbhb may play a role in regulating the progression of DbCM by influencing myocardial energy metabolic status. Additionally, another proteomic analysis of Kbhb in the liver of fasting mice showed that Kbhb-modified proteins exhibited significant enrichment in fatty acid β-oxidation, TCA cycle, and ATP metabolism [74]. Accordingly, Kbhb may play a role in the regulation of ATP synthesis, thereby participating in the disturbance of energy metabolism in DCM cardiomyocytes. However, further studies are needed to explore the underlying mechanisms by which Kbhb regulates myocardial energy metabolism. Endothelial dysfunction is often regarded as an early pathological feature of diabetes-related vascular complications [75]. Diabetic hyperglycemia may impair cardiovascular endothelial function in patients with DbCM by activating oxidative stress, inflammatory responses, and formation of advanced glycation end products (AGEs). Wu et al. demonstrated that exogenous β-HB supplementation in STZ-induced SD rats with type 2 diabetes upregulated the modification level of histone H3K9bhb in the rat heart in a dose-dependent manner, resulting in the expression of vascular endothelial growth factor (VEGF) and subsequently improving aortic endothelial injury in DbCM hearts [76]. Interestingly, this study found that the repair effect of β-HB on the vascular endothelium was most obvious at moderate concentrations, suggesting that Kbhb modification induced by different concentrations of β-HB may have some unique effects or mechanisms on the improvement of vascular endothelial injury in DbCM.

In summary, the potential role of Kbhb in diabetic cardiomyopathy has not yet been fully explored. Future studies will further reveal the epigenetic regulatory mechanism of Kbhb and provide a scientific basis for the prevention and treatment of diabetic cardiomyopathy.

Succinylation

Succinyl-CoA plays a crucial role in the intricate network of myocardial energy metabolism, as a key intermediate in the TCA cycle [77]. It is formed when α-ketoglutarate undergoes decarboxylation facilitated by the α-ketoglutarate dehydrogenase complex, resulting in the binding of an acetyl group to CoA. Within the metabolic framework of the TCA cycle, succinyl-CoA is catalyzed by succinyl-thiokinase, which hydrolyzes its thioester bonds, thereby releasing free energy for the synthesis of GTP, it is the major pathway for succinyl-CoA consumption. With or without enzymatic action, succinyl-CoA can transfer succinyl groups to lysine residues in cellular proteins, thereby achieving Ksucc. Metabolomics studies have shown that succinyl-CoA is the most abundant short-chain acyl-CoA in the mouse heart and that Ksucc is particularly pronounced in this energy-consuming organ, showing its potential importance in the regulation of cardiac function [78].

The regulatory mechanism of Ksucc is a complex process that involves various types of enzymes. Among these, writing enzymes such as P300/CBP, GCN5, and KAT2 are primarily responsible for catalyzing Ksucc, a process that induces significant changes in protein function and structure [79, 80]. Concurrently, erasing enzymes, including SIRT1, SIRT2, and SIRT5, remove the succinyl group, thereby restoring the protein to its original state and regulating the balance between different biological processes within the cell [81]. However, current research on the direct reading of enzymes involved in the regulatory mechanism of Ksucc is relatively scarce, and the existing literature and experimental data are limited, which hinders a comprehensive understanding of this field.

In the diabetic state, abnormal metabolism of energy substrates within cardiomyocytes, such as glucose, can lead to the accumulation of TCA cycle intermediates, particularly succinyl-CoA. As a result, high levels of succinyl-CoA promote Ksucc modifications of proteins, which may influence the activity and localization of certain key metabolic enzymes and signaling molecules in cardiomyocytes. Recently, with the extensive study of Ksucc, its role in the occurrence and progression of DbCM has been acknowledged. For instance, a study that integrated broadly targeted metabolomics, 4D proteomics, and Ksucc omics revealed that in a mouse model of DbCM heart failure, SIRT5 expression was significantly reduced, resulting in a substantial increase of the Ksucc of carnitine palmitoyl transferase 2 (CPT2) [82]. This change leads to the accumulation of myocardial lipids and increased lipid toxicity. Consequently, SIRT5-mediated Ksucc may represent a potential target for the treatment of DbCM, it is of great clinical significance to investigate the therapeutic effects of SIRT5 activators, such as investin and resveratrol, on DbCM and other metabolic diseases associated with fat deposition, as well as to determine whether Ksucc serves as the underlying mechanism [83]. However, it is crucial to note that this study was based on the db/db mouse model, which exhibits severe metabolic disorders and genetic variants. Thus, further validation using additional animal models of DbCM is warranted.

In summary, Ksucc is a significant PTMs for cardiomyocytes; however, its specific mechanisms in the occurrence and development of DbCM have not been fully elucidated. Therefore, future studies are needed to further explore the enzymatic mechanism of Ksucc, substrate specificity, and its interaction with other modification modes.

Malonylation

Using specific malonylated antibodies and LC–MS/MS, researchers first identified malonylation (Kma) in proteins from E. coli and HeLa cells in 2011 [84]. Malonylated acyl donor malonyl-CoA is an indispensable intermediate metabolite in fatty acid synthesis and β-oxidation. In cardiomyocytes, malonyl-CoA is produced by acetyl-CoA carboxylase (ACC) [85, 86], which is regulated by various hormones and nutritional signals. For example, AMPK is dephosphorylated when blood glucose levels rise, thereby inhibiting its phosphorylation of ACC, which leads to ACC activation and promotes malonyl-CoA synthesis [87]. The resulting malonyl-CoA is then transported to different regions within the cell and is involved in various metabolic pathways. In the process of fatty acid β-oxidation, malonyl-CoA, as a negative regulator, can reduce the rate of fatty acids entering mitochondria for β-oxidation by inhibiting the activity of carnitine palmitoyl transferase-1 (CPT-1), thereby regulating the energy supply of cardiomyocytes [88].

Recently, numerous studies have demonstrated that Kma is closely associated with the occurrence and progression of DbCM. Researchers have observed significantly elevated serum malonyl-CoA levels in DbCM patients, which may be associated with myocardial insulin resistance, hyperglycemia, and abnormal fatty acid metabolism. SIRT5 is one of the few known Kma-demodifying enzymes [89]. In 2023, Can Wei et al. found that knocking out SIRT5 resulted in a significant increase of GSTP1 protein Kma in hyperglycemia-stimulated mouse primary cardiomyocytes. This increase promotes mitochondrial dysfunction, pyrodeath, and DNA damage in the cardiomyocytes. Conversely, the overexpression of SIRT5 can mitigate myocardial damage associated with DbCM [90]. However, the study had several limitations. For instance, only western blotting was employed for detection, and the changes in Kma levels of all proteins in cardiomyocytes were not comprehensively evaluated using MS or other methods. Additionally, the wild-type mice used in the experiment originated from a different source than the SIRT5 knockout mice, which somewhat undermines the persuasive strength of the conclusions of the study. Other studies have demonstrated that quercetin, a component found in various medicinal plants, promotes desuccinylation of IDH2 through SIRT5. This process helps maintain mitochondrial homeostasis in mouse cardiomyocytes, improves myocardial inflammation and fibrosis, and ultimately contributes to a reduction in the incidence of heart failure caused by DbCM and other related diseases [91]. Malonylated proteomic analysis of a mouse model of myocardial hypertrophy indicated that malonylated protein enrichment was significantly associated with cardiac structure, systolic function, cGMP-PKG signaling pathway, and various metabolic processes [92]. Given that the heart also exhibits hypertrophy during the progression of heart failure in the context of DbCM, it may represent a promising therapeutic target for enhancing cardiac remodeling in DbCM.

In summary, research on Kma not only elucidates the pathogenesis of DbCM, but also has the potential to offer new therapeutic avenues for other causes of heart failure, thereby expanding treatment strategies in this field.

Palmitoylation

The palmitoylation of proteins is a PTM in which palmitic acid or palmitoyl-CoA serves as an acyl group donor [93]. Palmitic acid, a 16-carbon saturated fatty acid, is a key product of lipid metabolism in cardiomyocytes and can be obtained through the diet or synthesized endogenously. During β-oxidation, palmitic acid is initially converted to palmitoyl-CoA within the mitochondria of cardiomyocytes [94]. Subsequently, palmitoyl-CoA undergoes a series of enzymatic reactions that gradually degrade into short-chain fatty acyl-CoA fragments. These fragments then enter the TCA cycle, ultimately producing acetyl-CoA, NADH, FADH2, and a small amount of ATP, which collectively provides energy for cardiac muscle cells.

The enzymes responsible for palmitoylation, particularly the DHHC family of proteins, help add palmitoyl groups to cysteine residues in proteins [95, 96]. Instead, erasing enzymes (such as APT1) removes palmitoyl groups from these protein molecules [97]. The expression and activities of these enzymes in cardiomyocytes are strictly regulated to maintain palmitoylation homeostasis.

Recently, an increasing number of studies have indicated that palmitoylation modifications may play a role in the pathological processes of DbCM by influencing lipid metabolism, insulin secretion, and related signal transduction pathways in cardiomyocytes. CD36 is a transmembrane protein responsible for approximately two-thirds of the fatty acid intake in the heart. In certain instances, functional migration of CD36 across the plasma membrane can enhance fatty acid absorption. A recent study demonstrated that the specific knockout of TGR5 in the myocardial cells of mice with DbCM led to increased activity of DHHC4, the enzyme responsible for catalyzing CD36 palmitoylation. This results in enhanced localization of CD36 to the cell membrane, thereby increasing the uptake of long-chain fatty acids by cardiomyocytes, which ultimately contributes to hypertrophy, fibrosis, and dysfunction of the heart [98]. Therefore, intervention in palmitoylation modification within cardiomyocytes may play a crucial role in preventing cardiac lipid toxicity and DbCM development. In addition to DHHC4, both DHHC5 and DHHC6 have also been implicated in the palmitoylation modification of CD36 [99], and future studies are needed to explore their epigenetic regulatory role in the development of DbCM. Research conducted by Professor Guofang Dong et al. has demonstrated that APT1 plays a significant role in the pathogenesis of diabetes by regulating the palmitoylation status of Scamp1, which in turn affects the function of islet β cells and insulin secretion [97]. A similar mechanism might be present in cardiomyocytes. The abnormal function of APT1 could lead to impaired cardiomyocyte function and cardiac remodeling by influencing palmitoylation of proteins associated with the energy metabolism of cardiomyocytes. Future studies will provide deeper insights into the specific mechanisms of action of palmitoylation and its regulatory enzymes in DbCM, which will encourage us to explore ways to optimize the treatment strategies for DbCM.

Conclusions and prospects

With the rapid development of modern techniques, such as protein modification omics, researchers have confirmed that various novel protein acylation reactions are associated with disturbances in myocardial energy metabolism in DbCM [Fig. 2]. In addition to the novel PTMs highlighted here, current research suggests that O-linked β-N-acetylglucosamine-modification (O-GlcNAcylation) may also be involved in mitochondrial dysfunction in DbCM [100]. However, our understanding of the relationship between novel protein acylation modifications and DbCM remains limited, and many potential molecular mechanisms require further exploration and study. For example, it is unclear whether these novel PTMs are more likely to significantly affect changes in cardiac morphology and function through non-histone or histone acylation. Based on the available evidence, histone PTMs appear to play a more significant and direct role in the epigenetic regulation of DbCM. By modifying chromatin structure, histone PTMs influence DNA accessibility, thereby regulating gene expression and transcriptional activity. Additionally, the combinations of histone PTMs are notably complex and diverse. In contrast, the functions of non-histone PTMs are more prominently associated with cell signaling and metabolic regulation, with their direct impact on gene expression being relatively limited. Consequently, there is an urgent need for comprehensive and in-depth pathophysiological studies to elucidate the specific details of these mechanisms.

Fig. 2
figure 2

Metabolic perturbations in cardiomyocytes in DbCM and acyl donor formation of novel PTMs. Insulin resistance leads to reduced extracellular glucose uptake and glycolysis in diabetic cardiomyocytes. Additionally, aerobic oxidation of pyruvate within the mitochondria is inhibited, resulting in increased lactate levels in cardiomyocytes. Free fatty acids and ketone β-HB serve as alternative energy sources to glucose, enhancing their supply to cardiomyocytes and promoting the uptake of fatty acids and β-oxidation. However, the metabolism of ketone bodies in cardiomyocytes is bidirectional, with acyl-CoA being both formed and consumed during energy metabolism, thereby influencing the corresponding PTMs. DbCM, diabetic cardiomyopathy; β-HB, β-hydroxybutyrate; GLUT, glucose transporter; TCA cycle, tricarboxylic acid cycle; BDH1, β-hydroxybutyrate dehydrogenase 1; SCOT, succinyl-CoA:3-ketoacid CoA transferase; SCS, succinyl-CoA synthetase; AcAc, acetoacetic acid; FACS, fatty acyl-CoA synthetase; NADH, nicotinamide adenine dinucleotide; FADH2, flavin adenine dinucleotide hydrogen transmitter 2

Many protein acylation modifications are mediated by the same acyl-CoA synthetase, acyltransferase, and deacylase, which enhances the efficiency of identifying undiscovered regulatory enzymes and accelerates investigation of the biological functions of protein acylation modifications in DbCM [Table 1]. The researchers could further investigate whether the classic acyltransferase P300/CBP, along with the acyl-removing enzymes from the SIRT family, can catalyze the addition and removal of newly discovered acylated modifications. Additionally, they should continue to explore the roles of these regulatory enzymes in the modulation of these acylated modifications. Consequently, when exploring these reversible PTMs, it is crucial to thoroughly understand and closely monitor the dynamics of acyl donor levels under energy metabolic perturbations in DbCM as well as the dynamics of other modifications to gain a better understanding of the intricate regulatory network of protein acylation modifications in this pathological state.

Table 1 Summary of novel PTMs associated with myocardial metabolic perturbations in DbCM

Multiple PTMs can occur on the same protein, and these modifications can interact with one another, leading to either synergistic or antagonistic effects. Collectively, these interactions form the epigenetic background of DbCM. The interplay between PTMs may occur through mechanisms such as competition for the same modification site, structural alterations that render another site inaccessible to a different PTM modifier, or direct regulation of the activity of a secondary PTM modifier. Furthermore, whether amino acid residues within the same protein can undergo multiple modifications simultaneously in cardiomyocytes remains an open question. Consequently, it is essential to systematically investigate the functional effects of combinations of two or more novel PTMs within the same experiment as well as the potential interaction mechanisms between them.

Although PTMs have potential applications in the treatment of DbCM, several challenges and limitations remain. The high specificity and complexity of PTMs, along with the interactions and effects among different modifications, complicate precise control, leading to uncertainties regarding treatment efficacy and the potential for side effects. Furthermore, PTMs typically occur within cells, posing difficulties in directly targeting specific modifications through external administration. Even if a drug can be successfully delivered into the cell, ensuring that it specifically acts on the target PTMs presents a significant challenge. Additionally, it is likely that a drug targeting specific PTMs may also affect other modifications, resulting in off-target effects. This could reduce the treatment effectiveness for DbCM or increase the incidence of side effects.

In summary, novel PTMs associated with metabolic disorders are key to DbCM progression. Looking forward to the future, with the improvement of the precision of energy metabolity-related detection instruments, the significant improvement in the detection accuracy and application range of protein modification omics technology, and the development and marketing of more novel acylation modification site-specific antibodies, our understanding of energy metabolism disturbances in DbCM hearts will be more in-depth. Future studies should aim to establish a comprehensive network of regulatory relationships between PTMs and DbCM. Additionally, investigating PTMs in DbCM at the cellular level through techniques such as transcriptomics and modification omics will enhance our understanding of the functions and mechanisms underlying these associations. This knowledge will, in turn, facilitate the advancement and refinement of effective strategies for the prevention and treatment of DbCM.

Availability of data and materials

No datasets were generated or analyzed during the current study.

Abbreviations

DbCM:

Diabetic cardiomyopathy

PTMs:

Post-translational modifications

Kac:

Acetylation

Kla:

Lactylation

Kbhb:

β-Hydroxybutyrylation

Kma:

Malonylation

Ksucc:

Succinylation

β-HB:

β-Hydroxybutyrate

MS:

Mass spectrometry

TCA:

Tricarboxylic acid

ATP:

Adenosine triphosphate

STZ:

Streptozotocin

References

  1. Tong M, Saito T, Zhai P, Oka SI, Mizushima W, Nakamura M, Ikeda S, Shirakabe A, Sadoshima J. Mitophagy is essential for maintaining cardiac function during high fat diet-induced diabetic cardiomyopathy. Circ Res. 2019;124(9):1360–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/CIRCRESAHA.118.314607.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Li H, Zhu X, Cao X, Lu Y, Zhou J, Zhang X. Single-cell analysis reveals lysyl oxidase (Lox)+ fibroblast subset involved in cardiac fibrosis of diabetic mice. J Adv Res. 2023;54:223–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jare.2023.01.018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Pappritz K, Grune J, Klein O, Hegemann N, Dong F. Speckle-tracking echocardiography combined with imaging mass spectrometry assesses region-dependent alterations. Sci Rep. 2020;10(1):3629. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-020-60594-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Haas AV, McDonnell ME. Pathogenesis of cardiovascular disease in diabetes. Endocrinol Metab Clin N Am. 2018;47(1):51–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ecl.2017.10.010.

    Article  Google Scholar 

  5. Jensen MT, Fung K, Aung N, Sanghvi MM. Changes in cardiac morphology and function in individuals with diabetes mellitus: the UK biobank cardiovascular magnetic resonance substudy. Circ Cardiovasc Imaging. 2019;12(9): e009476. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/CIRCIMAGING.119.009476.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Chen R, Zhang H, Tang B, Luo Y, Yang Y, Zhong X, Chen S, Xu X. Macrophages in cardiovascular diseases: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther. 2024;9(1):130. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-024-01840-1.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Shu S, Cui H, Liu Z, Zhang H, Yang Y, Chen X. Suppression of RCAN1 alleviated lipid accumulation and mitochondrial fission in diabetic cardiomyopathy. Metabolism. 2024;158: 155977. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.metabol.2024.155977.

    Article  CAS  PubMed  Google Scholar 

  8. Ritchie RH, Abel ED. Basic mechanisms of diabetic heart disease. Circ Res. 2020;126(11):1501–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/CIRCRESAHA.120.315913.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zhou Y, Suo W, Zhang X, Yang Y, Zhao W, Li H, Ni Q. Targeting epigenetics in diabetic cardiomyopathy: therapeutic potential of flavonoids. Biomed Pharmacother. 2023;157: 114025. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2022.114025.

    Article  CAS  PubMed  Google Scholar 

  10. Ling C, Rönn T. Epigenetics in human obesity and type 2 diabetes. Cell Metab. 2019;29(5):1028–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2019.03.009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lee JM, Hammarén HM, Savitski MM, Baek SH. Control of protein stability by post-translational modifications. Nat Commun. 2023;14(1):201. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-023-35795-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Jazieh C, Arabi TZ, Asim Z, Sabbah BN, Alsaud AW, Alkattan K, Yaqinuddin A. Unraveling the epigenetic fabric of type 2 diabetes mellitus: pathogenic mechanisms and therapeutic implications. Front Endocrinol (Lausanne). 2024;15:1295967. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fendo.2024.1295967.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Li Z, Chen J, Huang H, Zhan Q, Wang F, Chen Z, Lu X, Sun G. Post-translational modifications in diabetic cardiomyopathy. J Cell Mol Med. 2024;28(7): e18158. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jcmm.18158.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Li X, Yang Y, Zhang B, Lin X, Fu X. Lactate metabolism in human health and disease. Signal Transduct Target Ther. 2022;7(1):305. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-022-01151-3. Erratum in: Signal Transduct Target Ther. 2022;7(1):372. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-022-01206-5.

  15. Zhou T, Cheng X, He Y, Xie Y, Xu F, Xu Y, Huang W. Function and mechanism of histone β-hydroxybutyrylation in health and disease. Front Immunol. 2022;13: 981285. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.981285.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gut P, Matilainen S, Meyer JG, Pällijeff P, Richard J, Carroll CJ, Euro L. SUCLA2 mutations cause global protein succinylation contributing to the pathomechanism of a hereditary mitochondrial disease. Nat Commun. 2020;11(1):5927. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-020-19743-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Cheng X, Wang K, Zhao Y, Wang K. Research progress on post-translational modification of proteins and cardiovascular diseases. Cell Death Discov. 2023;9(1):275. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41420-023-01560-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bornstein MR, Tian R, Arany Z. Human cardiac metabolism. Cell Metab. 2024;36(7):1456–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2024.06.003.

    Article  CAS  PubMed  Google Scholar 

  19. Da Dalt L, Cabodevilla AG, Goldberg IJ, Norata GD. Cardiac lipid metabolism, mitochondrial function, and heart failure. Cardiovasc Res. 2023;119(10):1905–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/cvr/cvad100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kazmi DH, Kapoor A, Sinha A. Role of metabolic manipulator trimetazidine in limiting percutaneous coronary intervention-induced myocardial injury. Indian Heart J. 2018;70(Suppl 3):S365–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ihj.2018.10.415.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Heather LC, Gopal K, Srnic N, Ussher JR. Redefining diabetic cardiomyopathy: perturbations in substrate metabolism at the heart of its pathology. Diabetes. 2024;73(5):659–70. https://doiorg.publicaciones.saludcastillayleon.es/10.2337/dbi23-0019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kota SK, Kota SK, Jammula S, Panda S, Modi KD. Effect of diabetes on alteration of metabolism in cardiac myocytes: therapeutic implications. Diabetes Technol Ther. 2011;13(11):1155–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/dia.2011.0120.

    Article  CAS  PubMed  Google Scholar 

  23. Rider OJ, Apps A, Miller JJJJ, Lau JYC, Lewis AJM. Noninvasive in vivo assessment of cardiac metabolism in the healthy and diabetic human heart using hyperpolarized 13C MRI. Circ Res. 2020;126(6):725–36. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/CIRCRESAHA.119.316260.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ju J, Xiao D, Shen N, Zhou T, Che H, Li X, Zhang S, Mokembo JN, Jha NK, Monayo SM, Wang Z, Zhang Y. miR-150 regulates glucose utilization through targeting GLUT4 in insulin-resistant cardiomyocytes. Acta Biochim Biophys Sin (Shanghai). 2020;52(10):1111–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/abbs/gmaa094.

    Article  CAS  PubMed  Google Scholar 

  25. Mansor LS, Sousa Fialho MDL, Yea G, Coumans WA. Inhibition of sarcolemmal FAT/CD36 by sulfo-N-succinimidyl oleate rapidly corrects metabolism and restores function in the diabetic heart following hypoxia/reoxygenation. Cardiovasc Res. 2017;113(7):737–48. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/cvr/cvx045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Montaigne D, Butruille L, Staels B. PPAR control of metabolism and cardiovascular functions. Nat Rev Cardiol. 2021;18(12):809–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41569-021-00569-6.

    Article  CAS  PubMed  Google Scholar 

  27. Finck BN, Lehman JJ, Leone TC, Welch MJ, Bennett MJ. The cardiac phenotype induced by PPARalpha overexpression mimics that caused by diabetes mellitus. J Clin Investig. 2002;109(1):121–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI14080.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Glatz JFC, Heather LC, Luiken JJFP. CD36 as a gatekeeper of myocardial lipid metabolism and therapeutic target for metabolic disease. Physiol Rev. 2024;104(2):727–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/physrev.00011.2023.

    Article  CAS  PubMed  Google Scholar 

  29. Park J, Song H, Moon S, Kim Y, Cho S. Cardiometabolic benefits of fenofibrate in heart failure related to obesity and diabetes. Cardiovasc Diabetol. 2024;23(1):343. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12933-024-02417-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cerf ME. Cardiac glucolipotoxicity and cardiovascular outcomes. Medicina (Kaunas). 2018;54(5):70. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/medicina54050070.

    Article  PubMed  Google Scholar 

  31. Puchalska P, Crawford PA. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 2017;25(2):262–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2016.12.022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Brahma MK, Ha CM, Pepin ME, Mia S. Increased glucose availability attenuates myocardial ketone body utilization. J Am Heart Assoc. 2020;9(15): e013039. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/JAHA.119.013039.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Abdurrachim D, Woo CC, Teo XQ, Chan WX. A new hyperpolarized 13C ketone body probe reveals an increase in acetoacetate utilization in the diabetic rat heart. Sci Rep. 2019;9(1):5532. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-019-39378-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Al Batran R, Gopal K, Capozzi ME, Chahade JJ, Saleme B. Pimozide alleviates hyperglycemia in diet-induced obesity by inhibiting skeletal muscle ketone oxidation. Cell Metab. 2020;31(5):909-919.e8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2020.03.017.

    Article  CAS  PubMed  Google Scholar 

  35. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21(3):263–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nm.3804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lopaschuk GD, Dyck JRB. Ketones and the cardiovascular system. Nat Cardiovasc Res. 2023;2(5):425–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s44161-023-00259-1.

    Article  CAS  PubMed  Google Scholar 

  37. Nagao M, Toh R, Irino Y, Mori T, Nakajima H, Hara T, Honjo T, Satomi-Kobayashi S, Shinke T, Tanaka H, Ishida T, Hirata K. β-Hydroxybutyrate elevation as a compensatory response against oxidative stress in cardiomyocytes. Biochem Biophys Res Commun. 2016;475(4):322–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbrc.2016.05.097.

    Article  CAS  PubMed  Google Scholar 

  38. Cieluch A, Uruska A, Zozulinska-Ziolkiewicz D. Can we prevent mitochondrial dysfunction and diabetic cardiomyopathy in type 1 diabetes mellitus? Pathophysiology and treatment options. Int J Mol Sci. 2020;21(8):2852. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms21082852.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dong S, Qian L, Cheng Z, Chen C, Wang K, Hu S, Zhang X, et al. Lactate and myocardiac energy metabolism. Front Physiol. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphys.2021.71508.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Ma XM, Geng K, Wang P, Jiang Z, Law BY, Xu Y. MCT4-dependent lactate transport: a novel mechanism for cardiac energy metabolism injury and inflammation in type 2 diabetes mellitus. Cardiovasc Diabetol. 2024;23(1):96. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12933-024-02178-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Singh RM, Waqar T, Howarth FC, Adeghate E, Bidasee K, Singh J. Hyperglycemia-induced cardiac contractile dysfunction in the diabetic heart. Heart Fail Rev. 2018;23(1):37–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10741-017-9663-y.

    Article  PubMed  Google Scholar 

  42. Sun L, Yu M, Zhou T, Zhang S, He G, Wang G, Gang X. Current advances in the study of diabetic cardiomyopathy: from clinicopathological features to molecular therapeutics (review). Mol Med Rep. 2019;20(3):2051–62. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/mmr.2019.10473.

    Article  CAS  PubMed  Google Scholar 

  43. Dabravolski SA, Sadykhov NK, Kartuesov AG. The role of mitochondrial abnormalities in diabetic cardiomyopathy. Int J Mol Sci. 2022;23(14):7863. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms23147863.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Verma S, Rawat S, Ho KL, Wagg CS, Zhang L. Empagliflozin increases cardiac energy production in diabetes: novel translational insights into the heart failure benefits of SGLT2 Inhibitors. JACC Basic Transl Sci. 2018;3(5):575–87. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jacbts.2018.07.006.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Oshima H, Miki T, Kuno A, Mizuno M. Empagliflozin, an SGLT2 inhibitor, reduced the mortality rate after acute myocardial infarction with modification of cardiac metabolomes and antioxidants in diabetic rats. J Pharmacol Exp Ther. 2019;368(3):524–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/jpet.118.253666.

    Article  CAS  PubMed  Google Scholar 

  46. Zhang E, Wu Y. Metabolic memory: mechanisms and implications for diabetic vasculopathies. Sci China Life Sci. 2014;57(8):845–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11427-014-4710-6.

    Article  CAS  PubMed  Google Scholar 

  47. Mews P, Donahue G, Drake AM, Luczak V, Abel T, Berger SL. Acetyl-CoA synthetase regulates histone acetylation and hippocampal memory. Nature. 2017;546(7658):381–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature22405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chakrabarty RP, Chandel NS. Mitochondria as signaling organelles control mammalian stem cell fate. Cell Stem Cell. 2021;28(3):394–408. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.stem.2021.02.011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. An C, Deng L, Zhai H, You Y, Wu F, Zhai Q, Goossens A, Li C. Regulation of jasmonate signaling by reversible acetylation of TOPLESS in Arabidopsis. Mol Plant. 2022;15(8):1329–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molp.2022.06.014.

    Article  CAS  PubMed  Google Scholar 

  50. Cheng YW, Zeng FM, Li DJ, Wang SH, He JZ, Guo ZC. P300/CBP-associated factor (PCAF)-mediated acetylation of Fascin at lysine 471 inhibits its actin-bundling activity and tumor metastasis in esophageal cancer. Cancer Commun (Lond). 2021;41(12):1398–416. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cac2.12221.

    Article  PubMed  Google Scholar 

  51. Shen Y, Wei W, Zhou DX. Histone acetylation enzymes coordinate metabolism and gene expression. Trends Plant Sci. 2015;20(10):614–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tplants.2015.07.005.

    Article  CAS  PubMed  Google Scholar 

  52. Wan L, Chong S, Xuan F, Liang A, Cui X, Gates L. Impaired cell fate through gain-of-function mutations in a chromatin reader. Nature. 2020;577(7788):121–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-019-1842-7.

    Article  CAS  PubMed  Google Scholar 

  53. Tang YJ, Zhang Z, Yan T, Chen K, Xu GF. Irisin attenuates type 1 diabetic cardiomyopathy by anti-ferroptosis via SIRT1-mediated deacetylation of p53. Cardiovasc Diabetol. 2024;23(1):116. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12933-024-02183-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gaikwad AB, Sayyed SG, Lichtnekert J, Tikoo K, Anders HJ. Renal failure increases cardiac histone h3 acetylation, dimethylation, and phosphorylation and the induction of cardiomyopathy-related genes in type 2 diabetes. Am J Pathol. 2010;176(3):1079–83. https://doiorg.publicaciones.saludcastillayleon.es/10.2353/ajpath.2010.090528.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Li L, Zeng H, He X, Chen JX. Sirtuin 3 alleviates diabetic cardiomyopathy by regulating TIGAR and cardiomyocyte metabolism. J Am Heart Assoc. 2021;10(5): e018913. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/JAHA.120.018913.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kronlage M, Dewenter M, Grosso J, Fleming T. O-GlcNAcylation of histone deacetylase 4 protects the diabetic heart from failure. Circulation. 2019;140(7):580–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/CIRCULATIONAHA.117.031942.

    Article  CAS  PubMed  Google Scholar 

  57. Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574(7779):575–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-019-1678-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Brooks GA. Lactate as a fulcrum of metabolism. Redox Biol. 2020;35: 101454. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.redox.2020.101454.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. d’Humières T, Bouvarel A, Boyer. Cardiac diastolic maladaptation is associated with the severity of exercise intolerance in sickle cell anemia patients. Sci Rep. 2024;14(1):11095. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-024-61689-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Brooks GA. Role of the heart in lactate shuttling. Front Nutr. 2021;8: 663560. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnut.2021.663560.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang L, Wang Y, Meng M, Ma N, Wei G. High-concentrate diet elevates histone lactylation mediated by p300/CBP through the upregulation of lactic acid and induces an inflammatory response in mammary gland of dairy cows. Microb Pathog. 2023;180: 106135. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.micpath.2023.106135.

    Article  CAS  PubMed  Google Scholar 

  62. Li Z, Gong T, Wu Q, Zhang Y, Zheng X, Li Y. Lysine lactylation regulates metabolic pathways and biofilm formation in Streptococcus mutans. Sci Signal. 2023;16(801):eadg1849. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/scisignal.adg1849.

    Article  CAS  PubMed  Google Scholar 

  63. Niu Z, Chen C, Wang S, Lu C, Wu Z, Wang A. HBO1 catalyzes lysine lactylation and mediates histone H3K9la to regulate gene transcription. Nat Commun. 2024;15(1):3561. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-024-47900-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Sun S, Xu Z, He L, Shen Y, Yan Y, Lv X, Zhu X. Metabolic regulation of cytoskeleton functions by HDAC6-catalyzed α-tubulin lactylation. Nat Commun. 2024;15(1):8377. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-024-52729-0.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Wang M, Lin H. Understanding the function of mammalian sirtuins and protein lysine acylation. Annu Rev Biochem. 2021;90:245–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-biochem-082520-125411.

    Article  CAS  PubMed  Google Scholar 

  66. Zhao SS, Liu J, Wu QC, Zhou XL. Lactate regulates pathological cardiac hypertrophy via histone lactylation modification. J Cell Mol Med. 2024;28(16): e70022. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jcmm.70022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Aksel T, Choe YuE, Sutton S, Ruppel KM, Spudich JA. Ensemble force changes that result from human cardiac myosin mutations and a small-molecule effector. Cell Rep. 2015;11(6):910–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2015.04.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Zhang N, Zhang Y, Xu J, Wang P, Wu B. α-Myosin heavy chain lactylation maintains sarcomeric structure and function and alleviates the development of heart failure. Cell Res. 2023;33(9):679–98. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41422-023-00844-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Li K, Wang WH, Wu JB, Xiao WH. β-hydroxybutyrate: a crucial therapeutic target for diverse liver diseases. Biomed Pharmacother. 2023;165: 115191. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2023.115191.

    Article  CAS  PubMed  Google Scholar 

  70. Han YM, Ramprasath T, Zou MH. β-Hydroxybutyrate and its metabolic effects on age-associated pathology. Exp Mol Med. 2020;52(4):548–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s12276-020-0415-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Evans M, McClure TS, Koutnik AP, Egan B. Exogenous ketone supplements in athletic contexts: past, present, and future. Sports Med. 2022;52(Suppl 1):25–67. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40279-022-01756-2.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Huang H, Zhang D, Weng Y, Delaney K, Tang Z. The regulatory enzymes and protein substrates for the lysine β-hydroxybutyrylation pathway. Sci Adv. 2021;7(9):eabe2771. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.abe2771.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Chriett S, Dąbek A, Wojtala M, Vidal H. Prominent action of butyrate over β-hydroxybutyrate as histone deacetylase inhibitor, transcriptional modulator and anti-inflammatory molecule. Sci Rep. 2019;9(1):742. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-018-36941-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Koronowski KB, Greco CM, Huang H, Kim JK. Ketogenesis impact on liver metabolism revealed by proteomics of lysine β-hydroxybutyrylation. Cell Rep. 2021;36(5): 109487. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2021.109487.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Karan A, Bhakkiyalakshmi E, Jayasuriya R, Sarada DVL, Ramkumar KM. The pivotal role of nuclear factor erythroid 2-related factor 2 in diabetes-induced endothelial dysfunction. Pharmacol Res. 2020;153: 104601. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.phrs.2019.104601.

    Article  CAS  PubMed  Google Scholar 

  76. Wu X, Miao D, Liu Z, Liu K, Zhang B. β-hydroxybutyrate antagonizes aortic endothelial injury by promoting generation of VEGF in diabetic rats. Tissue Cell. 2020;64: 101345. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tice.2020.101345.

    Article  CAS  PubMed  Google Scholar 

  77. Lancaster MS, Graham BH. Succinyl-CoA synthetase dysfunction as a mechanism of mitochondrial encephalomyopathy: more than just an oxidative energy deficit. Int J Mol Sci. 2023;24(13):10725. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms241310725.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Sadhukhan S, Liu X, Ryu D, Nelson OD. Metabolomics-assisted proteomics identifies succinylation and SIRT5 as important regulators of cardiac function. Proc Natl Acad Sci USA. 2016;113(16):4320–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.1519858113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Yuan Y, Yuan H, Yang G, Yun H, Zhao M. IFN-α confers epigenetic regulation of HBV cccDNA minichromosome by modulating GCN5-mediated succinylation of histone H3K79 to clear HBV cccDNA. Clin Epigenet. 2020;12(1):135. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13148-020-00928-z.

    Article  CAS  Google Scholar 

  80. Wang Y, Guo YR, Liu K, Yin Z, Liu R, Xia Y. KAT2A coupled with the α-KGDH complex acts as a histone H3 succinyltransferase. Nature. 2017;552(7684):273–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature25003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Fang Y, Tang S, Li X. Sirtuins in metabolic and epigenetic regulation of stem cells. Trends Endocrinol Metab. 2019;30(3):177–88. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tem.2018.12.002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Wu M, Tan J, Cao Z, Cai Y, Huang Z. Sirt5 improves cardiomyocytes fatty acid metabolism and ameliorates cardiac lipotoxicity in diabetic cardiomyopathy via CPT2 de-succinylation. Redox Biol. 2024;73: 103184. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.redox.2024.103184.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Mori M, Cazzaniga G, Meneghetti F, Villa S, Gelain A. Insights on the modulation of SIRT5 activity: a challenging balance. Molecules. 2022;27(14):4449. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/molecules27144449.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Peng C, Lu Z, Xie Z, Cheng Z, Chen Y. The first identification of lysine malonylation substrates and its regulatory enzyme. Mol Cell Proteomics. 2011;10(12):M111.012658. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/mcp.M111.012658.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Yu Y, Nie Q, Wang Z, Di Y, Chen X, Ren K. Targeting acetyl-CoA carboxylase 1 for cancer therapy. Front Pharmacol. 2023;14:1129010. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Yoshii Y, Furukawa T, Saga T, Fujibayashi Y. Acetate/acetyl-CoA metabolism associated with cancer fatty acid synthesis: overview and application. Cancer Lett. 2015;356(2 Pt A):211–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.canlet.2014.02.019.

    Article  CAS  PubMed  Google Scholar 

  87. Szkudelski T, Szkudelska K. The relevance of AMP-activated protein kinase in insulin-secreting β cells: a potential target for improving beta cell function? J Physiol Biochem. 2019;75(4):423–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13105-019-00706-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Folmes CD, Lopaschuk GD. Role of malonyl-CoA in heart disease and the hypothalamic control of obesity. Cardiovasc Res. 2007;73(2):278–87. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cardiores.2006.10.008.

    Article  CAS  PubMed  Google Scholar 

  89. Kwon OK, Bang IH, Choi SY, Jeon JM, Na AY, Gao Y, Cho SS, Ki SH, Choe Y, Lee JN, Ha YS, Bae EJ, Kwon TG, Park BH, Lee S. LDHA desuccinylase sirtuin 5 as a novel cancer metastatic stimulator in aggressive prostate cancer. Genomics Proteomics Bioinform. 2023;21(1):177–89. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.gpb.2022.02.004.

    Article  CAS  Google Scholar 

  90. Wei C, Shi M, Dong S, Li Z, Zhao B. SIRT5-related lysine demalonylation of GSTP1 contributes to cardiomyocyte pyroptosis suppression in diabetic cardiomyopathy. Int J Biol Sci. 2024;20(2):585–605. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/ijbs.83306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Chang X, Zhang T, Wang J, Liu Y. SIRT5-related desuccinylation modification contributes to quercetin-induced protection against heart failure and high-glucose-prompted cardiomyocytes injured through regulation of mitochondrial quality surveillance. Oxid Med Cell Longev. 2021;2021:5876841. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2021/5876841.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Wu LF, Wang DP, Shen J, Gao LJ. Global profiling of protein lysine malonylation in mouse cardiac hypertrophy. J Proteomics. 2022;266: 104667. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jprot.2022.104667.

    Article  CAS  PubMed  Google Scholar 

  93. Yang HQ, Martinez-Ortiz W, Hwang J, Fan X, Cardozo TJ, Coetzee WA. Palmitoylation of the KATP channel Kir6.2 subunit promotes channel opening by regulating PIP2 sensitivity. Proc Natl Acad Sci USA. 2020;117(19):10593–602. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.1918088117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Pei X, Li KY, Shen Y, Li JT. Palmitoylation of MDH2 by ZDHHC18 activates mitochondrial respiration and accelerates ovarian cancer growth. Sci China Life Sci. 2022;65(10):2017–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11427-021-2048-2.

    Article  CAS  PubMed  Google Scholar 

  95. Chang Y, Zhu J, Li X, Deng Y, Lai B. Palmitoylation regulates myelination by modulating the ZDHHC3-Cadm4 axis in the central nervous system. Signal Transduct Target Ther. 2024;9(1):254. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-024-01971-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Abazari D, Wild AR, Qiu T, Dickinson BC, Bamji SX. Activity-dependent post-translational regulation of palmitoylating and depalmitoylating enzymes in the hippocampus. J Cell Sci. 2023;136(7):jcs260629. https://doiorg.publicaciones.saludcastillayleon.es/10.1242/jcs.260629.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Dong G, Adak S, Spyropoulos G, Zhang Q. Palmitoylation couples insulin hypersecretion with β cell failure in diabetes. Cell Metab. 2023;35(2):332-344.e7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2022.12.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Wang H, Wang J, Cui H, Fan C, Xue Y. Inhibition of fatty acid uptake by TGR5 prevents diabetic cardiomyopathy. Nat Metab. 2024;6(6):1161–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s42255-024-01036-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Wang J, Hao JW, Wang X, Guo H. DHHC4 and DHHC5 facilitate fatty acid uptake by palmitoylating and targeting CD36 to the plasma membrane. Cell Rep. 2019;26(1):209-221.e5. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2018.12.022.

    Article  CAS  PubMed  Google Scholar 

  100. Qiu Z, Cui J, Huang Q, Qi B, Xia Z. Roles of O-GlcNAcylation in mitochondrial homeostasis and cardiovascular diseases. Antioxidants (Basel). 2024;13(5):571. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antiox13050571.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (No.82170834, No.U22A20286), Sichuan Science and Technology Program (No.2023ZYD0095), Sichuan Province cadre health research project (No. ZH2022-1501), Health Commission of Sichuan Province Medical Science and Technology Program (No.24CXTD02), Southwest Medical University Research Foundation for Clinical Medicine (No.2024LCYXZX12) and Luzhou-Southwest Medical University cooperation project (No.2021LZXNYD-P02, No.2021LZXNYD-G01).

Author information

Authors and Affiliations

Authors

Contributions

DL, LZ and QG wrote the main manuscript text, HD, CL, and TZ prepared Figs. 1 and 2 and Table 1, WH and YX revised the manuscript and provided funding support. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Wei Huang or Yong Xu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All authors approved the final manuscript for publication.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, D., Zhang, L., Gong, Q. et al. The role of myocardial energy metabolism perturbations in diabetic cardiomyopathy: from the perspective of novel protein post-translational modifications. Clin Epigenet 17, 15 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13148-025-01814-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13148-025-01814-2