Next Article in Journal
Application of Microorganisms for the Valorization of Side-Products of Rapeseed De-Oiling
Previous Article in Journal
Inflammatory Drug-Resistant Epilepsy Index (IDREI) as a Molecular Compound Biomarker in Focal Epilepsies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Insights into the Relationship Between Amino Acid Metabolism and Diabetic Cardiomyopathy

1
Department of Cardiology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
2
Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(7), 916; https://doi.org/10.3390/biom15070916 (registering DOI)
Submission received: 5 April 2025 / Revised: 30 May 2025 / Accepted: 19 June 2025 / Published: 22 June 2025
(This article belongs to the Section Molecular Medicine)

Abstract

:
Diabetes mellitus (DM) is a complex global pandemic that frequently leads to multiple complications. Diabetic cardiomyopathy (DCM) is the primary cause of heart failure in patients with type 1 and 2 diabetes and is fundamentally characterized by abnormalities in myocardial structure and function. Metabolic disorders occupy a leading role in the pathogenesis of DCM, manifesting as disrupted substrate metabolism, dysregulated signaling pathways, and energy imbalance. Given the limited benefits of conventional therapeutic strategies targeting glucolipid metabolism, increasing research efforts have focused on amino acid metabolism. Amino acids are involved in the synthesis of nitrogen-containing compounds and serve as an energy source under specific conditions. Moreover, emerging studies demonstrate that metabolic disturbances of specific amino acids—such as branched-chain amino acids (BCAAs), glutamine, and arginine—exacerbate mitochondrial dysfunction and oxidative stress, thereby promoting myocardial fibrosis and cardiomyocyte injury. Therefore, this review aims to summarize the general characteristics and regulatory pathways of amino acid metabolism, as well as the specific mechanisms by which metabolic alterations of amino acids contribute to the pathogenesis and progression of diabetic cardiomyopathy, with the hope of advancing more effective translational therapeutic approaches.

1. Introduction

Diabetes mellitus (DM), a prevalent metabolic disorder, has demonstrated a marked increase in global adult prevalence from 7% in 1990 to 14% in 2022, as evidenced by data from the Non-Communicable Diseases Risk Factor Collaboration (NCD-RisC) [1], highlighting that diabetes continues to be a significant global public health challenge. Cardiovascular disease (CVD) is the primary cause of morbidity and mortality in individuals with diabetes [2], with their risk being 2 to 4 times greater than that of individuals with normal blood glucose levels [3]. Diabetic cardiomyopathy (DCM), initially characterized by Rubler et al. in 1972, is defined as a condition of heart failure that occurs in the absence of coronary artery disease, hypertension, or valvular heart disease [4]. In June 2024, the European Society of Cardiology (ESC) expert consensus proposed a newly revised definition of DCM, characterized by myocardial systolic and/or diastolic dysfunction in the presence of diabetes, irrespective of other coexisting risk factors [5]. The updated definition significantly broadens the scope of diabetic cardiomyopathy, facilitating early detection and standardized interventions, thereby delaying heart failure progression and reducing morbidity and mortality.
Metabolic disorders, associated with disruptions in biochemical processes governing energy conversion and nutrient utilization, are fundamentally driven by the metabolic dysregulation of glucose, lipid and amino acid and frequently progress to chronic diseases, including diabetes mellitus and cardiovascular complications. Although glycemic control and heart failure medications exert beneficial effects on these conditions, currently no specific therapeutic approaches exist for targeting diabetic cardiomyopathy. Over the past decades, the pathophysiology of DCM has been preliminarily elucidated [6], with emerging evidence identifying the regulation of amino acid metabolism as a vital mediator and therapeutic target for invention in diabetes [7,8,9] and concurrent cardiovascular risk [10,11,12,13]. Therefore, given the current limitations in effective diagnostic and therapeutic methods [6] and these insightful clinical findings, we aim to systematically summarize the general principles and regulatory mechanisms of amino acid metabolism and review the critical role of amino acid metabolism in the development, diagnosis, treatment, and prognosis of diabetic cardiomyopathy.

2. Overview of Amino Acids and Their Metabolism

Approximately 20 amino acids participate in human protein synthesis, including essential amino acids (EAAs), which must be obtained from the diet, and non-essential amino acids (NEAAs), which are synthesized endogenously (Table 1). Beyond serving as protein building blocks, amino acids play critical roles in metabolic regulation, signaling, and immune function. Structurally, leucine, valine, and isoleucine are classified as branched-chain amino acids (BCAAs), while tryptophan, phenylalanine, and tyrosine are aromatic amino acids (AAAs). Amino acids are derived from both endogenous protein turnover and dietary intake, forming a metabolic pool that supports both anabolic processes (e.g., NEAA synthesis, protein formation) and catabolic pathways (e.g., transamination, deamination, and nitrogen metabolism).

3. Amino Acid Anabolism

Endogenous synthesis of non-essential amino acids (NEAAs) primarily relies on intermediates from glycolysis and the tricarboxylic acid (TCA) cycle, such as 3-phosphoglycerate and oxaloacetate. These substrates undergo transamination and other enzymatic reactions to yield NEAAs, a process essential for amino acid homeostasis and the conservation of essential amino acids (EAAs), particularly under stress conditions such as diabetic cardiomyopathy [14,15,16].
In contrast, dietary intake is the main source of EAAs for the human body. Intestinal absorption is mediated by various solute carrier (SLC) family transporters expressed on the apical membranes of epithelial cells. These amino acid transporters (AATs) include B0AT1 (SLC6A19) for neutral amino acids, b0,+AT (SLC7A9) for cationic amino acids, EAAT3 (SLC1A1) for anionic amino acids, PAT1 (SLC36A1) for glycine and proline, as well as β-amino acids, PepT1 (SLC15A1) for dipeptides and tripeptides, SIT (SLC6A20) for glycine and proline, and TauT (SLC6A6) for β-amino acids [17] Additionally, the large neutral amino acid transporter (LAT) family, which is also part of the SLC superfamily, is expressed on the basolateral membrane of intestinal epithelial cells and plays a crucial role in the coordination of amino acid excretion and reabsorption across the epithelium [18,19]. The antiporter LAT2 (SLC7A8) facilitates the transport of all neutral amino acids except proline [20], and y+LAT1 (SLC7A7) transports both cationic and neutral amino acids [21]. The uniporters TAT1 (SLC16A10) and LAT4 (SLC43A2) are responsible for the transport of AAAs and BCAAs, respectively [17] (LAT4 is also believed to participate in the transport of methionine and phenylalanine [22,23]).
In addition to dietary intake, EAA supplementation is vital in specific physiological or pathological states. For instance, lysine is used to support pediatric growth [24], and EAA mixtures combined with resistance training improve muscle function in sarcopenia [25]. Imbalanced EAA intake may contribute to conditions such as hepatic steatosis [26], neuronal dysfunction [27], or renal damage via excessive tryptophan metabolism [28]. Conversely, BCAA restriction is essential in disorders like maple syrup urine disease (MSUD) [29].

4. Amino Acid Catabolism

The degradation routes of amino acids exhibit both similarities and distinct characteristics. Amino acids absorbed through digestion, including alanine and aromatic amino acids, are metabolized predominantly in the liver. In contrast, the catabolism of BCAAs primarily occurs in skeletal muscle and affects their function simultaneously [30] Furthermore, several amino acid-degrading enzymes (AADEs), notably branched-chain aminotransferase (BCAT) and branched-chain keto acid dehydrogenase (BCKDH), are expressed in human intestinal epithelial cells [31], indicating that dietary amino acids, particularly BCAAs, can undergo degradation within the intestinal environment [32] (Table 1 and Figure 1).

4.1. Deamination of Amino Acids

Amino acid catabolism primarily occurs through deamination, with most amino acids undergoing trans-deamination catalyzed by aminotransferases and L-glutamate dehydrogenase, producing ammonia and α-keto acids—glutamate serving as a central intermediate. In the liver and kidney, some amino acids are further catabolized by L-amino acid oxidase, generating α-keto acids, H2O2, and ammonia. In skeletal and cardiac muscle, where glutamate dehydrogenase is less active, the purine nucleotide cycle (PNC) predominates. Certain amino acids such as histidine [33] and phenylalanine [34] also undergo non-oxidative deamination by gut microbes. The resulting α-keto acids can be funneled into energy production, NEAA synthesis, or converted into glucose and lipids via the TCA cycle. Notably, branched-chain α-keto acids (BCKAs) have been linked to tumor progression [35] and myocardial ischemia-reperfusion injury [36].

4.2. Metabolism of Blood Ammonia

The deamination of amino acids is the primary source of blood ammonia. In hepatocytes, glutamine (one of the carriers of ammonia) is transported into the mitochondrial matrix via the SLC1A5 transporter and hydrolyzed by glutaminase to produce glutamate and one molecule of ammonia. Thereafter, glutamate is further deaminated by L-glutamate dehydrogenase to yield α-ketoglutarate and another molecule of ammonia [37]. In the cytoplasm, alanine aminotransferase catalyzes the conversion of alanine and α-ketoglutarate to pyruvate and glutamate. Via carriers expressed on the inner mitochondrial membrane, including aspartate/glutamate carrier 2 (AGC2/SLC25A13) and glutamate carriers (GC1/SLC25A22 and GC2/SLC25A18) [38], glutamate is subsequently transported into the mitochondrial matrix. Ammonia can participate in the synthesis of NEAAs and other nitrogen-containing compounds, contributing to the maintenance of systemic ammonia homeostasis.

4.3. Metabolism of Individual Amino Acids

In addition to deamination and the metabolism of blood ammonia mentioned above, certain amino acids are involved in specialized metabolic pathways with significant physiological implications.

4.3.1. Aromatic Amino Acids

Under the catalysis of oxygenase, aromatic amino acids (AAAs) undergo decomposition to produce catecholamines, melanin, and multiple intermediates in glucose and lipid metabolism. Aromatic amino acid decarboxylase (AADC) mediates the production of aromatic amines by the gut microbiota and has been confirmed to stimulate colonic 5-hydroxytryptamine (5-HT) synthesis [39]. In addition to synthesizing proteins, tryptophan can be metabolized into kynurenine, serotonin (5-HT), indole derivatives, and nicotinic acid. Metabolites and enzymes involved in the kynurenine pathway (KP) have been implicated in lifespan extension [40], diabetic nephropathy [41], neuropsychiatric disorders [42], immune regulation [43], tolerance [44], and drug addiction [45].

4.3.2. Histidine

Histidine decarboxylation yields histamine; blockade of its receptors H1R/H2R exacerbates inflammatory immune responses via the NLRP3/caspase-1 proinflammatory cytokine pathway [46].

4.3.3. Serine

Serine hydroxymethyltransferase 2 (SHMT2) catalyzes the conversion of serine to glycine, generating a one-carbon unit that plays critical roles in development, immunity, and tumorigenesis. Additionally, one-carbon units support the synthesis of S-adenosylmethionine (SAM), the primary cellular methyl donor, which is closely associated with energy metabolism and epigenetic regulation [47,48].

4.3.4. Cysteine and Cystine

Cysteine and cystine are two other sulfur-containing amino acids that can interconvert with each other. Through a series of enzymatic reactions, cysteine generates endogenous hydrogen sulfide (H2S) and active sulfate (PAPS), which regulate vascular function, oxidative stress, inflammation, tumor proliferation, hormone homeostasis, and renal excretion [49].

4.3.5. Arginine

Arginine, under the catalysis of nitric oxide synthase (NOS), produces nitric oxide (NO), the first gasotransmitter discovered with complex cellular signaling functions. Over four decades of multidisciplinary research have established the pivotal role of NO in cardiovascular function, metabolism, neurotransmission, and immune modulation [50].

4.3.6. Branched-Chain Amino Acids

Following transportation into the mitochondria via the SLC25A44 carrier, BCAAs undergo transamination by branched-chain aminotransferase 2 (BCAT2) and oxidative decarboxylation by BCKDH, which generates BCKAs that participate in the TCA cycle and produce significant intermediates such as 3-hydroxyisobutyrate (3-HIB) and monomethyl branched-chain fatty acids (mmBCFAs) [51].

5. Regulation of Amino Acid Metabolism

Amino acid homeostasis refers to the dynamic equilibrium of amino acids and related metabolites within the body, which is the foundation for the proper functioning of protein synthesis, energy metabolism, and other vital biological processes. In addition to the regulation by enzymes (Figure 2), diet, signaling pathways, and amino acid transporters discussed below, amino acid metabolism is influenced to varying degrees by circadian rhythm [52], emotional health [53], and even climate change [54].

5.1. Amino Acid Metabolic Enzymes

As cellular biomolecules with catalytic capabilities, enzymes, particularly key enzymes, play a pivotal role in amino acid metabolism. For example, ATP-binding cassette (ABC) transporter proteins are among the rate-limiting factors in the kynurenine pathway (KP) of tryptophan, modulating the availability of tryptophan as a substrate for another rate-limiting enzyme, tryptophan-2,3-dioxygenase (TDO) [55]. Plant-derived bioactive compounds, such as quercetin [56], naringenin and naringin [57], along with organic acids found in hawthorn [58], can influence the activity of digestive enzymes that act on proteins in the gastrointestinal tract. Various vitamins, especially B vitamins, serve as coenzymes or cofactors and regulate the functional activity of enzymes involved in amino acid metabolism. According to the review of Nieraad et al., deficiencies in one or all vitamin B6, vitamin B12, and folic acid, due to factors such as inadequate intake, increased demand, or medication side effects, may lead to metabolic disturbances in one-carbon units and the incidence of hyperhomocysteinemia (HHCy) [59].
The transcription of amino acid-degrading enzymes (AADEs), which are predominantly localized in the liver and exhibit substrate specificity, is regulated by multiple factors, including diet, the gut microbiota, hormonal signaling, and transcription factors [60]. High-protein diets increase the activity and mRNA expression of amino acid-related metabolic enzymes in the liver while concurrently downregulating the expression of enzymes and mRNAs involved in carbohydrate and lipid metabolism. These alterations may be attributed to the utilization of excess amino acids as an energy source, with AMP-activated protein kinase (AMPK) as a potential regulatory factor [61,62,63]. Research has demonstrated that certain hormones, such as glucagon, corticosteroids, growth hormone, and insulin-like growth factor-1 (IGF-1), are involved in both the transcriptional and non-transcriptional regulation of amino acid metabolic enzymes, thereby influencing processes such as amino acid catabolism and the urea cycle [64,65,66] Moreover, several transcription factors contribute to this regulatory network. For example, zinc finger and BTB domain-containing protein 1 (ZBTB1) [67] and Krüppel-like factor (KLF6) [68] regulate the expression of asparagine synthetase (ASNS) and enzymes involved in the metabolism of BCAAs, particularly branched-chain keto acid dehydrogenase E1 subunit beta (BCKDHB). Additionally, Warnhoff et al. elucidated the role of hypoxia-inducible factor-1 (HIF-1) in the negative feedback regulation of cysteine homeostasis. In brief, the elevated level of cysteine promotes the generation of H2S signals, which subsequently stimulate HIF-1-mediated transcription of cdo-1 via the rhy-1/cysl-1/egl-9 signaling pathway, thereby enhancing the cysteine degradation process catalyzed by cysteine dioxygenase (CDO-1) [69].

5.2. Diet

Dietary composition plays a pivotal role in modulating amino acid metabolism. Both the source [70] and quantity [71] of dietary protein influence amino acid digestibility and absorption. Plant-derived proteins typically exhibit lower digestibility due to intrinsic factors such as high proline content, low solubility, and dense peptide structures, as well as extrinsic factors like antinutritional compounds and physical barriers [72] The absorption capacity of the small intestine is limited, and excess dietary proteins that reach the large intestine undergo fermentation into nitrogenous compounds and other metabolites [73]. Furthermore, diets rich in amylose significantly upregulate mRNA levels of amino acid transporters while downregulating amino acid-degrading enzymes (AADEs) in the ileum. These changes are accompanied by increased activation of the mTOR signaling pathway, as indicated by elevated levels of p-mTOR, p-4EBP1, and p-S6K1, thereby reducing amino acid consumption within the gut and promoting their systemic availability for protein synthesis [74] Compared with high-fat or high-protein diets, high-carbohydrate diets increase metabolic efficiency in amino acid utilization, an effect associated with changes in butyrate production [75]. These findings suggest that dietary structure can modulate amino acid metabolism and systemic homeostasis, with potential implications for metabolic disorders such as diabetic cardiomyopathy.

5.3. Amino Acid Transporters

Amino acid transporters (AATs) are regulated by a diverse array of transcription factors. Transcription factors that modulate the expression of excitatory amino acid transporter 1 (EAAT1) can be categorized into positive regulators (such as NF-κB, CREB, and β-catenin) and negative regulators (such as N-myc and YY1) [76] For excitatory amino acid transporter 2 (EAAT2), NF-κB and REST act on the promoter region to upregulate gene expression [77,78]. Similarly, the expression of AAT genes is modulated by the interaction between SIRT6 and ATF4 [79]. Under amino acid starvation stress, the protein kinase GCN2/EIF2AK4 phosphorylates eukaryotic translation initiation factor-2α (eIF2α). Phosphorylated eIF2α attenuates global protein synthesis but concurrently enhances the preferential expression of ATF4 and subsequent AAT genes, ensuring that cells can acquire sufficient amino acids to maintain vital functions during nutrient deprivation [80]. By stabilizing TFE3, glucose stress increases the levels of SLC36A1, a lysosomal amino acid transporter that is associated with mTOR activation and the cellular response to glucose starvation [81]. In immune-activated T cells, c-Myc controls the expression of AATs, particularly SLC7A5, inducing a positive feedforward loop to meet the increased amino acid demands for protein synthesis required for optimal T-cell function [82].
Epigenetic modifications are recognized as heritable and reversible alterations in gene activity that do not involve changes in the DNA sequence [83]. DNA methylation is involved in the regulation of several AAT genes, including slc1a2 (EAAT2) [84,85], slc1a3 (GLAST/EAAT1) [85], slc1a4 (ASCT1) [85], slc6a19 (B0AT1) [86], slc7a1 (CAT-1) [87], slc7a5 (LAT1) [79,85,88], slc7a8 (LAT2) [89,90], slc7a10 (ASC-1) [85], and slc7a11 (xCT) [85,91]. Histone modifications of AATs include methylation (e.g., H3K4me3 [92] and H3K27me3 [93]) and acetylation [94,95], which occur on N-terminal amino acid residues. As functional RNAs that do not encode proteins, non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), have been shown to regulate the expression of AAT genes through complementary base pairing with mRNAs. In particular, miRNAs (e.g., miR-23b-3p [96], miR-122 [97,98], miR-194-5p [98], and miR-328-3p [99]) and lncRNAs (e.g., GSTM3TV2) [100] have been shown to be involved in this regulation.
Additionally, 4F2hc/CD98hc is a single-pass transmembrane protein encoded by slc3a2 and plays a critical role in regulating the stability and transport function of AATs such as xCT [101], LAT1 [102], LAT2 [103], and Glut1 [104]. Similar interactions are observed with ACE2, which forms heterodimers with B0AT1 (SLC6A19) or SIT (SLC6A20) [105,106], as well as rBAT, which associates with B0,+AT (SLC7A9) [107]. Theoretically, any structural or functional changes in these subunits may impact the activity of AATs. For example, the glycosyltransferase B3GNT3 catalyzes the glycosylation of 4F2hc, thereby stabilizing it and enhancing its interaction with xCT. Knockdown of slc3a2 or knockout of B3GNT3 impairs the activity of xCT [108]. Moreover, deficiency of ACE2, a carboxypeptidase that assembles with B0AT1 to form a heterodimer, leads to reduced absorption of amino acids, decreased production of intestinal antimicrobial peptides, and increased susceptibility to inflammatory bowel diseases [106].

6. Amino Acid Metabolism in Diabetic Cardiomyopathy

In recent years, metabolomics analysis of amino acids has been increasingly utilized in the study of diabetes and diabetic cardiomyopathy (DCM) [109]. Several amino acids and intermediate products have been identified as being correlated with the risk of developing diabetes and diabetic cardiomyopathy (DCM), including BCAAs and AAAs [7,109] Some of these metabolites are considered potential specific biomarkers for DCM [110], although the precise mechanisms underlying their involvement remain to be fully elucidated. Herein, we review the latest research advancements in the metabolism of different amino acids in diabetic cardiomyopathy, including the clinical translation of drug targets. Table 2 summarizes recent preclinical studies related to amino acid metabolism in the context of diabetic cardiomyopathy. These include chemical agents, mimetic compounds, and investigational drugs that have shown potential in attenuating inflammation, oxidative stress, and fibrosis in diabetic cardiac models. Although clinical trials, such as randomized controlled trials (RCTs), are still lacking, these preclinical findings may offer valuable insights for future clinical translation.

6.1. Branched-Chain Amino Acids in DCM

BCAAs are the first amino acid group extensively studied in relation to the development of type 2 diabetes and remain the most widely researched to date [7] Branched-chain keto acid dehydrogenase kinase (BCKDK) is a kinase that inhibits BCKDH activity and targets branched-chain keto acid dehydrogenase E1 subunit alpha (BCKDHA), a gene that has been identified as one of the most likely candidate genes for T2DM [149]. PP2Cm, also known as Ppm1k, is the primary activator of BCKDH [150]. Compared with those in control groups, numerous studies have demonstrated significantly elevated plasma levels of BCAAs in both animal models of T1DM and T2DM [151,152] and in patients with these conditions [153,154]. Furthermore, impaired BCAA metabolism results in the accumulation of toxic metabolites [155]. In T1DM, the elevated levels of BCAAs originate from the imbalance between supply (enhanced muscle proteolysis) and consumption (reduced availability of amino group acceptors and diminished BCKDH activity) [151,156,157]. In addition to abnormal metabolic enzymes, genetic variations, overnutrition, and dysbiosis of the gut microbiota are also involved in obesity, insulin resistance and T2DM [156]. Multiple studies have established BCAAs as key markers of diabetes risk [7,8,9] with mechanisms involving interactions with mTOR, as well as increased insulin secretion and pancreatic β-cell depletion caused by BCAA metabolites [7] Recent evidence also suggests a potential link between BCAA metabolites and inflammation, as elevated isoleucine levels were found to correlate with NLRP3 inflammasome activation in patients with nascent metabolic syndrome [158] Such inflammatory activation may represent an underappreciated mechanism contributing to myocardial injury in diabetes-related conditions. Downregulation of BCAA oxidase levels has been shown in experimental models of heart failure and dilated cardiomyopathy [10]. Moreover, increased circulating levels of BCAAs and BCKAs are considered predictive factors for the incidence of coronary heart disease, congestive heart failure, and cardiovascular disease [110]. For example, 3-hydroxyisobutyrate (3-HIB), a valine metabolite, may be highly important for regulating the flexibility of lipid metabolism in the heart [159]. Although alterations in BCAA metabolism have been linked to various heart diseases, with mechanisms including cardiac insulin resistance, myocardial hypertrophy, and impaired myocardial contractility [10,116,156], research on this association in diabetic cardiomyopathy remains insufficient [10]. Given that BCAAs are not major substrates for cardiac fuel supply, it has been proposed that high levels of BCAAs and BCKAs act as signaling molecules to negatively regulate cardiac energy metabolism [10] (Figure 3).
Increased abundance of BCAA-producing bacteria in the gut of diabetic mice [112] and the reduced capacity to degrade BCAAs [160] result in elevated plasma levels of BCAAs. Excess circulating BCAA levels contribute to increased cardiac BCAA levels via the PPARα-FGF21-Zbtb7c-LAT1 axis, leading to cardiac fibrosis and dysfunction [160]. Moreover, dysregulation of the gut microbiota and BCAA metabolism is implicated in diabetes-induced autonomic imbalance, which ultimately leads to cardiac damage [112]. Periostin, an extracellular matrix protein, can be stimulated and upregulated by high-glucose conditions through a TGF-β/Smad-dependent mechanism. In cardiac fibroblasts, impaired BCAA catabolism aggravates cardiac fibrosis via the periostin/NAP1L2/SIRT3 pathway [111]. In rats with T2D, increased levels of AMP deaminase 3 (AMPD3), a negative regulator of BCKDH, impair cardiac energy metabolism. The interaction between AMPD3 and BCKDH offers novel insights into the pathogenesis of DCM [161].
Interventions targeting the metabolic pathway of BCAAs, particularly through the modulation of key enzymes, represent effective strategies for managing diabetes and diabetic cardiomyopathy. The overexpression of PP2Cm reduces BCAA catabolism and oxidative stress, thereby mitigating cardiac ischemia/reperfusion injury [116]. Glucosyringic acid (GA) specifically inhibits periostin expression, suggesting a potential but promising therapeutic approach for DCM [116]. In a mouse model of DCM, pyridostigmine (PYR) enhances vagal nerve activity, restores intestinal microbiota homeostasis, and decreases circulating BCAA levels. Pathologically, PYR alleviates cardiac dysfunction, hypertrophy, and fibrosis [112]. Similarly, the extract from Portulaca oleracea L. relieves T2DM by mediating gut microbiota modification and regulating the expression of BCAA-metabolizing enzymes [113]. Moreover, BT2, a selective allosteric inhibitor of BCKDK, has been demonstrated to increase BCAA catabolism, reduce circulating BCAA levels in ob/ob and diet-induced obesity (DIO) mice and significantly potentiate the hypoglycemic effects of metformin [114]. Several additional studies have similarly reported comparable findings, indicating that BT2 improves pathological remodeling and insulin sensitivity in failing hearts [115,116] and mitigates the adverse effects of 3-mercaptopyruvate sulfurtransferase (3-MST) deficiency on heart failure with a reduced ejection fraction (HFrEF) [117]. In addition to its inhibitory effect on BCKDK, the mitochondrial uncoupling property of BT2, which is a lipophilic weak acid, may account for its excellent efficacy in alleviating cardiovascular diseases [118].
The chemical agent BT2, previously mentioned, is not suitable for human application. Sodium phenylbutyrate (NaPB), a drug commonly employed in the management of urea cycle disorder (UCD), binds to the same allosteric pocket as BT2 to inhibit BCKDK [162], thereby reducing BCAA levels in the plasma of both UCD patients and healthy adults [163,164], as well as the BCAA concentration in culture media observed under experimental conditions [119,165]. Chronic exposure to elevated BCAA levels impairs cellular IRS1/AKT signaling pathways, while the administration of NaPB enhances AKT activation, which is posited as one potential mechanism by which NaPB improves insulin sensitivity [165]. In the liver, fibrates can suppress the expression of the BCKDK gene, thereby specifically enhancing the catabolism of BCAAs and reducing the plasma levels of BCAAs in both rodents and humans, as reported by Vanweert et al. [166]. In contrast to allosteric inhibitors, angiotensin II type 1 receptor (AT1R) blockers such as valsartan act as ATP-competitive inhibitors of BCKDK, increasing BCKDH activity in the rat liver and reducing plasma BCAA concentrations. Similar inhibitory effects on BCKDK have also been observed with candesartan and irbesartan [167].
Certain antidiabetic medications, including glucagon-like peptide-1 (GLP-1) receptor agonists and sodium-dependent glucose transporters 2 (SGLT-2) inhibitors, exhibit significant cardiovascular protective effects. In diabetic mice, empagliflozin has been shown to improve left ventricular diastolic function, an effect attributed to the downregulation of ryanodine receptor (RyR) phosphorylation and reduced spontaneous calcium release from the sarcoplasmic reticulum during diastole but not related to changes in cardiac BCAA metabolism [168]. Nevertheless, another study investigating the effects of empagliflozin in diabetic mice demonstrated that empagliflozin may mitigate DCM-associated myocardial injury by promoting BCAA catabolism and inhibiting the mTOR/p-ULK1 pathway to increase autophagy [120]. Moreover, research indicates that dapagliflozin exerts anti-inflammatory and fibrosis-lowering effects independent of its hypoglycemic action, suggesting the presence of alternative therapeutic targets [169,170]. Tirzepatide, a dual agonist of gastric inhibitory polypeptide (GIP) and GLP-1 receptors, stimulates the catabolism of BCAAs and BCKAs in brown adipose tissue (BAT), which potentially explains the observed improvements in insulin resistance and reductions in systemic BCAA levels in obese diabetic mice [121]. In contrast to SGLT 2 inhibitors, metformin may increase circulating BCAA and BCKA levels through the AMPK-induced inhibitory phosphorylation of BCKDHA [114,171], which could diminish the cardioprotective benefits of its hypoglycemic effects. In fact, recent studies have highlighted that cardiometabolic comorbidities such as diabetes mellitus not only exacerbate myocardial ischemia-reperfusion injury but also attenuate the efficacy of classical cardioprotective strategies, possibly due to impaired redox signaling and altered cellular stress responses, underscoring the need for tailored interventions in diabetic hearts [172,173].
In addition to pharmacological interventions, endurance training promotes the oxidation of BCAAs and enhances the activity and gene expression of BCKDH; intermittent protein restriction (IPR) exerts positive metabolic effects independent of BCKDH activity and mitigates metformin-induced elevations in plasma BCAA and BCKA levels; cold exposure increases the capacity of mitochondria in BAT to utilize and clear BCAAs [114,166,174,175]; these might be potential alternative strategies for reducing plasma BCAA levels. Furthermore, by targeting the endosomal mTOR-ATPase axis, supplementation with specific amino acids (leucine, lysine, arginine) improves the cardiac insulin resistance and contractile dysfunction induced by lipid overload, with the amino acid transporter SLC38A9 also involved in these beneficial effects [124].

6.2. Aromatic Amino Acids in DCM

Similar to BCAAs, the levels of AAAs are regarded as predictors of T2MD [8,9] Interestingly, elevated BCAA levels have been shown to increase AAA levels through competitive inhibition of LAT1 [176].
Phenylalanine levels are regulated by tetrahydrobiopterin-dependent phenylalanine hydroxylase (PAH). Elevated plasma levels of phenylalanine contribute to age-related cardiac aging, characterized by progressive myocardial hypertrophy and interstitial fibrosis and accompanied by diastolic and systolic dysfunction. Administration of tetrahydrobiopterin or dietary restriction of phenylalanine can reverse the age-related elevation of phenylalanine levels and associated cardiac abnormal changes [11]. In diabetic mouse, a significant reduction in tryptophan metabolites and an increase in phenylalanine levels were observed. Meanwhile, rice wine polyphenols (RWPH) and rice wine polypeptides (RWPE) within Chinese rice wine improved these metabolic changes [122]. Administration of the ethanol extract of S. fusiforme to a similar mouse model may enhance glucose tolerance by reducing intestinal BCAA and AAA levels, while also alleviating pathological remodeling in cardiac tissue [123].
Several studies have demonstrated a significant increase in the density and number of sympathetic nerve fibers in diabetic hearts, which are corroborated by elevated levels of norepinephrine (NE) and tyrosine hydroxylase (TH) in the myocardium of T2MD animal models [125,126,127]. The activation of the renin-angiotensin-aldosterone system (RAAS) results in increased NADPH oxidase activity, which may directly promote cardiac fibrosis by triggering the TGF-β1/Smad 2/3 signaling pathway [177]. Treatment with SGLT2 inhibitors (e.g., dapagliflozin) and GIP-1 receptor agonists (e.g., liraglutide), exercise, Stevia rebaudiana (R) extract, and RAAS blockers (e.g., enalapril and losartan) significantly reduces myocardial NE and TH levels, suggesting that attenuation of sympathetic nerve activity may be a key mechanism underlying the cardioprotective effects observed in DCM [125,126,127]. Correspondingly, bilateral percutaneous renal sympathetic denervation improved heart failure and cardiac remodeling in animal models of DCM, further supporting this hypothesis [177,178].
Abnormal tryptophan metabolism has been implicated in the pathogenesis of several cardiovascular diseases, including heart failure [12,13]. Tryptophan undergoes hydroxylation and decarboxylation to form serotonin (5-HT), which, upon binding to the 5-HT2B receptor, can protect the mitochondria in cardiomyocytes from damage. In diabetes, elevated levels of 5-HT in the gut and serum, coupled with reduced expression of cardiac 5-HT2B receptors, are observed and closely associated with the development of DCM. Lactobacillus plantarum and insulin mitigate cardiac apoptosis and fibrosis in diabetic mice by reversing these alterations [128]. Moreover, tryptophan serves as a critical substrate for de novo NAD+ synthesis, and cardiac NAD+ redox imbalance exacerbates diabetic cardiomyopathy [179]. Increased expression of the key enzyme ACMSD in myocardial endothelial cells impairs NAD+ synthesis and increases the risk of cardiac diastolic dysfunction. Inhibition of ACMSD may improve DCM, potentially through activation of the Sirt1/eNOS pathway [13].

6.3. Other Amino Acids in DCM

In addition to the extensively studied branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs), the associations between the metabolism of other categories of amino acids and diabetic cardiomyopathy have been investigated to varying degrees in recent years. Nevertheless, existing studies on several amino acids such as threonine and histidine remain limited. Future investigations should focus on exploring the feasibility and potential mechanisms in these uncharted areas to advance our understanding and therapeutic approaches.

6.3.1. Glycine

As precursors to the natural antioxidant glutathione, glycine and glutamic acid levels in the cardiac tissues of rats with DCM are significantly decreased, which may partially explain the link between impaired glutathione synthesis and myocardial injury resulting from oxidative stress [129]. Glycine and serine are interconverted via serine hydroxymethyltransferase (SHMT), whereas disrupted glucose metabolism in diabetic patients leads to serine deficiency and subsequent glycine depletion [176]. Erzhi Pill, a traditional Chinese medicine, effectively improves the metabolism of glycine and glutamate in the cardiac tissue of diabetic patients [129], suggesting a novel therapeutic strategy for DCM. Glycine supplementation ameliorates the integrity of mitochondria-associated endoplasmic reticulum membranes (MAMs) and insulin signaling in liver cells in vitro but does not confer overall metabolic benefits in mice fed high-fat and high-sugar diets [180]. Future studies should investigate the long-term effects of glycine supplementation on T2DM and associated complications.

6.3.2. Serine

In diabetes, as mentioned earlier, depletion of precursors caused by glycolysis increased hepatic and renal consumption due to gluconeogenesis, and decreased expression of enzymes regulating de novo synthesis, coupled with upregulated expression of catabolic enzymes, collectively contribute to serine deficiency [176,181]. Serine deficiency impairs sphingolipid synthesis and leads to the accumulation of neurotoxic deoxysphinganines, which may play a role in the pathogenesis of diabetic neuropathy, including cardiac autonomic neuropathy [176]. In diabetic models, dietary serine restriction in conjunction with a high-fat diet (HFD) accelerates the development of neuropathy in diabetic mice, whereas dietary serine supplementation can mitigate disease progression [181]. Furthermore, serine deficiency is associated with elevated homocysteine (Hcy) levels, likely due to the reduced availability of key methyl donors and impaired activity of cystathionine β-synthase [176]. The implications of elevated Hcy levels will be discussed in subsequent sections.

6.3.3. Methionine

Reduced levels of insulin-like growth factor 1 (IGF-1) and vitamin B12 are observed in patients with T1DM, and a decrease in IGF-1 may contribute to the pathogenesis of DCM. Research indicates that high-dose oral supplementation with vitamin B12 prevents and reverses signs of diabetic cardiomyopathy, likely through direct clearance of reactive oxygen species (ROS) and restoration of SAMe-DNMT-SOCS1/3-IGF-1 signaling [130]. Elevated serum Hcy is considered a risk factor for cardiac fibrosis in diabetes [131], which is partly mediated by differential expression of miRNAs and changes in β-adrenergic signaling transduction [182]. In DCM mice with hyperhomocysteinemia (HHcy), folic acid treatment improved HHcy but did not alleviate the progression of DCM [183]. Moreover, ginger extract reduces cardiac fibrosis and elevates plasma Hcy levels in diabetic rats [131]. Moreover, Hui Tao et al. investigated the mechanism by which DNMT1-mediated methylation of the androgen receptor (AR) triggers homocysteine-induced autophagy in cardiac fibroblasts, suggesting potential roles for AR and DNA methyltransferase 1 (DNMT1) as novel biomarkers of diabetic cardiac fibrosis [184]. S-Adenosylhomocysteine hydrolase (SAHH) catalyzes the hydrolysis of S-adenosylhomocysteine (SAH); elevated plasma levels of SAH caused by the inhibition of SAHH induce endothelial dysfunction. By means of ultrasound-targeted microbubble technology with cationic microbubbles (CMBs) as carriers, delivery of the SAHH gene to cardiomyocytes improved ventricular function in DCM rats, in which the activation of the AMPK/FOXO3/SIRT3 signaling pathway may play a critical role [185].

6.3.4. Cysteine

Hydrogen sulfide (H2S), the third gasotransmitter identified to possess signal transduction capabilities, is generated primarily in vivo through the catabolism of cysteine. The physiological roles of endogenous H2S have been reviewed [49] and described previously. In recent years, the sources and supplementation of H2S have emerged as a research focus for elucidating the relationship between cysteine metabolism and DCM. For example, the novel endogenous H2S modulator S-propargyl-cysteine activates insulin receptor signal transduction, thereby exerting beneficial effects on DCM [132]. Exogenous H2S supplementation in the form of sodium hydrosulfide (NaHS) prevents lipid deposits in cardiomyocytes by increasing the degradation of sterol regulatory element-binding protein 1 (SREBP1), inhibiting SREBP1 nuclear translocation [133], and regulating Parkin-dependent mitophagy by promoting the S-sulfhydration of ubiquitin-specific protease 8 (USP8) [134], thereby ameliorating DCM. In addition to H2S, which is catalyzed by alanine aminotransferase (AST, also known as AAT), cysteine can be converted into another gaseous signaling molecule, endogenous sulfur dioxide (SO2), which inhibits the autophagic apoptosis of cardiomyocytes and improves myocardial fibrosis in rats with T2D through the PI3K/AKT pathway [135].

6.3.5. Glutamic Acid and Glutamine

The ratio of glutamic acid to glutamine is considered an indicator of overall energy metabolism, and the imbalance between them may lead to the development of T2DM, with potential mechanisms including oxidative damage, dysfunction, and limited neogenesis of β cells [186]. Previous studies have identified several signaling pathways regulated by glutamine metabolism as key therapeutic targets for DCM. For example, piceatannol [136], empagliflozin [137,138], the angiotensin receptor-neprilysin inhibitor (ARNI) LCZ696 [139] and pioglitazone and curcumin [140] relieve diabetes-induced myocardial oxidative damage and cardiac fibrosis by modulating the Nrf2 and/or NF-κB signaling pathways. Additionally, sulforaphane prevents ferroptosis and associated DCM through activation of the AMPK/Nrf2 pathway [141]. Vitamin D downregulates the expression of the AGE cellular receptor (RAGE) gene and O-glycosylation mediated by the hexosamine pathway and reduces cardiac NF-κB activity, thereby alleviating diabetic cardiomyopathy [142]. These discoveries provide new ideas for the prevention and treatment of DCM in terms of oxidative stress, inflammation, and apoptosis.

6.3.6. Lysine

The currently known degradation pathways of L-lysine include the saccharopine pathway and the pipecolic acid pathway. The former is regarded as the predominant metabolic route, and the detailed mechanisms of the latter remain to be fully elucidated. In the saccharopine pathway, lysine and 2-oxoglutaric acid (OG), derived from the tryptophan metabolic pathway, are enzymatically converted into saccharopine [187]. Saccharopine has been identified as a mitochondrial toxin, and its levels are significantly elevated in patients with DCM compared with healthy controls and those with T2DM without myocardial injury, suggesting a potential link among disruption of the lysine metabolic pathway, mitochondrial dysfunction, and the onset of DCM [110]. The level of 2-aminoadipic acid (2-AAA), a downstream metabolite in the saccharopine pathway, is associated with obesity and metabolic syndrome and can predict the future risk of T2DM. Interestingly, an analysis by Cristina Razquin et al. revealed that higher lysine levels were correlated with an increased future risk of CVD exclusively in diabetic patients [188].

6.3.7. Arginine

Nitric oxide (NO) synthesis is a significant metabolic pathway of arginine. In the presence of adequate tetrahydrobiopterin (BH4) as a cofactor and L-arginine as a substrate, endothelial nitric oxide synthase (eNOS) catalyzes the conversion of L-arginine to NO. In T2DM, increased conversion of BH4 to BH2 leads to eNOS uncoupling, resulting in electron transfer to oxygen and consequently increased production of reactive oxygen species (ROS) and reduced NO generation. Additionally, T2DM enhances arginase activity and increases arginine consumption. The combined supplementation of sepiapterin, the precursor of BH4, and L-citrulline, the precursor of L-arginine, improved the status of diabetic cardiomyopathy in T2DM mice [143]. Moreover, previous studies have demonstrated that arginase inhibitors can reverse the exacerbation of cellular oxidative stress induced by high-glucose stimulation [189]. NG-dimethyl-L-arginine (ADMA), a natural L-arginine analog, can be metabolized by dimethylarginine dimethylaminohydrolase 2 (DDAH2), competitively inhibiting eNOS and leading to its uncoupling. The overexpression of DDAH2 improves myocardial fibrosis and cardiac function by activating the DDAH/ADMA/eNOS/NO pathway, thereby delaying the progression of DCM [190].
Polyamines (PAs) represent another critical group of metabolic products of arginine, including putrescine, spermidine, and spermine. Metabolomic analysis has confirmed that spermine mitigates myocardial injury in DCM by modulating the metabolic pathways of lipids and amino acids, with acyl-CoA thioesterase 1 (Acot1) potentially serving as one of the key targets [191]. In DCM, the expression levels of various polyamines and metabolic enzymes, such as ornithine decarboxylase (ODC), are altered. Therapy with polyamines attenuates diabetic cardiomyopathy by inhibiting the downregulation of calcium-sensing receptors [144] alleviating endoplasmic reticulum stress, and modulating Wnt signaling pathways [145]. Tyler N Kambis et al. reviewed the changes in miRNAs in hearts with DCM, which regulate cell death, oxidative stress, myocardial hypertrophy and fibrosis, thereby influencing cardiac remodeling [192]. Notably, miRNAs also play crucial regulatory roles in polyamine synthesis, suggesting a potential new approach for targeting polyamine metabolism to improve DCM [192].
The application of L-arginine to the neonatal rat cardiomyocyte cell line H9c2 protects human serum albumin (HSA) from glycosylation, ensuring the proper function of nuclear factor erythroid 2-related factor 2 (Nrf-2), which suggests the potential of L-arginine in mitigating accelerated glycosylation and oxidative stress-associated DCM [146]. Moreover, L-arginine supplementation prevents the development of DCM in mice with T2DM by enhancing mitochondrial function [193]. The combined administration of L-arginine and the cannabinoid 2 (CB2) receptor agonist β-caryophyllene has also been revealed to alleviate inflammation-induced cardiac dysfunction by downregulating NF-κB expression in diabetic hearts [147].

6.3.8. Alanine

Elevated alanine aminotransferase (ALT) levels are observed in high-fat diet (HFD)-induced T2DM mice, suggesting progressive hepatic injury. Concurrently, echocardiographic assessments reveal manifestations of cardiomyopathy characterized by a reduced left ventricular ejection fraction (EF%), fractional shortening (FS%), and fractional area change (FAC%) [194]. In diabetes, the relationship between elevated ALT levels and the development of mild cardiomyopathy, in which metabolic disturbances secondary to liver damage may play a contributory role, remains to be elucidated.

6.3.9. Aspartic Acid and Asparagine

As a serum biomarker indicative of cardiac injury, the level of aspartate aminotransferase (AST) relatively increases when myocardial injury occurs. Several plant-derived compounds that act on DCM animal models have been shown to reduce elevated serum AST levels and exhibit cardioprotective effects against hyperglycemia-induced damage, including Lycium chinense leaf extract [195], Artemisia vulgaris extract [196], olive leaf extract [197], ginger extract [198], sodium houttuyfonate [199], cinnamon [200], quercetin 4′-O-glucoside [148], and thymoquinone [201]. Similar benefits were observed in moderate-to-high intensity endurance exercise [202] and β-aminoisobutyric acid treatment [201]. A study based on the China Cardiometabolic Disease and Cancer Cohort (4C) revealed that during the stage of normal glucose tolerance, abnormal asparagine metabolism may also be indicative of an increased risk of future T2DM development [203]. Moreover, in T1DM, reduced levels of asparagine and glutamine are associated with the progression of cardiovascular autonomic neuropathy induced by diabetes [204]. We hypothesize that, given the close relationship between asparagine and glutamine and the tricarboxylic acid cycle, metabolic disturbances may impact the glycolytic pathway, which is essential for peripheral nerve function; however, more research is needed to elucidate further the mechanisms involved.

6.3.10. Proline

Metabolic syndrome (MetS) is a preclinical high-risk state that is associated with the development of T2DM and cardiovascular diseases. According to research from the China Suboptimal Health Cohort, the metabolism of proline, arginine, and glutathione is influenced in individuals with metabolic syndrome [205]. Fibrosis is a key pathological feature of DCM. Elevated expression of glutamyl-prolyl-tRNA synthetase (EPRS), the sole enzyme in the mammalian cytoplasm responsible for catalyzing the synthesis of prolyl-tRNA, leads to increased formation of prolyl-tRNA, which directly enhances the translation of proline-rich extracellular matrix proteins, thereby promoting myocardial fibrosis under pathological conditions. The inhibition of EPRS can alleviate the profibrotic effects of proline-rich collagens and TGF-β, potentially ameliorating cardiac fibrosis in diabetic cardiomyopathy [206].

7. Conclusions and Perspectives

Although the dysregulation of glucose and lipid metabolism remains the primary initiating factor of DCM, the crosstalk among metabolic pathways and signaling networks underscores the indispensable role of amino acid metabolism in maintaining homeostasis and supporting growth and development. In fact, amino acid metabolism critically drives DCM progression through multifaceted mechanisms, such as (i) the vicious cycle between BCAA metabolism disorders and insulin resistance; (ii) arginine metabolism imbalances and microcirculation disorders; (iii) disturbances in sulfur-containing amino acid metabolism and oxidative stress damage; and (iv) glutamine metabolism reprogramming and the energy crisis. This review systematically summarizes the fundamental pathways of amino acid metabolism, highlights several critical regulatory targets, and aims to provide deeper insights for the understanding and management of diabetic cardiomyopathy.
Here, we discuss various pharmacological agents that have demonstrated efficacy in animal models and in vitro cells by reducing cardiac fibrosis and ventricular pathological remodeling and alleviating diastolic and systolic dysfunction to ameliorate DCM. More significantly, certain drugs and amino acid supplements may offer clinical benefits for specific patient groups. In a randomized controlled trial, NaPB increased peripheral insulin sensitivity in T2DM patients and simultaneously reduced the levels of BCAAs and glucose [207]. Fibrates, as classic lipid-lowering drugs used to reduce cardiovascular risk, have also been shown to improve insulin sensitivity and BCAA metabolism [166]. Although the specific mechanism remains unclear, these findings imply that fibrates may protect diabetic hearts through the glucose, lipid, and amino acid metabolic pathways. Similarly, in addition to blocking AT1R, the inhibitory effect of valsartan on BDKDK may represent another pharmacological mechanism for sartans in the treatment of diabetic heart diseases. In elderly populations, GlyNAC (a combination of glycine and N-acetylcysteine) supplementation improves oxidative stress, endothelial dysfunction, and insulin resistance [208]. Additionally, glutamine supplementation stimulates insulin secretion via increased GLP-1 production, thereby lowering blood glucose levels and ameliorating cardiac risk factors in T2DM patients [186,209]. However, excessive glutamine may exacerbate myocardial injury through increased glutamate production or O-GlcNAcylation [210], which reflects the complexity of amino acid metabolic regulation. Future research should build upon preclinical findings to validate the efficacy and safety of pharmacological agents across both clinical trial settings and diverse patient populations.
Clearly, the management of diabetes complications, particularly diabetic cardiomyopathy, represents a complex and long-term systemic endeavor that requires multidisciplinary collaboration, sustained government support, and the integration of societal resources. Despite significant advancements in understanding the pathophysiology of diabetes and its cardiovascular sequelae, numerous challenges remain unresolved. In this context, it is anticipated that emerging research on amino acid metabolism—including the integration of artificial intelligence-empowered metabolomic profiling and advanced multimodal cardiac imaging techniques—will contribute significantly to improving the prevention, diagnosis, and treatment of diabetes and its cardiac complications [205,211,212,213].
Amino acid metabolism plays a pivotal and multifaceted role in the pathogenesis of diabetic cardiomyopathy. Beyond the classical view of glucose and lipid dysregulation, targeting specific amino acid pathways offers novel opportunities for diagnosis, risk stratification, and therapeutic intervention. Advancing our understanding of these mechanisms may pave the way for more precise and individualized management strategies for patients with diabetes and cardiovascular complications.

Author Contributions

Conceptualization, Y.W.; writing—original draft preparation, Y.W.; visualization, Y.W.; supervision, X.M., S.M., Q.W. and J.Y.; funding acquisition, J.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by grants from the Hubei Key Research and Development Program (No. 2021BCA121) and the National Natural Science Foundation of China (Grant number: 82170392).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The figures in this review article were created via Adobe Illustrator (28.0.0.28).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

5-HT2B 5-hydroxytryptamine receptor 2B
AAA aromatic amino acid
AADE amino acid-degrading enzyme
AAT amino acid transporter
AGE advanced glycation end product
AKT protein kinase B
AMPK AMP-activated protein kinase
ARNI angiotensin receptor-neprilysin inhibitor
AT1R angiotensin II type 1 receptor
B0AT1 broad neutral amino acid transporter 1
BCAA branched-chain amino acid
BCAT branched-chain aminotransferase
BCKA branched-chain α-keto acid
BCKDH branched-chain keto acid dehydrogenase
BCKDHA branched-chain keto acid dehydrogenase E1 subunit alpha
BCKDHB branched-chain keto acid dehydrogenase E1 subunit beta
BCKDK branched-chain keto acid dehydrogenase kinase
BT2 3:6-dichlorobenzo[b]thiophene-2-carboxylic acid
CVD cardiovascular disease
DCM diabetic cardiomyopathy
DIO diet-induced obesity
DM diabetes mellitus
EAA essential amino acid
EF ejection fraction
EGF epidermal growth factor
FGF fibroblast growth factor
H1R histamine receptor 1
H3K4me3 trimethylated histone H3 at lysine 4
HFD high-fat diet
HFrEF heart failure with reduced ejection fraction
HHCy hyperhomocysteinemia
IGF-1 insulin-like growth factor 1
IIS insulin and insulin-like growth factor signaling
IR insulin resistance
LAT large neutral amino acid transporter
LC-MS liquid chromatography-mass spectrometry
MetS metabolic syndrome
mTOR mammalian target of rapamycin
NaPB sodium phenylbutyrate
NEAA non-essential amino acid
RAAS renin-angiotensin-aldosterone system
RAGE advanced glycation end product (AGE) cellular receptor
ROS reactive oxygen species
SCFA short-chain fatty acid
SGLT-2 sodium-glucose cotransporter 2
Sirt1 sirtuin 1
SLC1A1 solute carrier family 1 member 1
TAC tricarboxylic acid
T1DM type 1 diabetes mellitus
TGF-β transforming growth factor beta

References

  1. NCD Risk Factor Collaboration (NCD-RisC). Worldwide trends in diabetes prevalence and treatment from 1990 to 2022: A pooled analysis of 1108 population-representative studies with 141 million participants. Lancet 2024, 404, 2077–2093. [Google Scholar] [CrossRef]
  2. Wong, N.D.; Sattar, N. Cardiovascular risk in diabetes mellitus: Epidemiology, assessment and prevention. Nat. Rev. Cardiol. 2023, 20, 685–695. [Google Scholar] [CrossRef] [PubMed]
  3. Marx, N.; Federici, M.; Schütt, K.; Müller-Wieland, D.; Ajjan, R.A.; Antunes, M.J.; Christodorescu, R.M.; Crawford, C.; Di Angelantonio, E.; Eliasson, B.; et al. 2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes. Eur. Heart J. 2023, 44, 4043–4140. [Google Scholar] [CrossRef]
  4. Rubler, S.; Dlugash, J.; Yuceoglu, Y.Z.; Kumral, T.; Branwood, A.W.; Grishman, A. New type of cardiomyopathy associated with diabetic glomerulosclerosis. Am. J. Cardiol. 1972, 30, 595–602. [Google Scholar] [CrossRef] [PubMed]
  5. Seferović, P.M.; Paulus, W.J.; Rosano, G.; Polovina, M.; Petrie, M.C.; Jhund, P.S.; Tschöpe, C.; Sattar, N.; Piepoli, M.; Papp, Z.; et al. Diabetic myocardial disorder. A clinical consensus statement of the Heart Failure Association of the ESC and the ESC Working Group on Myocardial & Pericardial Diseases. Eur. J. Heart Fail. 2024, 26, 1893–1903. [Google Scholar] [CrossRef]
  6. Tan, Y.; Zhang, Z.; Zheng, C.; Wintergerst, K.A.; Keller, B.B.; Cai, L. Mechanisms of diabetic cardiomyopathy and potential therapeutic strategies: Preclinical and clinical evidence. Nat. Rev. Cardiol. 2020, 17, 585–607. [Google Scholar] [CrossRef] [PubMed]
  7. Morze, J.; Wittenbecher, C.; Schwingshackl, L.; Danielewicz, A.; Rynkiewicz, A.; Hu, F.B.; Guasch-Ferré, M. Metabolomics and Type 2 Diabetes Risk: An Updated Systematic Review and Meta-analysis of Prospective Cohort Studies. Diabetes Care 2022, 45, 1013–1024. [Google Scholar] [CrossRef]
  8. Rivas-Tumanyan, S.; Pacheco, L.S.; Haslam, D.E.; Morou-Bermudez, E.; Liang, L.; Tucker, K.L.; Joshipura, K.J.; Bhupathiraju, S.N. Branched-Chain and Aromatic Amino Acids, Type 2 Diabetes, and Cardiometabolic Risk Factors among Puerto Rican Adults. Nutrients 2024, 16, 2562. [Google Scholar] [CrossRef]
  9. Wang, M.; Ou, Y.; Yuan, X.-L.; Zhu, X.-F.; Niu, B.; Kang, Z.; Zhang, B.; Ahmed, A.; Xing, G.-Q.; Su, H. Heterogeneously elevated branched-chain/aromatic amino acids among new-onset type-2 diabetes mellitus patients are potentially skewed diabetes predictors. World J. Diabetes 2024, 15, 53–71. [Google Scholar] [CrossRef]
  10. Karwi, Q.G.; Lopaschuk, G.D. Branched-Chain Amino Acid Metabolism in the Failing Heart. Cardiovasc. Drugs Ther. 2023, 37, 413–420. [Google Scholar] [CrossRef]
  11. Czibik, G.; Mezdari, Z.; Murat Altintas, D.; Bréhat, J.; Pini, M.; d’Humières, T.; Delmont, T.; Radu, C.; Breau, M.; Liang, H.; et al. Dysregulated Phenylalanine Catabolism Plays a Key Role in the Trajectory of Cardiac Aging. Circulation 2021, 144, 559–574. [Google Scholar] [CrossRef]
  12. Lund, A.; Nordrehaug, J.E.; Slettom, G.; Solvang, S.-E.H.; Pedersen, E.K.R.; Midttun, Ø.; Ulvik, A.; Ueland, P.M.; Nygård, O.; Giil, L.M. Plasma kynurenines and prognosis in patients with heart failure. PLoS ONE 2020, 15, e0227365. [Google Scholar] [CrossRef]
  13. Zeng, F.; Zhou, P.; Wang, M.; Xie, L.; Huang, X.; Wang, Y.; Huang, J.; Shao, X.; Yang, Y.; Liu, W.; et al. ACMSD mediated de novo NAD+ biosynthetic impairment in cardiac endothelial cells as a potential therapeutic target for diabetic cardiomyopathy. Diabetes Res. Clin. Pract. 2023, 206, 111014. [Google Scholar] [CrossRef] [PubMed]
  14. El-Hattab, A.W. Serine biosynthesis and transport defects. Mol. Genet. Metab. 2016, 118, 153–159. [Google Scholar] [CrossRef] [PubMed]
  15. Chiu, M.; Taurino, G.; Bianchi, M.G.; Kilberg, M.S.; Bussolati, O. Asparagine Synthetase in Cancer: Beyond Acute Lymphoblastic Leukemia. Front. Oncol. 2019, 9, 1480. [Google Scholar] [CrossRef] [PubMed]
  16. Kalhan, S.C.; Hanson, R.W. Resurgence of serine: An often neglected but indispensable amino Acid. J. Biol. Chem. 2012, 287, 19786–19791. [Google Scholar] [CrossRef]
  17. Bröer, S.; Gauthier-Coles, G. Amino Acid Homeostasis in Mammalian Cells with a Focus on Amino Acid Transport. J. Nutr. 2022, 152, 16–28. [Google Scholar] [CrossRef]
  18. Hushmandi, K.; Einollahi, B.; Saadat, S.H.; Lee, E.H.C.; Farani, M.R.; Okina, E.; Huh, Y.S.; Nabavi, N.; Salimimoghadam, S.; Kumar, A.P. Amino acid transporters within the solute carrier superfamily: Underappreciated proteins and novel opportunities for cancer therapy. Mol. Metab. 2024, 84, 101952. [Google Scholar] [CrossRef]
  19. Wang, Q.; Holst, J. L-type amino acid transport and cancer: Targeting the mTORC1 pathway to inhibit neoplasia. Am. J. Cancer Res. 2015, 5, 1281–1294. [Google Scholar]
  20. Segawa, H.; Fukasawa, Y.; Miyamoto, K.; Takeda, E.; Endou, H.; Kanai, Y. Identification and functional characterization of a Na+-independent neutral amino acid transporter with broad substrate selectivity. J. Biol. Chem. 1999, 274, 19745–19751. [Google Scholar] [CrossRef]
  21. Gauthier-Coles, G.; Fairweather, S.J.; Bröer, A.; Bröer, S. Do Amino Acid Antiporters Have Asymmetric Substrate Specificity? Biomolecules 2023, 13, 301. [Google Scholar] [CrossRef]
  22. Oparija, L.; Rajendran, A.; Poncet, N.; Verrey, F. Anticipation of food intake induces phosphorylation switch to regulate basolateral amino acid transporter LAT4 (SLC43A2) function. J. Physiol. 2019, 597, 521–542. [Google Scholar] [CrossRef] [PubMed]
  23. Rajendran, A.; Poncet, N.; Oparija-Rogenmozere, L.; Herzog, B.; Verrey, F. Tissue-specific deletion of mouse basolateral uniporter LAT4 (Slc43a2) reveals its crucial role in small intestine and kidney amino acid transport. J. Physiol. 2020, 598, 5109–5132. [Google Scholar] [CrossRef] [PubMed]
  24. Gunarathne, R.; Guan, X.; Feng, T.; Zhao, Y.; Lu, J. L-lysine dietary supplementation for childhood and adolescent growth: Promises and precautions. J. Adv. Res. 2025, 70, 571–586. [Google Scholar] [CrossRef] [PubMed]
  25. Jang, J.; Kim, Y.; Song, T.; Park, S.; Kim, H.-J.; Koh, J.-H.; Cho, Y.; Park, S.-Y.; Sadayappan, S.; Kwak, H.-B.; et al. Free essential amino acid feeding improves endurance during resistance training via DRP1-dependent mitochondrial remodelling. J. Cachexia Sarcopenia Muscle 2024, 15, 1651–1663. [Google Scholar] [CrossRef]
  26. Zhang, Y.; Lin, S.; Peng, J.; Liang, X.; Yang, Q.; Bai, X.; Li, Y.; Li, J.; Dong, W.; Wang, Y.; et al. Amelioration of hepatic steatosis by dietary essential amino acid-induced ubiquitination. Mol. Cell 2022, 82, 1528–1542.e10. [Google Scholar] [CrossRef]
  27. Knaus, L.S.; Basilico, B.; Malzl, D.; Gerykova Bujalkova, M.; Smogavec, M.; Schwarz, L.A.; Gorkiewicz, S.; Amberg, N.; Pauler, F.M.; Knittl-Frank, C.; et al. Large neutral amino acid levels tune perinatal neuronal excitability and survival. Cell 2023, 186, 1950–1967.E25. [Google Scholar] [CrossRef]
  28. Hu, D.; Liu, J.; Yu, W.; Li, C.; Huang, L.; Mao, W.; Lu, Z. Tryptophan intake, not always the more the better. Front. Nutr. 2023, 10, 1140054. [Google Scholar] [CrossRef]
  29. Skvorak, K.; Liu, J.; Kruse, N.; Mehmood, R.; Das, S.; Jenne, S.; Chng, C.; Lao, U.L.; Duan, D.; Asfaha, J.; et al. Oral enzyme therapy for maple syrup urine disease (MSUD) suppresses plasma leucine levels in intermediate MSUD mice and healthy nonhuman primates. J. Inherit. Metab. Dis. 2023, 46, 1089–1103. [Google Scholar] [CrossRef]
  30. Kaspy, M.S.; Hannaian, S.J.; Bell, Z.W.; Churchward-Venne, T.A. The effects of branched-chain amino acids on muscle protein synthesis, muscle protein breakdown and associated molecular signalling responses in humans: An update. Nutr. Res. Rev. 2024, 37, 273–286. [Google Scholar] [CrossRef]
  31. Harper, A.E.; Miller, R.H.; Block, K.P. Branched-chain amino acid metabolism. Annu. Rev. Nutr. 1984, 4, 409–454. [Google Scholar] [CrossRef] [PubMed]
  32. Ma, N.; Ma, X. Dietary Amino Acids and the Gut-Microbiome-Immune Axis: Physiological Metabolism and Therapeutic Prospects. Compr. Rev. Food Sci. Food Saf. 2019, 18, 221–242. [Google Scholar] [CrossRef] [PubMed]
  33. Salas-Garrucho, F.M.; Carrillo-Moreno, A.; Contreras, L.M.; Rodríguez-Vico, F.; Clemente-Jiménez, J.M.; Las Heras-Vázquez, F.J. Exploring the Kinetics and Thermodynamics of a Novel Histidine Ammonia-Lyase from Geobacillus kaustophilus. Int. J. Mol. Sci. 2024, 25, 163. [Google Scholar] [CrossRef]
  34. Boros, K.; Moisă, M.E.; Nagy, C.L.; Paizs, C.; Toşa, M.I.; Bencze, L.C. Robust, site-specifically immobilized phenylalanine ammonia-lyases for the enantioselective ammonia addition of cinnamic acids. Catal. Sci. Technol. 2021, 11, 5553–5563. [Google Scholar] [CrossRef]
  35. Qian, L.; Li, N.; Lu, X.-C.; Xu, M.; Liu, Y.; Li, K.; Zhang, Y.; Hu, K.; Qi, Y.-T.; Yao, J.; et al. Enhanced BCAT1 activity and BCAA metabolism promotes RhoC activity in cancer progression. Nat. Metab. 2023, 5, 1159–1173. [Google Scholar] [CrossRef]
  36. Li, Y.; Xiong, Z.; Yan, W.; Gao, E.; Cheng, H.; Wu, G.; Liu, Y.; Zhang, L.; Li, C.; Wang, S.; et al. Branched chain amino acids exacerbate myocardial ischemia/reperfusion vulnerability via enhancing GCN2/ATF6/PPAR-α pathway-dependent fatty acid oxidation. Theranostics 2020, 10, 5623–5640. [Google Scholar] [CrossRef] [PubMed]
  37. Yoo, H.C.; Park, S.J.; Nam, M.; Kang, J.; Kim, K.; Yeo, J.H.; Kim, J.-K.; Heo, Y.; Lee, H.S.; Lee, M.Y.; et al. A Variant of SLC1A5 Is a Mitochondrial Glutamine Transporter for Metabolic Reprogramming in Cancer Cells. Cell Metab. 2020, 31, 267–283.E12. [Google Scholar] [CrossRef]
  38. Kunji, E.R.S.; King, M.S.; Ruprecht, J.J.; Thangaratnarajah, C. The SLC25 Carrier Family: Important Transport Proteins in Mitochondrial Physiology and Pathology. Physiology 2020, 35, 302–327. [Google Scholar] [CrossRef]
  39. Sugiyama, Y.; Mori, Y.; Nara, M.; Kotani, Y.; Nagai, E.; Kawada, H.; Kitamura, M.; Hirano, R.; Shimokawa, H.; Nakagawa, A.; et al. Gut bacterial aromatic amine production: Aromatic amino acid decarboxylase and its effects on peripheral serotonin production. Gut Microbes. 2022, 14, 2128605. [Google Scholar] [CrossRef]
  40. Gabrawy, M.M.; Westbrook, R.; King, A.; Khosravian, N.; Ochaney, N.; DeCarvalho, T.; Wang, Q.; Yu, Y.; Huang, Q.; Said, A.; et al. Dual treatment with kynurenine pathway inhibitors and NAD+ precursors synergistically extends life span in Drosophila. Aging Cell 2024, 23, e14102. [Google Scholar] [CrossRef]
  41. Liu, J.-J.; Ching, J.; Wee, H.N.; Liu, S.; Gurung, R.L.; Lee, J.; M, Y.; Zheng, H.; Lee, L.S.; Ang, K.; et al. Plasma Tryptophan-Kynurenine Pathway Metabolites and Risk for Progression to End-Stage Kidney Disease in Patients With Type 2 Diabetes. Diabetes Care 2023, 46, 2223–2231. [Google Scholar] [CrossRef] [PubMed]
  42. Cheng, D.; Qin, Z.-S.; Zheng, Y.; Xie, J.-Y.; Liang, S.-S.; Zhang, J.-L.; Feng, Y.-B.; Zhang, Z.-J. Minocycline, a classic antibiotic, exerts psychotropic effects by normalizing microglial neuroinflammation-evoked tryptophan-kynurenine pathway dysregulation in chronically stressed male mice. Brain Behav. Immun. 2023, 107, 305–318. [Google Scholar] [CrossRef] [PubMed]
  43. Stone, T.W.; Williams, R.O. Modulation of T cells by tryptophan metabolites in the kynurenine pathway. Trends Pharmacol. Sci. 2023, 44, 442–456. [Google Scholar] [CrossRef]
  44. Liang, H.; Zhan, J.; Chen, Y.; Xing, Z.; He, Z.N.T.; Liu, Y.; Li, X.; Chen, Y.; Li, Z.; Kuang, C.; et al. Tryptophan deficiency induced by indoleamine 2,3-dioxygenase 1 results in glucose transporter 1-dependent promotion of aerobic glycolysis in pancreatic cancer. MedComm 2024, 5, e555. [Google Scholar] [CrossRef]
  45. Morales-Puerto, N.; Giménez-Gómez, P.; Pérez-Hernández, M.; Abuin-Martínez, C.; Gil de Biedma-Elduayen, L.; Vidal, R.; Gutiérrez-López, M.D.; O’Shea, E.; Colado, M.I. Addiction and the kynurenine pathway: A new dancing couple? Pharmacol. Ther. 2021, 223, 107807. [Google Scholar] [CrossRef] [PubMed]
  46. Song, F.; Yang, X.; Zhu, B.; Xiong, Y.; Song, Z.; Yang, X.; Zheng, Y. Histamine deficiency deteriorates LPS-induced periodontal diseases in a murine model via NLRP3/Caspase-1 pathway. Int. Immunopharmacol. 2023, 115, 109630. [Google Scholar] [CrossRef]
  47. Pham, V.N.; Bruemmer, K.J.; Toh, J.D.W.; Ge, E.J.; Tenney, L.; Ward, C.C.; Dingler, F.A.; Millington, C.L.; Garcia-Prieto, C.A.; Pulos-Holmes, M.C.; et al. Formaldehyde regulates S-adenosylmethionine biosynthesis and one-carbon metabolism. Science 2023, 382, eabp9201. [Google Scholar] [CrossRef]
  48. McBride, M.J.; Hunter, C.J.; Zhang, Z.; TeSlaa, T.; Xu, X.; Ducker, G.S.; Rabinowitz, J.D. Glycine homeostasis requires reverse SHMT flux. Cell Metab. 2024, 36, 103–115.E4. [Google Scholar] [CrossRef]
  49. Hu, X.; Xiao, Y.; Sun, J.; Ji, B.; Luo, S.; Wu, B.; Zheng, C.; Wang, P.; Xu, F.; Cheng, K.; et al. New possible silver lining for pancreatic cancer therapy: Hydrogen sulfide and its donors. Acta Pharm. Sin. B 2021, 11, 1148–1157. [Google Scholar] [CrossRef]
  50. Lundberg, J.O.; Weitzberg, E. Nitric oxide signaling in health and disease. Cell 2022, 185, 2853–2878. [Google Scholar] [CrossRef]
  51. McGarrah, R.W.; White, P.J. Branched-chain amino acids in cardiovascular disease. Nat. Rev. Cardiol. 2023, 20, 77–89. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, L.; Jain, M.K. Circadian regulation of cardiac metabolism. J. Clin. Investig. 2021, 131, e148276. [Google Scholar] [CrossRef]
  53. Radjabzadeh, D.; Bosch, J.A.; Uitterlinden, A.G.; Zwinderman, A.H.; Ikram, M.A.; van Meurs, J.B.J.; Luik, A.I.; Nieuwdorp, M.; Lok, A.; van Duijn, C.M.; et al. Gut microbiome-wide association study of depressive symptoms. Nat. Commun. 2022, 13, 7128. [Google Scholar] [CrossRef] [PubMed]
  54. Söllinger, A.; Séneca, J.; Borg Dahl, M.; Motleleng, L.L.; Prommer, J.; Verbruggen, E.; Sigurdsson, B.D.; Janssens, I.; Peñuelas, J.; Urich, T.; et al. Down-regulation of the bacterial protein biosynthesis machinery in response to weeks, years, and decades of soil warming. Sci. Adv. 2022, 8, eabm3230. [Google Scholar] [CrossRef] [PubMed]
  55. Oxenkrug, G.; Navrotska, V. Extension of life span by down-regulation of enzymes catalyzing tryptophan conversion into kynurenine: Possible implications for mechanisms of aging. Exp. Biol. Med. (Maywood) 2023, 248, 573–577. [Google Scholar] [CrossRef]
  56. Cheng, Y.; Liu, Y.; Chen, D.; Zhou, Y.; Yu, S.; Lin, H.; Liao, C.K.; Lin, H.; Xu, P.; Huang, M. Dual effects of quercetin on protein digestion and absorption in the digestive tract. Food Chem. 2021, 358, 129891. [Google Scholar] [CrossRef]
  57. Li, X.; Liu, H.; Wu, X.; Xu, R.; Ma, X.; Zhang, C.; Song, Z.; Peng, Y.; Ni, T.; Xu, Y. Exploring the interactions of naringenin and naringin with trypsin and pepsin: Experimental and computational modeling approaches. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2021, 258, 119859. [Google Scholar] [CrossRef]
  58. Wang, K.; Luo, L.; Xu, X.; Chen, X.; He, Q.; Zou, Z.; Wang, S.; Liang, S. LC-MS-based plasma metabolomics study of the intervention effect of different polar parts of hawthorn on gastrointestinal motility disorder rats. Biomed. Chromatogr. 2021, 35, e5076. [Google Scholar] [CrossRef]
  59. Nieraad, H.; Pannwitz, N.; Bruin, N.d.; Geisslinger, G.; Till, U. Hyperhomocysteinemia: Metabolic Role and Animal Studies with a Focus on Cognitive Performance and Decline-A Review. Biomolecules 2021, 11, 1546. [Google Scholar] [CrossRef]
  60. Torres, N.; Tobón-Cornejo, S.; Velazquez-Villegas, L.A.; Noriega, L.G.; Alemán-Escondrillas, G.; Tovar, A.R. Amino Acid Catabolism: An Overlooked Area of Metabolism. Nutrients 2023, 15, 3378. [Google Scholar] [CrossRef]
  61. Alam, M.S.; Liang, X.-F.; Liu, L.; He, S.; Kuang, Y.; Hoseinifar, S.H.; Dawar, F.U. Growth and Metabolic Response of Chinese Perch to Different Dietary Protein-to-Energy Ratios in Artificial Diets. Int. J. Mol. Sci. 2019, 20, 5983. [Google Scholar] [CrossRef] [PubMed]
  62. Yu, Y.; Ai, C.; Luo, C.; Yuan, J. Effect of Dietary Crude Protein and Apparent Metabolizable Energy Levels on Growth Performance, Nitrogen Utilization, Serum Parameter, Protein Synthesis, and Amino Acid Metabolism of 1- to 10-Day-Old Male Broilers. Int. J. Mol. Sci. 2024, 25, 7431. [Google Scholar] [CrossRef] [PubMed]
  63. Heibel, S.K.; McGuire, P.J.; Haskins, N.; Majumdar, H.D.; Rayavarapu, S.; Nagaraju, K.; Hathout, Y.; Brown, K.; Tuchman, M.; Caldovic, L. AMP-activated protein kinase signaling regulated expression of urea cycle enzymes in response to changes in dietary protein intake. J. Inherit. Metab. Dis. 2019, 42, 1088–1096. [Google Scholar] [CrossRef] [PubMed]
  64. Elmelund, E.; Galsgaard, K.D.; Johansen, C.D.; Trammell, S.A.J.; Bomholt, A.B.; Winther-Sørensen, M.; Hunt, J.E.; Sørensen, C.M.; Kruse, T.; Lau, J.F.; et al. Opposing effects of chronic glucagon receptor agonism and antagonism on amino acids, hepatic gene expression, and alpha cells. iScience 2022, 25, 105296. [Google Scholar] [CrossRef]
  65. Dahabiyeh, L.A.; Malkawi, A.K.; Wang, X.; Colak, D.; Mujamammi, A.H.; Sabi, E.M.; Li, L.; Dasouki, M.; Abdel Rahman, A.M. Dexamethasone-Induced Perturbations in Tissue Metabolomics Revealed by Chemical Isotope Labeling LC-MS analysis. Metabolites 2020, 10, 42. [Google Scholar] [CrossRef]
  66. Grøfte, T.; Wolthers, T.; Jensen, S.A.; Møller, N.; Jørgensen, J.O.; Tygstrup, N.; Orskov, H.; Vilstrup, H. Effects of growth hormone and insulin-like growth factor-I singly and in combination on in vivo capacity of urea synthesis, gene expression of urea cycle enzymes, and organ nitrogen contents in rats. Hepatology 1997, 25, 964–969. [Google Scholar] [CrossRef]
  67. Williams, R.T.; Guarecuco, R.; Gates, L.A.; Barrows, D.; Passarelli, M.C.; Carey, B.; Baudrier, L.; Jeewajee, S.; La, K.; Prizer, B.; et al. ZBTB1 Regulates Asparagine Synthesis and Leukemia Cell Response to L-Asparaginase. Cell Metab. 2020, 31, 852–861.e6. [Google Scholar] [CrossRef]
  68. Piret, S.E.; Guo, Y.; Attallah, A.A.; Horne, S.J.; Zollman, A.; Owusu, D.; Henein, J.; Sidorenko, V.S.; Revelo, M.P.; Hato, T.; et al. Krüppel-like factor 6-mediated loss of BCAA catabolism contributes to kidney injury in mice and humans. Proc. Natl. Acad. Sci. USA 2021, 118, e2024414118. [Google Scholar] [CrossRef]
  69. Warnhoff, K.; Bhattacharya, S.; Snoozy, J.; Breen, P.C.; Ruvkun, G. Hypoxia-inducible factor induces cysteine dioxygenase and promotes cysteine homeostasis in Caenorhabditis elegans. Elife 2024, 12, RP89173. [Google Scholar] [CrossRef]
  70. Huang, Z.; Shi, X.; Zhou, G.; Li, C. Dietary soy, pork and chicken proteins induce distinct nitrogen metabolism in rat liver. Food Chem. Mol. Sci. 2021, 3, 100050. [Google Scholar] [CrossRef]
  71. Zhao, J.; Zhang, X.; Liu, H.; Brown, M.A.; Qiao, S. Dietary Protein and Gut Microbiota Composition and Function. Curr. Protein Pept. Sci. 2019, 20, 145–154. [Google Scholar] [CrossRef]
  72. Joye, I. Protein Digestibility of Cereal Products. Foods 2019, 8, 199. [Google Scholar] [CrossRef]
  73. Bartlett, A.; Kleiner, M. Dietary protein and the intestinal microbiota: An understudied relationship. iScience 2022, 25, 105313. [Google Scholar] [CrossRef]
  74. Lv, X.; Zhou, C.; Ran, T.; Jiao, J.; Liu, Y.; Tan, Z.; Tang, S.; Kang, J.; Xie, J.; Chen, L.; et al. Dietary amylose:amylopectin ratio influences the expression of amino acid transporters and enzyme activities for amino acid metabolism in the gastrointestinal tract of goats. Br. J. Nutr. 2022, 127, 1121–1131. [Google Scholar] [CrossRef]
  75. Dong, Y.; Zhu, Q.; Li, Y.; Wang, R.; Xu, W.; Tang, X.; Li, X.; Lv, X.; Kong, X.; Cai, L.; et al. Longevity extension in rats via improved redox homeostasis with high carbohydrate diet intervention from weaning to adulthood: A comprehensive multi-omics study. Food Funct. 2024, 15, 7920–7935. [Google Scholar] [CrossRef]
  76. Liu, L.; Tian, X.; Li, W. Mechanistic study of the anti-excitatory amino acid toxicity of Bushen Zhichan decoction for Parkinson’s disease based on the transcriptional regulation of EAAT1 by YY1. J. Ethnopharmacol. 2024, 325, 117857. [Google Scholar] [CrossRef]
  77. Gan, Z.; Guo, Y.; Zhao, M.; Ye, Y.; Liao, Y.; Liu, B.; Yin, J.; Zhou, X.; Yan, Y.; Yin, Y.; et al. Excitatory amino acid transporter supports inflammatory macrophage responses. Sci. Bull. 2024, 69, 2405–2419. [Google Scholar] [CrossRef]
  78. Pajarillo, E.; Digman, A.; Nyarko-Danquah, I.; Son, D.-S.; Soliman, K.F.A.; Aschner, M.; Lee, E. Astrocytic transcription factor REST upregulates glutamate transporter EAAT2, protecting dopaminergic neurons from manganese-induced excitotoxicity. J. Biol. Chem. 2021, 297, 101372. [Google Scholar] [CrossRef]
  79. Zhang, N.; Yang, X.; Yuan, F.; Zhang, L.; Wang, Y.; Wang, L.; Mao, Z.; Luo, J.; Zhang, H.; Zhu, W.-G.; et al. Increased Amino Acid Uptake Supports Autophagy-Deficient Cell Survival upon Glutamine Deprivation. Cell Rep. 2018, 23, 3006–3020. [Google Scholar] [CrossRef]
  80. Augusto, L.; Amin, P.H.; Wek, R.C.; Sullivan, W.J. Regulation of arginine transport by GCN2 eIF2 kinase is important for replication of the intracellular parasite Toxoplasma gondii. PLoS Pathog. 2019, 15, e1007746. [Google Scholar] [CrossRef] [PubMed]
  81. Lv, S.; Zhang, Z.; Li, Z.; Ke, Q.; Ma, X.; Li, N.; Zhao, X.; Zou, Q.; Sun, L.; Song, T. TFE3-SLC36A1 axis promotes resistance to glucose starvation in kidney cancer cells. J. Biol. Chem. 2024, 300, 107270. [Google Scholar] [CrossRef]
  82. Marchingo, J.M.; Sinclair, L.V.; Howden, A.J.; Cantrell, D.A. Quantitative analysis of how Myc controls T cell proteomes and metabolic pathways during T cell activation. Elife 2020, 9, e53725. [Google Scholar] [CrossRef]
  83. Davalos, V.; Esteller, M. Cancer epigenetics in clinical practice. CA Cancer J. Clin. 2023, 73, 376–424. [Google Scholar] [CrossRef]
  84. Alam, M.A.; Datta, P.K. Epigenetic Regulation of Excitatory Amino Acid Transporter 2 in Neurological Disorders. Front. Pharmacol. 2019, 10, 1510. [Google Scholar] [CrossRef]
  85. Simner, C.; Novakovic, B.; Lillycrop, K.A.; Bell, C.G.; Harvey, N.C.; Cooper, C.; Saffery, R.; Lewis, R.M.; Cleal, J.K. DNA methylation of amino acid transporter genes in the human placenta. Placenta 2017, 60, 64–73. [Google Scholar] [CrossRef]
  86. Tümer, E.; Bröer, A.; Balkrishna, S.; Jülich, T.; Bröer, S. Enterocyte-specific regulation of the apical nutrient transporter SLC6A19 (B(0)AT1) by transcriptional and epigenetic networks. J. Biol. Chem. 2013, 288, 33813–33823. [Google Scholar] [CrossRef]
  87. Li, Y.; Ren, Q.; Zhu, L.; Li, Y.; Li, J.; Zhang, Y.; Zheng, G.; Han, T.; Sun, S.; Feng, F. Involvement of methylation of MicroRNA-122, -125b and -106b in regulation of Cyclin G1, CAT-1 and STAT3 target genes in isoniazid-induced liver injury. BMC Pharmacol. Toxicol. 2018, 19, 11. [Google Scholar] [CrossRef]
  88. Warmbrunn, M.V.; Attaye, I.; Aron-Wisnewsky, J.; Rampanelli, E.; van der Vossen, E.W.J.; Hao, Y.; Koopen, A.; Bergh, P.-O.; Stols-Gonçalves, D.; Mohamed, N.; et al. Oral histidine affects gut microbiota and MAIT cells improving glycemic control in type 2 diabetes patients. Gut Microbes. 2024, 16, 2370616. [Google Scholar] [CrossRef]
  89. Howe, C.G.; Zhou, M.; Wang, X.; Pittman, G.S.; Thompson, I.J.; Campbell, M.R.; Bastain, T.M.; Grubbs, B.H.; Salam, M.T.; Hoyo, C.; et al. Associations between Maternal Tobacco Smoke Exposure and the Cord Blood DNA Methylome. Environ. Health Perspect. 2019, 127, 047009. [Google Scholar] [CrossRef]
  90. Panda, S.K.; Kim, D.-H.; Desai, P.; Rodrigues, P.F.; Sudan, R.; Gilfillan, S.; Cella, M.; Van Dyken, S.J.; Colonna, M. SLC7A8 is a key amino acids supplier for the metabolic programs that sustain homeostasis and activation of type 2 innate lymphoid cells. Proc. Natl. Acad. Sci. USA 2022, 119, e2215528119. [Google Scholar] [CrossRef]
  91. Nath, P.; Alfarsi, L.H.; El-Ansari, R.; Masisi, B.K.; Erkan, B.; Fakroun, A.; Ellis, I.O.; Rakha, E.A.; Green, A.R. The amino acid transporter SLC7A11 expression in breast cancer. Cancer Biol. Ther. 2024, 25, 2291855. [Google Scholar] [CrossRef]
  92. Chen, X.; Wang, Z.; Zhao, X.; Zhang, L.; Zhou, L.; Li, X.; Ge, C.; Zhao, F.; Chen, T.; Xie, H.; et al. STAT5A modulates CDYL2/SLC7A6 pathway to inhibit the proliferation and invasion of hepatocellular carcinoma by targeting to mTORC1. Oncogene 2022, 41, 2492–2504. [Google Scholar] [CrossRef]
  93. Xie, Z.; Zhang, W.; Zhang, Y. Loss of Slc38a4 imprinting is a major cause of mouse placenta hyperplasia in somatic cell nuclear transferred embryos at late gestation. Cell Rep. 2022, 38, 110407. [Google Scholar] [CrossRef]
  94. Tomblin, J.K.; Arthur, S.; Primerano, D.A.; Chaudhry, A.R.; Fan, J.; Denvir, J.; Salisbury, T.B. Aryl hydrocarbon receptor (AHR) regulation of L-Type Amino Acid Transporter 1 (LAT-1) expression in MCF-7 and MDA-MB-231 breast cancer cells. Biochem. Pharmacol. 2016, 106, 94–103. [Google Scholar] [CrossRef]
  95. Paullin, T.; Powell, C.; Menzie, C.; Hill, R.; Cheng, F.; Martyniuk, C.J.; Westerheide, S.D. Spheroid growth in ovarian cancer alters transcriptome responses for stress pathways and epigenetic responses. PLoS ONE 2017, 12, e0182930. [Google Scholar] [CrossRef]
  96. Bacci, M.; Lorito, N.; Ippolito, L.; Ramazzotti, M.; Luti, S.; Romagnoli, S.; Parri, M.; Bianchini, F.; Cappellesso, F.; Virga, F.; et al. Reprogramming of Amino Acid Transporters to Support Aspartate and Glutamate Dependency Sustains Endocrine Resistance in Breast Cancer. Cell Rep. 2019, 28, 104–118.E8. [Google Scholar] [CrossRef]
  97. Sengupta, D.; Cassel, T.; Teng, K.-Y.; Aljuhani, M.; Chowdhary, V.K.; Hu, P.; Zhang, X.; Fan, T.W.M.; Ghoshal, K. Regulation of hepatic glutamine metabolism by miR-122. Mol. Metab. 2020, 34, 174–186. [Google Scholar] [CrossRef]
  98. Liu, X.; Nishikubo, K.; Ohgaki, R.; Okanishi, H.; Okuda, S.; Xu, M.; Kanai, Y. Identification of tumor-suppressive miRNAs that target amino acid transporter LAT1 and exhibit anti-proliferative effects on cholangiocarcinoma cells. J. Pharmacol. Sci. 2024, 154, 301–311. [Google Scholar] [CrossRef]
  99. Yi, W.; Tu, M.-J.; Liu, Z.; Zhang, C.; Batra, N.; Yu, A.-X.; Yu, A.-M. Bioengineered miR-328-3p modulates GLUT1-mediated glucose uptake and metabolism to exert synergistic antiproliferative effects with chemotherapeutics. Acta Pharm. Sin. B 2020, 10, 159–170. [Google Scholar] [CrossRef]
  100. Xiong, G.; Liu, C.; Yang, G.; Feng, M.; Xu, J.; Zhao, F.; You, L.; Zhou, L.; Zheng, L.; Hu, Y.; et al. Long noncoding RNA GSTM3TV2 upregulates LAT2 and OLR1 by competitively sponging let-7 to promote gemcitabine resistance in pancreatic cancer. J. Hematol. Oncol. 2019, 12, 97. [Google Scholar] [CrossRef]
  101. Oda, K.; Lee, Y.; Wiriyasermkul, P.; Tanaka, Y.; Takemoto, M.; Yamashita, K.; Nagamori, S.; Nishizawa, T.; Nureki, O. Consensus mutagenesis approach improves the thermal stability of system xc—transporter, xCT, and enables cryo-EM analyses. Protein Sci. 2020, 29, 2398–2407. [Google Scholar] [CrossRef]
  102. Yan, R.; Zhao, X.; Lei, J.; Zhou, Q. Structure of the human LAT1-4F2hc heteromeric amino acid transporter complex. Nature 2019, 568, 127–130. [Google Scholar] [CrossRef]
  103. Jeckelmann, J.-M.; Fotiadis, D. Sub-Nanometer Cryo-EM Density Map of the Human Heterodimeric Amino Acid Transporter 4F2hc-LAT2. Int. J. Mol. Sci. 2020, 21, 7094. [Google Scholar] [CrossRef]
  104. Ohno, H.; Nakatsu, Y.; Sakoda, H.; Kushiyama, A.; Ono, H.; Fujishiro, M.; Otani, Y.; Okubo, H.; Yoneda, M.; Fukushima, T.; et al. 4F2hc stabilizes GLUT1 protein and increases glucose transport activity. Am. J. Physiol. Cell Physiol. 2011, 300, C1047–C1054. [Google Scholar] [CrossRef]
  105. Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 2020, 367, 1444–1448. [Google Scholar] [CrossRef]
  106. Camargo, S.M.R.; Vuille-Dit-Bille, R.N.; Meier, C.F.; Verrey, F. ACE2 and gut amino acid transport. Clin. Sci. 2020, 134, 2823–2833. [Google Scholar] [CrossRef]
  107. Yan, R.; Li, Y.; Shi, Y.; Zhou, J.; Lei, J.; Huang, J.; Zhou, Q. Cryo-EM structure of the human heteromeric amino acid transporter b0,+AT-rBAT. Sci. Adv. 2020, 6, eaay6379. [Google Scholar] [CrossRef]
  108. Ma, H.; Chen, X.; Mo, S.; Zhang, Y.; Mao, X.; Chen, J.; Liu, Y.; Tong, W.-M.; Lu, Z.; Yu, S.; et al. Targeting N-glycosylation of 4F2hc mediated by glycosyltransferase B3GNT3 sensitizes ferroptosis of pancreatic ductal adenocarcinoma. Cell Death Differ. 2023, 30, 1988–2004. [Google Scholar] [CrossRef]
  109. Chen, Z.-Z.; Gerszten, R.E. Metabolomics and Proteomics in Type 2 Diabetes. Circ. Res. 2020, 126, 1613–1627. [Google Scholar] [CrossRef]
  110. Xiong, R.-Q.; Li, Y.-P.; Lin, L.-P.; Yao, J.-Y. Identification of potential biomarkers for diabetic cardiomyopathy using LC-MS-based metabolomics. Endocr. Connect. 2024, 13, e230384. [Google Scholar] [CrossRef]
  111. Lu, Q.-B.; Fu, X.; Liu, Y.; Wang, Z.-C.; Liu, S.-Y.; Li, Y.-C.; Sun, H.-J. Disrupted cardiac fibroblast BCAA catabolism contributes to diabetic cardiomyopathy via a periostin/NAP1L2/SIRT3 axis. Cell. Mol. Biol. Lett. 2023, 28, 93. [Google Scholar] [CrossRef]
  112. Yang, Y.; Zhao, M.; He, X.; Wu, Q.; Li, D.-L.; Zang, W.-J. Pyridostigmine Protects Against Diabetic Cardiomyopathy by Regulating Vagal Activity, Gut Microbiota, and Branched-Chain Amino Acid Catabolism in Diabetic Mice. Front. Pharmacol. 2021, 12, 647481. [Google Scholar] [CrossRef] [PubMed]
  113. Bao, M.; Hou, K.; Xin, C.; Zeng, D.; Cheng, C.; Zhao, H.; Wang, Z.; Wang, L. Portulaca oleracea L. Extract Alleviated Type 2 Diabetes Via Modulating the Gut Microbiota and Serum Branched-Chain Amino Acid Metabolism. Mol. Nutr. Food Res. 2022, 66, e2101030. [Google Scholar] [CrossRef]
  114. Zhao, X.; Zhang, X.; Pei, J.; Liu, Y.; Niu, W.; Sun, H. Targeting BCAA metabolism to potentiate metformin’s therapeutic efficacy in the treatment of diabetes in mice. Diabetologia 2023, 66, 2139–2153. [Google Scholar] [CrossRef] [PubMed]
  115. Chen, M.; Gao, C.; Yu, J.; Ren, S.; Wang, M.; Wynn, R.M.; Chuang, D.T.; Wang, Y.; Sun, H. Therapeutic Effect of Targeting Branched-Chain Amino Acid Catabolic Flux in Pressure-Overload Induced Heart Failure. J. Am. Heart Assoc. 2019, 8, e011625. [Google Scholar] [CrossRef] [PubMed]
  116. Lian, K.; Guo, X.; Wang, Q.; Liu, Y.; Wang, R.-T.; Gao, C.; Li, C.-Y.; Li, C.-X.; Tao, L. PP2Cm overexpression alleviates MI/R injury mediated by a BCAA catabolism defect and oxidative stress in diabetic mice. Eur. J. Pharmacol. 2020, 866, 172796. [Google Scholar] [CrossRef]
  117. Li, Z.; Xia, H.; Sharp, T.E.; LaPenna, K.B.; Elrod, J.W.; Casin, K.M.; Liu, K.; Calvert, J.W.; Chau, V.Q.; Salloum, F.N.; et al. Mitochondrial H2S Regulates BCAA Catabolism in Heart Failure. Circ. Res. 2022, 131, 222–235. [Google Scholar] [CrossRef]
  118. Acevedo, A.; Jones, A.E.; Danna, B.T.; Turner, R.; Montales, K.P.; Benincá, C.; Reue, K.; Shirihai, O.S.; Stiles, L.; Wallace, M.; et al. The BCKDK inhibitor BT2 is a chemical uncoupler that lowers mitochondrial ROS production and de novo lipogenesis. bioRxiv 2023. [Google Scholar] [CrossRef]
  119. Crossland, H.; Smith, K.; Idris, I.; Phillips, B.E.; Atherton, P.J.; Wilkinson, D.J. Phenylbutyrate, a branched-chain amino acid keto dehydrogenase activator, promotes branched-chain amino acid metabolism and induces muscle catabolism in C2C12 cells. Exp. Physiol. 2021, 106, 585–592. [Google Scholar] [CrossRef]
  120. Zhang, L.; Zhang, H.; Xie, X.; Tie, R.; Shang, X.; Zhao, Q.; Xu, J.; Jin, L.; Zhang, J.; Ye, P. Empagliflozin ameliorates diabetic cardiomyopathy via regulated branched-chain amino acid metabolism and mTOR/p-ULK1 signaling pathway-mediated autophagy. Diabetol. Metab. Syndr. 2023, 15, 93. [Google Scholar] [CrossRef]
  121. Samms, R.J.; Zhang, G.; He, W.; Ilkayeva, O.; Droz, B.A.; Bauer, S.M.; Stutsman, C.; Pirro, V.; Collins, K.A.; Furber, E.C.; et al. Tirzepatide induces a thermogenic-like amino acid signature in brown adipose tissue. Mol. Metab. 2022, 64, 101550. [Google Scholar] [CrossRef] [PubMed]
  122. Yang, J.; Song, J.; Zhou, J.; Lin, H.; Wu, Z.; Liu, N.; Xie, W.; Guo, H.; Chi, J. Functional components of Chinese rice wine can ameliorate diabetic cardiomyopathy through the modulation of autophagy, apoptosis, gut microbiota, and metabolites. Front. Cardiovasc. Med. 2022, 9, 940663. [Google Scholar] [CrossRef] [PubMed]
  123. Wu, S.; Zuo, J.; Cheng, Y.; Zhang, Y.; Zhang, Z.; Wu, M.; Yang, Y.; Tong, H. Ethanol extract of Sargarsum fusiforme alleviates HFD/STZ-induced hyperglycemia in association with modulation of gut microbiota and intestinal metabolites in type 2 diabetic mice. Food Res. Int. 2021, 147, 110550. [Google Scholar] [CrossRef]
  124. Wang, S.; Schianchi, F.; Neumann, D.; Wong, L.-Y.; Sun, A.; van Nieuwenhoven, F.A.; Zeegers, M.P.; Strzelecka, A.; Col, U.; Glatz, J.F.C.; et al. Specific amino acid supplementation rescues the heart from lipid overload-induced insulin resistance and contractile dysfunction by targeting the endosomal mTOR-v-ATPase axis. Mol. Metab. 2021, 53, 101293. [Google Scholar] [CrossRef]
  125. Hussein, A.M.; Eid, E.A.; Taha, M.; Elshazli, R.M.; Bedir, R.F.; Lashin, L.S. Comparative Study of the Effects of GLP1 Analog and SGLT2 Inhibitor against Diabetic Cardiomyopathy in Type 2 Diabetic Rats: Possible Underlying Mechanisms. Biomedicines 2020, 8, 43. [Google Scholar] [CrossRef]
  126. Hussein, A.M.; Eid, E.A.; Bin-Jaliah, I.; Taha, M.; Lashin, L.S. Exercise and Stevia Rebaudiana (R) Extracts Attenuate Diabetic Cardiomyopathy in Type 2 Diabetic Rats: Possible Underlying Mechanisms. Endocr. Metab. Immune. Disord. Drug Targets 2020, 20, 1117–1132. [Google Scholar] [CrossRef]
  127. Yehya, Y.M.; Hussein, A.M.; Ezam, K.; Eid, E.A.; Ibrahim, E.M.; Sarhan, M.A.F.E.; Elsayed, A.; Sarhan, M.E. Blockade of Renin Angiotensin System Ameliorates the Cardiac Arrhythmias and Sympathetic Neural Remodeling in Hearts of Type 2 DM Rat Model. Endocr. Metab. Immune. Disord. Drug Targets 2020, 20, 464–478. [Google Scholar] [CrossRef] [PubMed]
  128. Sefidgari-Abrasi, S.; Roshangar, L.; Karimi, P.; Morshedi, M.; Rahimiyan-Heravan, M.; Saghafi-Asl, M. From the gut to the heart: L. plantarum and inulin administration as a novel approach to control cardiac apoptosis via 5-HT2B and TrkB receptors in diabetes. Clin. Nutr. 2021, 40, 190–201. [Google Scholar] [CrossRef]
  129. Peng, M.; Xia, T.; Zhong, Y.; Zhao, M.; Yue, Y.; Liang, L.; Zhong, R.; Zhang, H.; Li, C.; Cao, X.; et al. Integrative pharmacology reveals the mechanisms of Erzhi Pill, a traditional Chinese formulation, against diabetic cardiomyopathy. J. Ethnopharmacol. 2022, 296, 115474. [Google Scholar] [CrossRef]
  130. Kakoki, M.; Ramanathan, P.V.; Hagaman, J.R.; Grant, R.; Wilder, J.C.; Taylor, J.M.; Charles Jennette, J.; Smithies, O.; Maeda-Smithies, N. Cyanocobalamin prevents cardiomyopathy in type 1 diabetes by modulating oxidative stress and DNMT-SOCS1/3-IGF-1 signaling. Commun. Biol. 2021, 4, 775. [Google Scholar] [CrossRef]
  131. Ilkhanizadeh, B.; Shirpoor, A.; Khadem Ansari, M.H.; Nemati, S.; Rasmi, Y. Protective Effects of Ginger (Zingiber officinale) Extract against Diabetes-Induced Heart Abnormality in Rats. Diabetes Metab. J. 2016, 40, 46–53. [Google Scholar] [CrossRef] [PubMed]
  132. Li, Y.; Xie, K.-F.; Chang, Y.-H.; Wang, C.; Chen, Y.; Wang, M.-J.; Zhu, Y.-C. S-Propargyl-Cysteine Attenuates Diabetic Cardiomyopathy in db/db Mice Through Activation of Cardiac Insulin Receptor Signaling. Front. Cardiovasc. Med. 2021, 8, 737191. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, S.; Wang, M.; Li, H.; Li, Q.; Liu, N.; Dong, S.; Zhao, Y.; Pang, K.; Huang, J.; Ren, C.; et al. Exogenous H2S promotes ubiquitin-mediated degradation of SREBP1 to alleviate diabetic cardiomyopathy via SYVN1 S-sulfhydration. J. Cachexia Sarcopenia Muscle. 2023, 14, 2719–2732. [Google Scholar] [CrossRef] [PubMed]
  134. Sun, Y.; Lu, F.; Yu, X.; Wang, B.; Chen, J.; Lu, F.; Peng, S.; Sun, X.; Yu, M.; Chen, H.; et al. Exogenous H2S Promoted USP8 Sulfhydration to Regulate Mitophagy in the Hearts of db/db Mice. Aging Dis. 2020, 11, 269–285. [Google Scholar] [CrossRef]
  135. Zhao, J.; Wu, Q.; Yang, T.; Nie, L.; Liu, S.; Zhou, J.; Chen, J.; Jiang, Z.; Xiao, T.; Yang, J.; et al. Gaseous signal molecule SO2 regulates autophagy through PI3K/AKT pathway inhibits cardiomyocyte apoptosis and improves myocardial fibrosis in rats with type II diabetes. Korean J. Physiol. Pharmacol. 2022, 26, 541–556. [Google Scholar] [CrossRef]
  136. Li, H.; Shi, Y.; Wang, X.; Li, P.; Zhang, S.; Wu, T.; Yan, Y.; Zhan, Y.; Ren, Y.; Rong, X.; et al. Piceatannol alleviates inflammation and oxidative stress via modulation of the Nrf2/HO-1 and NF-κB pathways in diabetic cardiomyopathy. Chem. Biol. Interact. 2019, 310, 108754. [Google Scholar] [CrossRef]
  137. Cai, W.; Chong, K.; Huang, Y.; Huang, C.; Yin, L. Empagliflozin improves mitochondrial dysfunction in diabetic cardiomyopathy by modulating ketone body metabolism and oxidative stress. Redox. Biol. 2024, 69, 103010. [Google Scholar] [CrossRef]
  138. Li, C.; Zhang, J.; Xue, M.; Li, X.; Han, F.; Liu, X.; Xu, L.; Lu, Y.; Cheng, Y.; Li, T.; et al. SGLT2 inhibition with empagliflozin attenuates myocardial oxidative stress and fibrosis in diabetic mice heart. Cardiovasc. Diabetol. 2019, 18, 15. [Google Scholar] [CrossRef]
  139. Ge, Q.; Zhao, L.; Ren, X.-M.; Ye, P.; Hu, Z.-Y. LCZ696, an angiotensin receptor-neprilysin inhibitor, ameliorates diabetic cardiomyopathy by inhibiting inflammation, oxidative stress and apoptosis. Exp. Biol. Med. (Maywood). 2019, 244, 1028–1039. [Google Scholar] [CrossRef]
  140. Gbr, A.A.; Abdel Baky, N.A.; Mohamed, E.A.; Zaky, H.S. Cardioprotective effect of pioglitazone and curcumin against diabetic cardiomyopathy in type 1 diabetes mellitus: Impact on CaMKII/NF-κB/TGF-β1 and PPAR-γ signaling pathway. Naunyn Schmiedebergs Arch. Pharmacol. 2021, 394, 349–360. [Google Scholar] [CrossRef]
  141. Wang, X.; Chen, X.; Zhou, W.; Men, H.; Bao, T.; Sun, Y.; Wang, Q.; Tan, Y.; Keller, B.B.; Tong, Q.; et al. Ferroptosis is essential for diabetic cardiomyopathy and is prevented by sulforaphane via AMPK/NRF2 pathways. Acta Pharm. Sin. B 2022, 12, 708–722. [Google Scholar] [CrossRef] [PubMed]
  142. Derakhshanian, H.; Djazayery, A.; Javanbakht, M.H.; Eshraghian, M.R.; Mirshafiey, A.; Jahanabadi, S.; Ghadbeigi, S.; Zarei, M.; Alvandi, E.; Djalali, M. Vitamin D downregulates key genes of diabetes complications in cardiomyocyte. J. Cell Physiol. 2019, 234, 21352–21358. [Google Scholar] [CrossRef]
  143. Baumgardt, S.L.; Paterson, M.; Leucker, T.M.; Fang, J.; Zhang, D.X.; Bosnjak, Z.J.; Warltier, D.C.; Kersten, J.R.; Ge, Z.-D. Chronic Co-Administration of Sepiapterin and L-Citrulline Ameliorates Diabetic Cardiomyopathy and Myocardial Ischemia/Reperfusion Injury in Obese Type 2 Diabetic Mice. Circ. Heart Fail. 2016, 9, e002424. [Google Scholar] [CrossRef] [PubMed]
  144. Kambis, T.N.; Tofilau, H.M.N.; Gawargi, F.I.; Chandra, S.; Mishra, P.K. Regulating Polyamine Metabolism by miRNAs in Diabetic Cardiomyopathy. Curr. Diab. Rep. 2021, 21, 52. [Google Scholar] [CrossRef]
  145. Wang, Y.; Chen, J.; Li, S.; Zhang, X.; Guo, Z.; Hu, J.; Shao, X.; Song, N.; Zhao, Y.; Li, H.; et al. Exogenous spermine attenuates rat diabetic cardiomyopathy via suppressing ROS-p53 mediated downregulation of calcium-sensitive receptor. Redox. Biol. 2020, 32, 101514. [Google Scholar] [CrossRef] [PubMed]
  146. Thakur, M.R.; Nachane, S.S.; Tupe, R.S. Alleviation of albumin glycation-induced diabetic cardiomyopathy by L-Arginine: Insights into Nrf-2 signaling. Int. J. Biol. Macromol. 2024, 264, 130478. [Google Scholar] [CrossRef]
  147. Kumawat, V.S.; Kaur, G. Cannabinoid 2 receptor agonist and L-arginine combination attenuates diabetic cardiomyopathy in rats via NF-ĸβ inhibition. Can. J. Physiol. Pharmacol. 2022, 100, 259–271. [Google Scholar] [CrossRef]
  148. Liu, H.; Zhang, Z.; Zhang, L.; Yao, X.; Zhong, X.; Cheng, G.; Wang, L.; Wan, Q. Spiraeoside protects human cardiomyocytes against high glucose-induced injury, oxidative stress, and apoptosis by activation of PI3K/Akt/Nrf2 pathway. J. Biochem. Mol. Toxicol. 2020, 34, e22548. [Google Scholar] [CrossRef]
  149. Zhou, F.; Sheng, C.; Ma, X.; Li, T.; Ming, X.; Wang, S.; Tan, J.; Yang, Y.; Sun, H.; Lu, J.; et al. BCKDH kinase promotes hepatic gluconeogenesis independent of BCKDHA. Cell Death Dis. 2024, 15, 736. [Google Scholar] [CrossRef]
  150. Nishi, K.; Yoshii, A.; Abell, L.; Zhou, B.; Frausto, R.; Ritterhoff, J.; McMillen, T.S.; Sweet, I.; Wang, Y.; Gao, C.; et al. Branched-chain keto acids inhibit mitochondrial pyruvate carrier and suppress gluconeogenesis in hepatocytes. Cell Rep. 2023, 42, 112641. [Google Scholar] [CrossRef]
  151. Holeček, M.; Vodeničarovová, M.; Fingrová, R. Dual Effects of Beta-Hydroxy-Beta-Methylbutyrate (HMB) on Amino Acid, Energy, and Protein Metabolism in the Liver and Muscles of Rats with Streptozotocin-Induced Type 1 Diabetes. Biomolecules 2020, 10, 1475. [Google Scholar] [CrossRef] [PubMed]
  152. Yuan, Z.; Qiao, H.; Wang, Z.; Wang, H.; Han, M.; Zhang, W.; Zhou, Y.; Hassan, H.M.; Zhao, W.; Qin, T. Taohe Chengqi decoction alleviated metabolic-associated fatty liver disease by boosting branched chain amino acids catabolism in the skeletal muscles of type 2 diabetes mellitus. Phytomedicine 2024, 126, 155315. [Google Scholar] [CrossRef] [PubMed]
  153. Karusheva, Y.; Strassburger, K.; Markgraf, D.F.; Zaharia, O.-P.; Bódis, K.; Kössler, T.; Tura, A.; Pacini, G.; Burkart, V.; Roden, M.; et al. Branched-Chain Amino Acids Associate Negatively With Postprandial Insulin Secretion in Recent-Onset Diabetes. J. Endocr. Soc. 2021, 5, bvab067. [Google Scholar] [CrossRef]
  154. Liu, M.; Yang, Y.; Liu, Y.; Peng, X.; Hou, Y.; Zhang, X.; Sun, H.; Shan, C. Serum branched chain amino acids: An effective indicator of diabetic kidney disease. Front. Endocrinol. 2023, 14, 1269633. [Google Scholar] [CrossRef]
  155. Neinast, M.; Murashige, D.; Arany, Z. Branched Chain Amino Acids. Annu. Rev. Physiol. 2019, 81, 139–164. [Google Scholar] [CrossRef]
  156. Dimou, A.; Tsimihodimos, V.; Bairaktari, E. The Critical Role of the Branched Chain Amino Acids (BCAAs) Catabolism-Regulating Enzymes, Branched-Chain Aminotransferase (BCAT) and Branched-Chain α-Keto Acid Dehydrogenase (BCKD), in Human Pathophysiology. Int. J. Mol. Sci. 2022, 23, 4022. [Google Scholar] [CrossRef]
  157. Bai, X.; Long, X.; Song, F.; Chen, B.; Sheng, C.; Tang, C.; Li, L.; Zhang, J.; Zhang, R.; Zhang, J.; et al. High doses of rosuvastatin induce impaired branched-chain amino acid catabolism and lead to insulin resistance. Exp. Physiol. 2023, 108, 961–974. [Google Scholar] [CrossRef] [PubMed]
  158. Jialal, I.; Patel, A.; Devaraj, S.; Adams-Huet, B. Metabolites that activate the inflammasome in nascent metabolic syndrome. J. Diabetes Complicat. 2021, 35, 107836. [Google Scholar] [CrossRef]
  159. Mardinoglu, A.; Gogg, S.; Lotta, L.A.; Stančáková, A.; Nerstedt, A.; Boren, J.; Blüher, M.; Ferrannini, E.; Langenberg, C.; Wareham, N.J.; et al. Elevated Plasma Levels of 3-Hydroxyisobutyric Acid Are Associated With Incident Type 2 Diabetes. EBioMedicine 2018, 27, 151–155. [Google Scholar] [CrossRef]
  160. Zheng, H.; Zhang, X.; Li, C.; Wang, D.; Shen, Y.; Lu, J.; Zhao, L.; Li, X.; Gao, H. BCAA mediated microbiota-liver-heart crosstalk regulates diabetic cardiomyopathy via FGF21. Microbiome. 2024, 12, 157. [Google Scholar] [CrossRef]
  161. Ogawa, T.; Kouzu, H.; Osanami, A.; Tatekoshi, Y.; Sato, T.; Kuno, A.; Fujita, Y.; Ino, S.; Shimizu, M.; Toda, Y.; et al. Downregulation of extramitochondrial BCKDH and its uncoupling from AMP deaminase in type 2 diabetic OLETF rat hearts. Physiol. Rep. 2023, 11, e15608. [Google Scholar] [CrossRef] [PubMed]
  162. Tso, S.-C.; Gui, W.-J.; Wu, C.-Y.; Chuang, J.L.; Qi, X.; Skvora, K.J.; Dork, K.; Wallace, A.L.; Morlock, L.K.; Lee, B.H.; et al. Benzothiophene carboxylate derivatives as novel allosteric inhibitors of branched-chain α-ketoacid dehydrogenase kinase. J. Biol. Chem. 2014, 289, 20583–20593. [Google Scholar] [CrossRef] [PubMed]
  163. Burrage, L.C.; Jain, M.; Gandolfo, L.; Lee, B.H.; Nagamani, S.C.S. Sodium phenylbutyrate decreases plasma branched-chain amino acids in patients with urea cycle disorders. Mol. Genet. Metab. 2014, 113, 131–135. [Google Scholar] [CrossRef] [PubMed]
  164. Osaka, S.; Nakano, S.; Mizuno, T.; Hiraoka, Y.; Minowa, K.; Hirai, S.; Mizutani, A.; Sabu, Y.; Miura, Y.; Shimizu, T.; et al. A randomized trial to examine the impact of food on pharmacokinetics of 4-phenylbutyrate and change in amino acid availability after a single oral administration of sodium 4-phenylbutyrarte in healthy volunteers. Mol. Genet. Metab. 2021, 132, 220–226. [Google Scholar] [CrossRef]
  165. Crossland, H.; Smith, K.; Idris, I.; Phillips, B.E.; Atherton, P.J.; Wilkinson, D.J. Exploring mechanistic links between extracellular branched-chain amino acids and muscle insulin resistance: An in vitro approach. Am. J. Physiol. Cell Physiol. 2020, 319, C1151–C1157. [Google Scholar] [CrossRef]
  166. Vanweert, F.; Schrauwen, P.; Phielix, E. Role of branched-chain amino acid metabolism in the pathogenesis of obesity and type 2 diabetes-related metabolic disturbances BCAA metabolism in type 2 diabetes. Nutr. Diabetes 2022, 12, 35. [Google Scholar] [CrossRef]
  167. Kitaura, Y.; Shindo, D.; Ogawa, T.; Sato, A.; Shimomura, Y. Antihypertensive drug valsartan as a novel BDK inhibitor. Pharmacol. Res. 2021, 167, 105518. [Google Scholar] [CrossRef]
  168. Moellmann, J.; Klinkhammer, B.M.; Droste, P.; Kappel, B.; Haj-Yehia, E.; Maxeiner, S.; Artati, A.; Adamski, J.; Boor, P.; Schütt, K.; et al. Empagliflozin improves left ventricular diastolic function of db/db mice. Biochim. Biophys. Acta-Mol. Basis Dis. 2020, 1866, 165807. [Google Scholar] [CrossRef]
  169. Ye, Y.; Bajaj, M.; Yang, H.-C.; Perez-Polo, J.R.; Birnbaum, Y. SGLT-2 Inhibition with Dapagliflozin Reduces the Activation of the Nlrp3/ASC Inflammasome and Attenuates the Development of Diabetic Cardiomyopathy in Mice with Type 2 Diabetes. Further Augmentation of the Effects with Saxagliptin, a DPP4 Inhibitor. Cardiovasc. Drugs. Ther. 2017, 31, 119–132. [Google Scholar] [CrossRef]
  170. Tian, J.; Zhang, M.; Suo, M.; Liu, D.; Wang, X.; Liu, M.; Pan, J.; Jin, T.; An, F. Dapagliflozin alleviates cardiac fibrosis through suppressing EndMT and fibroblast activation via AMPKα/TGF-β/Smad signalling in type 2 diabetic rats. J. Cell Mol. Med. 2021, 25, 7642–7659. [Google Scholar] [CrossRef]
  171. Rivera, M.E.; Lyon, E.S.; Vaughan, R.A. Effect of metformin on myotube BCAA catabolism. J. Cell Biochem. 2020, 121, 816–827. [Google Scholar] [CrossRef]
  172. Yoneshiro, T.; Wang, Q.; Tajima, K.; Matsushita, M.; Maki, H.; Igarashi, K.; Dai, Z.; White, P.J.; McGarrah, R.W.; Ilkayeva, O.R.; et al. BCAA catabolism in brown fat controls energy homeostasis through SLC25A44. Nature 2019, 572, 614–619. [Google Scholar] [CrossRef] [PubMed]
  173. Penna, C.; Andreadou, I.; Aragno, M.; Beauloye, C.; Bertrand, L.; Lazou, A.; Falcão-Pires, I.; Bell, R.; Zuurbier, C.J.; Pagliaro, P.; et al. Effect of hyperglycaemia and diabetes on acute myocardial ischaemia-reperfusion injury and cardioprotection by ischaemic conditioning protocols. Br. J. Pharmacol. 2020, 177, 5312–5335. [Google Scholar] [CrossRef]
  174. Andreadou, I.; Daiber, A.; Baxter, G.F.; Brizzi, M.F.; Di Lisa, F.; Kaludercic, N.; Lazou, A.; Varga, Z.V.; Zuurbier, C.J.; Schulz, R.; et al. Influence of cardiometabolic comorbidities on myocardial function, infarction, and cardioprotection: Role of cardiac redox signaling. Free Radic. Biol. Med. 2021, 166, 33–52. [Google Scholar] [CrossRef] [PubMed]
  175. Kamei, Y.; Hatazawa, Y.; Uchitomi, R.; Yoshimura, R.; Miura, S. Regulation of Skeletal Muscle Function by Amino Acids. Nutrients 2020, 12, 261. [Google Scholar] [CrossRef] [PubMed]
  176. Holeček, M. Role of Impaired Glycolysis in Perturbations of Amino Acid Metabolism in Diabetes Mellitus. Int. J. Mol. Sci. 2023, 24, 1724. [Google Scholar] [CrossRef]
  177. Liu, Y.; Li, B.; Li, M.; Yu, Y.; Wang, Z.; Chen, S. Improvement of cardiac dysfunction by bilateral surgical renal denervation in animals with diabetes induced by high fructose and high fat diet. Diabetes Res. Clin. Pract. 2016, 115, 140–149. [Google Scholar] [CrossRef]
  178. Huo, J.-Y.; Jiang, W.-Y.; Zhang, S.-G.; Lyu, Y.-T.; Geng, J.; Chen, M.; Chen, Y.-Y.; Jiang, Z.-X.; Shan, Q.-J. Renal denervation ameliorates cardiac metabolic remodeling in diabetic cardiomyopathy rats by suppressing renal SGLT2 expression. Lab. Investig. 2022, 102, 341–351. [Google Scholar] [CrossRef]
  179. Chiao, Y.A.; Chakraborty, A.D.; Light, C.M.; Tian, R.; Sadoshima, J.; Shi, X.; Gu, H.; Lee, C.F. NAD+ Redox Imbalance in the Heart Exacerbates Diabetic Cardiomyopathy. Circ. Heart Fail. 2021, 14, e008170. [Google Scholar] [CrossRef]
  180. Alves, A.; Lamarche, F.; Lefebvre, R.; Drevet Mulard, E.; Bassot, A.; Chanon, S.; Loizon, E.; Pinteur, C.; Bloise, A.M.N.d.L.G.; Godet, M.; et al. Glycine Supplementation in Obesity Worsens Glucose Intolerance through Enhanced Liver Gluconeogenesis. Nutrients 2022, 15, 96. [Google Scholar] [CrossRef]
  181. Handzlik, M.K.; Gengatharan, J.M.; Frizzi, K.E.; McGregor, G.H.; Martino, C.; Rahman, G.; Gonzalez, A.; Moreno, A.M.; Green, C.R.; Guernsey, L.S.; et al. Insulin-regulated serine and lipid metabolism drive peripheral neuropathy. Nature 2023, 614, 118–124. [Google Scholar] [CrossRef] [PubMed]
  182. Kar, S.; Shahshahan, H.R.; Kambis, T.N.; Yadav, S.K.; Li, Z.; Lefer, D.J.; Mishra, P.K. Hydrogen Sulfide Ameliorates Homocysteine-Induced Cardiac Remodeling and Dysfunction. Front. Physiol. 2019, 10, 598. [Google Scholar] [CrossRef]
  183. Mishra, P.K.; Givvimani, S.; Metreveli, N.; Tyagi, S.C. Attenuation of beta2-adrenergic receptors and homocysteine metabolic enzymes cause diabetic cardiomyopathy. Biochem. Biophys. Res. Commun. 2010, 401, 175–181. [Google Scholar] [CrossRef]
  184. Tao, H.; Shi, P.; Xuan, H.-Y.; Ding, X.-S. DNA methyltransferase-1 inactivation of androgen receptor axis triggers homocysteine induced cardiac fibroblast autophagy in diabetic cardiac fibrosis. Arch. Biochem. Biophys. 2020, 692, 108521. [Google Scholar] [CrossRef]
  185. Guo, X.; Chen, K.; Ji, L.; Wang, S.; Ye, X.; Xu, L.; Feng, L. Ultrasound-targeted microbubble technology facilitates SAHH gene delivery to treat diabetic cardiomyopathy by activating AMPK pathway. iScience 2024, 27, 108852. [Google Scholar] [CrossRef] [PubMed]
  186. Liu, X.; Zheng, Y.; Guasch-Ferré, M.; Ruiz-Canela, M.; Toledo, E.; Clish, C.; Liang, L.; Razquin, C.; Corella, D.; Estruch, R.; et al. High plasma glutamate and low glutamine-to-glutamate ratio are associated with type 2 diabetes: Case-cohort study within the PREDIMED trial. Nutr. Metab. Cardiovasc. Dis. 2019, 29, 1040–1049. [Google Scholar] [CrossRef] [PubMed]
  187. Leandro, J.; Houten, S.M. The lysine degradation pathway: Subcellular compartmentalization and enzyme deficiencies. Mol. Genet. Metab. 2020, 131, 14–22. [Google Scholar] [CrossRef]
  188. Razquin, C.; Ruiz-Canela, M.; Clish, C.B.; Li, J.; Toledo, E.; Dennis, C.; Liang, L.; Salas-Huetos, A.; Pierce, K.A.; Guasch-Ferré, M.; et al. Lysine pathway metabolites and the risk of type 2 diabetes and cardiovascular disease in the PREDIMED study: Results from two case-cohort studies. Cardiovasc. Diabetol. 2019, 18, 151. [Google Scholar] [CrossRef]
  189. Zhou, L.; Sun, C.-B.; Liu, C.; Fan, Y.; Zhu, H.-Y.; Wu, X.-W.; Hu, L.; Li, Q.-P. Upregulation of arginase activity contributes to intracellular ROS production induced by high glucose in H9c2 cells. Int. J. Clin. Exp. Pathol. 2015, 8, 2728–2736. [Google Scholar]
  190. Zhu, Z.-D.; Ye, J.-M.; Fu, X.-M.; Wang, X.-C.; Ye, J.-Y.; Wu, X.-R.; Hua, P.; Liao, Y.-Q.; Xuan, W.; Duan, J.-L.; et al. DDAH2 alleviates myocardial fibrosis in diabetic cardiomyopathy through activation of the DDAH/ADMA/NOS/NO pathway in rats. Int. J. Mol. Med. 2019, 43, 749–760. [Google Scholar] [CrossRef]
  191. Wei, C.; Song, T.; Yuan, H.; Li, X.; Zhang, X.; Liang, X.; Fan, Y. Transcriptomics Coupled to Proteomics Reveals Novel Targets for the Protective Role of Spermine in Diabetic Cardiomyopathy. Oxid. Med. Cell Longev. 2022, 2022, 5909378. [Google Scholar] [CrossRef] [PubMed]
  192. Hu, J.; Lu, X.; Zhang, X.; Shao, X.; Wang, Y.; Chen, J.; Zhao, B.; Li, S.; Xu, C.; Wei, C. Exogenous spermine attenuates myocardial fibrosis in diabetic cardiomyopathy by inhibiting endoplasmic reticulum stress and the canonical Wnt signaling pathway. Cell Biol. Int. 2020, 44, 1660–1670. [Google Scholar] [CrossRef] [PubMed]
  193. Fiordelisi, A.; Cerasuolo, F.A.; Avvisato, R.; Buonaiuto, A.; Maisto, M.; Bianco, A.; D’Argenio, V.; Mone, P.; Perrino, C.; D’Apice, S.; et al. L-Arginine supplementation as mitochondrial therapy in diabetic cardiomyopathy. Cardiovasc. Diabetol. 2024, 23, 450. [Google Scholar] [CrossRef] [PubMed]
  194. Al Zoubi, S.; Chen, J.; Murphy, C.; Martin, L.; Chiazza, F.; Collotta, D.; Yaqoob, M.M.; Collino, M.; Thiemermann, C. Linagliptin Attenuates the Cardiac Dysfunction Associated With Experimental Sepsis in Mice With Pre-existing Type 2 Diabetes by Inhibiting NF-κB. Front. Immunol. 2018, 9, 2996. [Google Scholar] [CrossRef]
  195. Wen, C.; Liu, C.; Li, Y.; Xia, T.; Zhang, X.; Xue, S.; Olatunji, O.J. Ameliorative potentials of the ethanolic extract from Lycium chinense leaf extract against diabetic cardiomyopathy. Insight into oxido-inflammatory and apoptosis modulation. Biomed. Pharmacother. 2022, 154, 113583. [Google Scholar] [CrossRef]
  196. Liza; Hussain, G.; Malik, A.; Akhtar, S.; Anwar, H. Artemisia vulgaris Extract as a Novel Therapeutic Approach for Reversing Diabetic Cardiomyopathy in a Rat Model. Pharmaceuticals 2024, 17, 46. [Google Scholar] [CrossRef]
  197. Asghari, A.A.; Mahmoudabady, M.; Shabab, S.; Niazmand, S. Anti-inflammatory, anti-oxidant and anti-apoptotic effects of olive leaf extract in cardiac tissue of diabetic rats. J. Pharm. Pharmacol. 2022, 74, 961–972. [Google Scholar] [CrossRef]
  198. Abdi, T.; Mahmoudabady, M.; Marzouni, H.Z.; Niazmand, S.; Khazaei, M. Ginger (Zingiber officinale Roscoe) Extract Protects the Heart Against Inflammation and Fibrosis in Diabetic Rats. Can. J. Diabetes 2021, 45, 220–227. [Google Scholar] [CrossRef]
  199. Yao, P.-A.; Wei, K.-Z.; Feng, J.-H.; Liu, X.-N.; Xu, X.; Cui, H.-Y.; Zhang, X.-C.; Gao, J.-P. Sodium houttuyfonate protects against cardiac injury by regulating cardiac energy metabolism in diabetic rats. Eur. J. Pharmacol. 2022, 932, 175236. [Google Scholar] [CrossRef]
  200. Farazandeh, M.; Mahmoudabady, M.; Asghari, A.A.; Niazmand, S. Diabetic cardiomyopathy was attenuated by cinnamon treatment through the inhibition of fibro-inflammatory response and ventricular hypertrophy in diabetic rats. J. Food Biochem. 2022, 46, e14206. [Google Scholar] [CrossRef]
  201. Gur, F.M.; Aktas, I. The ameliorative effects of thymoquinone and beta-aminoisobutyric acid on streptozotocin-induced diabetic cardiomyopathy. Tissue Cell. 2021, 71, 101582. [Google Scholar] [CrossRef]
  202. Shabab, S.; Mahmoudabady, M.; Gholamnezhad, Z.; Fouladi, M.; Asghari, A.A. Diabetic cardiomyopathy in rats was attenuated by endurance exercise through the inhibition of inflammation and apoptosis. Heliyon 2024, 10, e23427. [Google Scholar] [CrossRef]
  203. Wang, S.; Li, M.; Lin, H.; Wang, G.; Xu, Y.; Zhao, X.; Hu, C.; Zhang, Y.; Zheng, R.; Hu, R.; et al. Amino acids, microbiota-related metabolites, and the risk of incident diabetes among normoglycemic Chinese adults: Findings from the 4C study. Cell Rep. Med. 2022, 3, 100727. [Google Scholar] [CrossRef]
  204. Mathew, A.V.; Jaiswal, M.; Ang, L.; Michailidis, G.; Pennathur, S.; Pop-Busui, R. Impaired Amino Acid and TCA Metabolism and Cardiovascular Autonomic Neuropathy Progression in Type 1 Diabetes. Diabetes 2019, 68, 2035–2044. [Google Scholar] [CrossRef] [PubMed]
  205. Wang, H.; Wang, Y.; Li, X.; Deng, X.; Kong, Y.; Wang, W.; Zhou, Y. Machine learning of plasma metabolome identifies biomarker panels for metabolic syndrome: Findings from the China Suboptimal Health Cohort. Cardiovasc. Diabetol. 2022, 21, 288. [Google Scholar] [CrossRef]
  206. Wu, J.; Subbaiah, K.C.V.; Xie, L.H.; Jiang, F.; Khor, E.-S.; Mickelsen, D.; Myers, J.R.; Tang, W.H.W.; Yao, P. Glutamyl-Prolyl-tRNA Synthetase Regulates Proline-Rich Pro-Fibrotic Protein Synthesis During Cardiac Fibrosis. Circ. Res. 2020, 127, 827–846. [Google Scholar] [CrossRef] [PubMed]
  207. Vanweert, F.; Neinast, M.; Tapia, E.E.; van de Weijer, T.; Hoeks, J.; Schrauwen-Hinderling, V.B.; Blair, M.C.; Bornstein, M.R.; Hesselink, M.K.C.; Schrauwen, P.; et al. A randomized placebo-controlled clinical trial for pharmacological activation of BCAA catabolism in patients with type 2 diabetes. Nat. Commun. 2022, 13, 3508. [Google Scholar] [CrossRef] [PubMed]
  208. Kumar, P.; Liu, C.; Hsu, J.W.; Chacko, S.; Minard, C.; Jahoor, F.; Sekhar, R.V. Glycine and N-acetylcysteine (GlyNAC) supplementation in older adults improves glutathione deficiency, oxidative stress, mitochondrial dysfunction, inflammation, insulin resistance, endothelial dysfunction, genotoxicity, muscle strength, and cognition: Results of a pilot clinical trial. Clin. Transl. Med. 2021, 11, e372. [Google Scholar] [CrossRef]
  209. Mansour, A.; Mohajeri-Tehrani, M.R.; Qorbani, M.; Heshmat, R.; Larijani, B.; Hosseini, S. Effect of glutamine supplementation on cardiovascular risk factors in patients with type 2 diabetes. Nutrition 2015, 31, 119–126. [Google Scholar] [CrossRef]
  210. Zhang, Y. The essential role of glutamine metabolism in diabetic cardiomyopathy: A review. Medicine 2023, 102, e36299. [Google Scholar] [CrossRef]
  211. Arslan, A.K.; Yagin, F.H.; Algarni, A.; Karaaslan, E.; Al-Hashem, F.; Ardigò, L.P. Enhancing type 2 diabetes mellitus prediction by integrating metabolomics and tree-based boosting approaches. Front. Endocrinol. 2024, 15, 1444282. [Google Scholar] [CrossRef] [PubMed]
  212. Wamil, M.; Goncalves, M.; Rutherford, A.; Borlotti, A.; Pellikka, P.A. Multi-modality cardiac imaging in the management of diabetic heart disease. Front. Cardiovasc. Med. 2022, 9, 1043711. [Google Scholar] [CrossRef] [PubMed]
  213. Li, J.; Guan, Z.; Wang, J.; Cheung, C.Y.; Zheng, Y.; Lim, L.-L.; Lim, C.C.; Ruamviboonsuk, P.; Raman, R.; Corsino, L.; et al. Integrated image-based deep learning and language models for primary diabetes care. Nat. Med. 2024, 30, 2886–2896. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Pathways of amino acid metabolism. Most amino acids undergo catabolic processes in the liver, primarily through deamination to form α-keto acids and ammonia (NH3/NH4+). The resulting α-keto acids are oxidized in the mitochondria for energy production, converted into glucose and fatty acids, or resynthesized into NEAAs. Ammonia is predominantly detoxified via the ornithine cycle to form urea and ultimately excreted by the kidneys. Moreover, certain amino acids can be decarboxylated to yield carbon dioxide and amines. The arrows in the figure represent either the transport of substances or the course of biochemical processes.
Figure 1. Pathways of amino acid metabolism. Most amino acids undergo catabolic processes in the liver, primarily through deamination to form α-keto acids and ammonia (NH3/NH4+). The resulting α-keto acids are oxidized in the mitochondria for energy production, converted into glucose and fatty acids, or resynthesized into NEAAs. Ammonia is predominantly detoxified via the ornithine cycle to form urea and ultimately excreted by the kidneys. Moreover, certain amino acids can be decarboxylated to yield carbon dioxide and amines. The arrows in the figure represent either the transport of substances or the course of biochemical processes.
Biomolecules 15 00916 g001
Figure 2. Regulation of amino acid metabolic enzymes. Enzymes involved in amino acid metabolism are regulated by a variety of factors. Bioactive compounds in food, such as quercetin and organic acids, can inhibit or activate the activity of different digestive enzymes. Cofactor dysfunction may lead to impaired enzyme activity and disease pathogenesis. In addition, the transcription of enzymes is regulated by factors such as diet, the gut microbiota, hormone levels, and transcription factors. + and − represent the activation and inhibition of digestive enzyme activity, respectively.
Figure 2. Regulation of amino acid metabolic enzymes. Enzymes involved in amino acid metabolism are regulated by a variety of factors. Bioactive compounds in food, such as quercetin and organic acids, can inhibit or activate the activity of different digestive enzymes. Cofactor dysfunction may lead to impaired enzyme activity and disease pathogenesis. In addition, the transcription of enzymes is regulated by factors such as diet, the gut microbiota, hormone levels, and transcription factors. + and − represent the activation and inhibition of digestive enzyme activity, respectively.
Biomolecules 15 00916 g002
Figure 3. Branched-Chain Amino Acids Metabolism in DCM. Under physiological conditions, BCAAs undergo catabolism via a series of enzymatic reactions in the mitochondria of skeletal muscle, myocardium, and hepatic tissues. In diabetes, the interplay of impaired glucose/lipid metabolism, vagal dysfunction, and gut microbiota disruption alters the status of substrates and enzymes within the myocardial BCAA metabolic pathway, which contributes to the pathogenesis of DCM. Mechanistically, autophagy dysregulation, oxidative stress, and inflammatory responses collectively drive myocardial hypertrophy and fibrosis, ultimately manifesting as impaired cardiac function. An upward arrow indicates an increase in a substance or the promotion of a biological process, while a downward arrow indicates a decrease in a substance or the inhibition of a biological process.
Figure 3. Branched-Chain Amino Acids Metabolism in DCM. Under physiological conditions, BCAAs undergo catabolism via a series of enzymatic reactions in the mitochondria of skeletal muscle, myocardium, and hepatic tissues. In diabetes, the interplay of impaired glucose/lipid metabolism, vagal dysfunction, and gut microbiota disruption alters the status of substrates and enzymes within the myocardial BCAA metabolic pathway, which contributes to the pathogenesis of DCM. Mechanistically, autophagy dysregulation, oxidative stress, and inflammatory responses collectively drive myocardial hypertrophy and fibrosis, ultimately manifesting as impaired cardiac function. An upward arrow indicates an increase in a substance or the promotion of a biological process, while a downward arrow indicates a decrease in a substance or the inhibition of a biological process.
Biomolecules 15 00916 g003
Table 1. Classification, derivatives, and main degrading enzymes of amino acids.
Table 1. Classification, derivatives, and main degrading enzymes of amino acids.
Name
(Abbreviation, Symbol)
Nutritional
Classification
Metabolic
Transformation
Important Nitrogenous DerivativeMain AADEs
(Abbreviation)
Alanine (Ala, A)NEAAGlucogenic-Glutamic pyruvate transaminase (GPT)
Cysteine (Cys, C)NEAAGlucogenicTaurineCysteine dioxygenase 1 (CDO1)
Aspartic acid (Asp, D)NEAAGlucogenicPurine base, pyrimidine baseGlutamic-oxaloacetic transaminase 1 (GOT1)
Glutamic acid (Glu, E)NEAAGlucogenicGABAGlutamate dehydrogenase 1 (GLUD1)
Phenylalanine (Phe, F)EAAGlucogenic and ketogenicCA, thyroxine, melaninPhenylalanine hydroxylase (PAH)
Glycine (Gly, G)NEAAGlucogenicPurine base, porphyrin,
creatine, creatine phosphate
Serine hydroxymethyltransferase 1 (SHMT1)
Histidine (His, H)EAAGlucogenicHistamineHistidine ammonia-lyase (HAL)
Isoleucine (Ile, I)EAAGlucogenic and ketogenic-Branched-chain amino acid transaminase (BCATc, BCATm)
Branched-chain keto acid dehydrogenase (BCKDH)
Lysine (Lys, K)EAAKetogenicCrotonyl-CoAGlutaryl-CoA dehydrogenase (GCDH)
Leucine (Leu, L)EAAKetogenic-see Isoleucine
Methionine (Met, M)EAAGlucogenicSpermidine, spermine,
creatine, creatine phosphate
Methionine adenosyl transferase 1A (MAT1A)
Asparagine (Asn, N)NEAAGlucogenic-Asparaginase (ASPG)
Proline (Pro, P)NEAAGlucogenic-Proline dehydrogenase 1 (PRODH1)
Glutamine (Gln, Q)NEAAGlucogenicPurine baseGlutaminase 1 (GLS1)
Glutaminase 2 (GLS2)
Arginine (Arg, R)NEAAGlucogenicNO,
Creatine, creatine phosphate
Arginase 1 (ARG1)
Serine (Ser, S)NEAAGlucogenic-Serine dehydratase (SDS)
Threonine (Thr, T)EAAGlucogenic and ketogenic-Serine dehydratase like (SDSL)
Valine (Val, V)EAAGlucogenic-see Isoleucine
Tryptophan (Trp, W)EAAGlucogenic and ketogenic5-HT, nicotinic acidTryptophan 2,3-dioxygenase (TDO2)
Aminocarboxymuconate semialdehyde decarboxylase (ACMSD)
Tryptophan hydroxylase 1 (TPH1)
Tyrosine (Tyr, Y)NEAAGlucogenic and ketogenicCA, thyroxine, melaninTyrosinase (TYR)
Tyrosine hydroxylase (TH)
Tyrosine aminotransferase (TAT)
Table 2. Preclinical research on amino acid metabolism in diabetic cardiomyopathy.
Table 2. Preclinical research on amino acid metabolism in diabetic cardiomyopathy.
TargetDrugModelMain FindingsRefs
BCAT2, PP2CmGlucosyringic acid (GA)Male C57BL/6 J mice induced by HFD and SFZGA restored normal BCAA metabolism in diabetic mouse heart via targeting and inhibiting the periostin/NAP1L2/SIRT3 axis[111]
BCAT2, PP2Cm, BCKDK, BCKDHPyridostigmineMale C57BL/6 J mice induced by HFD and SFZPyridostigmine improved disrupted BCAA metabolic enzymes and intestinal microbiota homeostasis, and enhanced vagal nerve activity[112]
PP2Cm, BCKDK, BCKDHPortulaca oleracea
L. extracts (PE)
Mice induced by HFD and STZPE improved disrupted BCAA metabolic enzymes and intestinal microbiota homeostasis[113]
BCKDK3,6-dichlorobenzo[b]
thiophene-2-carboxylic acid (BT2)
Male ob/ob mice, wild-type C57BL/6 J mice, leptin gene mutant ob/ob mice and BCKDK-Alb cre+ mice, induced by HFDBT2 inhibited BCKDK, reduced the level of p-BCKDH and thus enhanced BCAA catabolism, and potentiated the hypoglycemic effect of metformin[114]
BCKDKBT2Male C57BL/6 N mice performed with TACBT2 enhanced BCAA catabolism, and improved TAC-induced cardiac contractile dysfunction and pathological state[115]
BCKDKBT2C57BL/6 mice induced by HFD and SFZBT2 enhanced BCAA catabolism, ameliorated the impaired heart function and reduced the infarction area in diabetic mice with myocardial infarction[116]
BCKDKBT23-MST KO mice and C57BL/6 J mice performed with TACBT2 enhanced BCAA catabolism and ameliorated the severity of TAC-induced heart failure in 3-MST KO mice[117]
The oxidative respiration process within the mitochondriaBT2NRVMs and iPSC-derived cardiomyocytesIndependent of the inhibitory effect on BCKDK, BT2 decreased mitochondrial membrane potential, increased proton conductance across the mitochondrial inner membrane, and reduced the production of ROS[118]
IRS1/Akt signaling pathwaySodium Phenylbutyrate (NaPB)Murine C2C12 myoblasts, passage 6–8, treated with elevated (4×) media BCAA concentrationsUnder high BCAA conditions, NaPB treatment elevated Akt and AS160 phosphorylation, while decreased glycogen synthesis and BCAA concentrations[119]
PP2Cm and mTOR/p-ULK1 signaling pathwayEmpagliflozinMale KK-Ay mice aged 8 weeks induced by HFDEmpagliflozin promoted BCAA degradation through the upregulation of PP2Cm and inhibited mTOR/p-ULK1 to enhance autophagy[120]
BCAA/BCKA metabolism in BATTirzepatideMale C57BL/6 J mice induced by HFDTirzepatide stimulated catabolism of BCAAs/BCKAs in BAT, as demonstrated by increased BCAA/BCKA-derived metabolites[121]
PAH in liversTetrahydrobiopterinGlobal p21−/− mice backcrossed to C57BL/6 J background for at least 10 generationsIn naturally aged mice, consistent with siRNA-mediated p21 knockdown, tetrahydrobiopterin treatment restored healthy cardiac structure and function through reviving hepatic PAH activity and normalizing plasma phenylalanine levels[11]
Gut microbiotaRice wine polyphenols and polypeptides within Chinese rice wineMale db/db and db/m miceFunctional components of Chinese rice wine provided a cardioprotective effect against DCM via increasing tryptophan metabolism-associated metabolites and reducing serum phenylalanine by modulating the composition and metabolic function of the gut microbiota[122]
Gut microbiotaEthanol extract of S. fusiforme (EE)Male ICR mice aged 8 weeks induced by HFD and SFZEE altered the composition of gut microbiota, reduced the levels of BCAAs and AAAs, and improved glucose tolerance as well as pathological changes in the heart[123]
mTORC1-v-ATPase axis, adaptor protein Ragulator, and SLC38A9Specific cocktail of amino acids (lysine/leucine/arginine)Male Lewis rats induced by HFD, and cardiomyocyte models (aRCMs, HL-1 and hiPSC-CM)Lysine/leucine/arginine stimulated mTORC1-v-ATPase axis, reinternalized CD36, and reduced cardiac lipid uptake[124]
Myocardial NE and THLiraglutide and dapagliflozinMale SD rats induced by HFD and SFZBoth liraglutide and dapagliflozin significantly reduced TH density and myocardial NE contents, and dapagliflozin exhibited more reduction than liraglutide[125]
Myocardial THStevia Rebaudiana (R) extractsMale SD rats induced by HFD and SFZStevia R extracts significantly attenuated myocardial TH density[126]
Myocardial NE and THRAAS blockers (enalapril and losartan)Male SD rats induced by HFD and SFZBlockade of RAAS attenuated myocardial TH density and NE contents[127]
5-HT and its cardiac receptor (5-HT2B receptor)L. plantarum and inulinMale Wistar rats induced by HFD and SFZIncrease in intestinal and serum 5-HT as well as decrease in cardiac 5-HT and 5-HT2B receptor were observed in diabetic rats, which were reversed by L. plantarum and insulin administration[128]
Amino acid metabolism and AMPK and PI3K/Akt/FoxO3a signaling pathways in the heart tissueErzhi PillMale SD rats induced by HFD and SFZErzhi Pill balanced amino acid metabolism similar to glutamic acid and glycine, and regulated the AMPK and PI3K/Akt/FoxO3a signaling pathways[129]
SAMe and DNMT-SOCS1/3-IGF-1 signalingVitamin B12Male C57BL/6 J mice carrying Elmo1H/H and Ins2Akita/+ genesHigh oral dose of vitamin B12 normalized the decreased levels of SAMe and DNMTs, modulated oxidative stress, and improved the echocardiographic indices[130]
Hcy, etc.Ginger extractMale Wistar rats induced by SFZGinger extract restored the increased levels of Hcy and alleviated heart structural abnormalities[131]
Cardiac CSE and H2S as well as insulin receptor and Akt/GSK-3β signalingS-Propargyl-Cysteine (SPRC)Male C57BLKS/J db/db miceSPRC increased CSE expression and H2S content, activated cardiac insulin receptor and Akt/GSK-3β signaling[132]
Synoviolin-1 (SYVN1/Hrd1)NaHS and the novel hydrogen sulfide-releasing molecule GYY4137Female db/db mice and HL-1 cells treated with palmitate and oleateExogenous H2S improved H2S levels in cardiomyocytes, prevented LDs formation by restoring SYVN1 S sulfhydration and promoting SREBP1 ubiquitination[133]
USP8/parkin signaling pathwayNaHSMale and female db/db miceExogenous H2S activated USP8 S sulfhydration, promoted parkin-dependent mitophagy and ameliorated cardiac impairment[134]
PI3K/Akt pathwayExogenous SO2 donor (Na2SO3/NaHSO3)Male SD rats induced by high-fat high-sucrose diet (HFHSD) and SFZSO2 activated autophagy to antagonize cardiomyocyte apoptosis and fibrosis by downregulating the excessive activation of PI3K/Akt pathway[135]
Nrf2/HO-1 and NF-κB pathwaysPiceatannol (PIC)Male SD rats induced by SFZ, and HG-induced H9C2 cardiac myoblastsPIC suppressed HG-induced NF-κB activation by upregulating Nrf2 and HO-1 expression, and alleviated inflammation and oxidative stress in DCM rats[136]
Nrf2/ARE signaling pathwayEmpagliflozinMale db/db miceEmpagliflozin inhibited oxidative stress via activating Nrf2/ARE signaling, modulated ketone body metabolism, and improved mitochondrial dysfunction in DCM[137]
Nrf2/ARE signaling and TGF-β/Smad pathwayEmpagliflozinKK-Ay mice induced by HFDEmpagliflozin attenuated oxidative stress and fibrosis in diabetic heart by activating Nrf2/ARE and suppressing TGF-β/Smad signaling[138]
JNK/p38 MAPK and NF-κB pathwaysLCZ696 (an ARNI)Male C57BL/6 mice induced by SFZ, and HG-induced H9C2 cardiomyocytesLCZ696 inhibited inflammation and oxidative stress by suppressing JNK/p38 MAPK phosphorylation and NF-κB nuclear translocation[139]
CaMKII/NF-κB/TGF-β1 and PPAR-γ signaling pathwaysPioglitazone and curcumin (Pio/Cur)Male adult SD rats induced by SFZPio/Cur treatment ameliorated DCM in T1DM via inhibition of CaMKII/NF-κB/TGF-β1 and activation of PPAR-γ pathways[140]
AMPK/Nrf2 pathwaysSulforaphane (SFN)Engineered cardiac tissue and AMPKα2-KO mice induced by HFD and SFZSFN prevented ferroptosis and associated cardiac pathogenesis via AMPK-mediated Nrf2 activation[141]
RAGE, OGT, and GFAT and NF-κB in heart tissueVitamin DMale SD rats induced by SFZVitamin D alleviated DCM by down-regulating the RAGE expression and HBP-mediated O-glycosylation, while reducing NF-κB activity[142]
BH4/eNOS/NO pathwaySepiapterin (SEP) and L-citrulline (L-Cit)db/db mice and HG-induced ECs stimulating I/R or H/R conditionsCoadministration of SEP and L-Cit protected diabetic heart, via improvements in coronary arterial endothelial function, cardiac BH4 concentrations, and eNOS function[143]
Nrf2-ROS-p53-MuRF1 axisSpermineMale Wistar rats induced by SFZExogenous spermine attenuated DCM by suppressing ROS-p53 mediated downregulation of cell membrane calcium-sensitive receptor[144]
Wnt/β-catenin signalingSpermineMale Wistar rats induced by SFZ, and HG-induced CFs from neonatal Wistar ratsExogenous spermine attenuated myocardial fibrosis by inhibiting ERS and the canonical Wnt/β-catenin signaling pathway[145]
Nrf2 signalingL-ArginineNeonatal rat cardiomyocytes H9c2 (2-1) cell line, incubated with MGO to stimulate glycationL-Arginine exerted protective effects in DCM due to the inhibition of HSA glycation as well as the activation and nuclear translocation of Nrf2[146]
NF-κβ pathwayβ-caryophyllene (BCP) and L-Arginine (LA)Male SD rats induced by SFZCoadministration of BCP and LA led to a reduction in collagen deposition and cardiac fibrosis via NF-ĸβ inhibition[147]
PI3K/Akt/Nrf2 pathwaySpiraeosideHG-induced AC16 cellsSpiraeoside protected HG-stimulated cardiomyocytes through its antioxidant and antiapoptotic activities via the activation of PI3K/Akt/Nrf2 pathway[148]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wen, Y.; Ma, X.; Mei, S.; Wuyun, Q.; Yan, J. Emerging Insights into the Relationship Between Amino Acid Metabolism and Diabetic Cardiomyopathy. Biomolecules 2025, 15, 916. https://doi.org/10.3390/biom15070916

AMA Style

Wen Y, Ma X, Mei S, Wuyun Q, Yan J. Emerging Insights into the Relationship Between Amino Acid Metabolism and Diabetic Cardiomyopathy. Biomolecules. 2025; 15(7):916. https://doi.org/10.3390/biom15070916

Chicago/Turabian Style

Wen, Yi, Xiaozhu Ma, Shuai Mei, Qidamugai Wuyun, and Jiangtao Yan. 2025. "Emerging Insights into the Relationship Between Amino Acid Metabolism and Diabetic Cardiomyopathy" Biomolecules 15, no. 7: 916. https://doi.org/10.3390/biom15070916

APA Style

Wen, Y., Ma, X., Mei, S., Wuyun, Q., & Yan, J. (2025). Emerging Insights into the Relationship Between Amino Acid Metabolism and Diabetic Cardiomyopathy. Biomolecules, 15(7), 916. https://doi.org/10.3390/biom15070916

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop