Skip to main content
The American Journal of Pathology logoLink to The American Journal of Pathology
. 2024 Apr;194(4):510–524. doi: 10.1016/j.ajpath.2023.12.006

Amino Acid Metabolism and Atherosclerotic Cardiovascular Disease

Sumit Kumar Anand , Theresea-Anne Governale , Xiangyu Zhang , Babak Razani , Arif Yurdagul Jr ∗,†,§, Christopher B Pattillo †,§,, Oren Rom ∗,†,§,∗∗
PMCID: PMC10988767  PMID: 38171450

Abstract

Despite significant advances in medical treatments and drug development, atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of death worldwide. Dysregulated lipid metabolism is a well-established driver of ASCVD. Unfortunately, even with potent lipid-lowering therapies, ASCVD-related deaths have continued to increase over the past decade, highlighting an incomplete understanding of the underlying risk factors and mechanisms of ASCVD. Accumulating evidence over the past decades indicates a correlation between amino acids and disease state. This review explores the emerging role of amino acid metabolism in ASCVD, uncovering novel potential biomarkers, causative factors, and therapeutic targets. Specifically, the significance of arginine and its related metabolites, homoarginine and polyamines, branched-chain amino acids, glycine, and aromatic amino acids, in ASCVD are discussed. These amino acids and their metabolites have been implicated in various processes characteristic of ASCVD, including impaired lipid metabolism, endothelial dysfunction, increased inflammatory response, and necrotic core development. Understanding the complex interplay between dysregulated amino acid metabolism and ASCVD provides new insights that may lead to the development of novel diagnostic and therapeutic approaches. Although further research is needed to uncover the precise mechanisms involved, it is evident that amino acid metabolism plays a role in ASCVD.


Atherosclerosis, a chronic disease of the arteries driven by impaired lipid metabolism and an imbalanced inflammatory response, is the major cause of cardiovascular diseases (CVDs), including myocardial infarction and stroke.1 The deposition of lipoproteins containing apolipoprotein B in the subendothelial regions of the intima layer in the arterial wall causes a persistent proinflammatory response that drives atherosclerotic plaque formation.2,3 As the disease progresses, atherosclerotic plaques accumulate extracellular matrix proteins and calcium minerals, whereas necrotic cores develop as lipoprotein-rich macrophages that accumulate and die.3 Advanced atherosclerotic plaques can cause ischemia by occluding the artery lumen and impeding blood flow; however, most cardiovascular events are caused by occlusive thrombi from ruptured unstable plaques.4,5 Despite advances in medical treatments and pharmacologic therapies, atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of death worldwide. Alarmingly, a significant increase in deaths due to CVD has been observed during the past decade,6 highlighting the incomplete understanding of underlying risk factors and pathophysiological mechanisms that drive ASCVD.

Dysregulated cholesterol and lipid metabolism plays a central role in the pathogenesis of ASCVD and has been extensively studied for over a century.7,8 More recently, mounting evidence has suggested a role for impaired amino acid metabolism. Beyond their obvious role as building blocks of proteins, amino acids play a role in cardiovascular health.9 The first analysis of amino acid composition in human atherosclerotic plaques was published in 1965 (Figure 1).10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21

Figure 1.

Figure 1

Landmark findings in the field of amino acids and their metabolism in atherosclerotic cardiovascular disease over the past decades. BCAA, branched-chain amino acid; CAD, coronary artery disease; CHD, coronary heart disease; GWAS, genome-wide association study; IDO, indoleamine 2,3-dioxygenase.

Another landmark study, published in 1997, identified the amino acid metabolite, homocysteine, as a predictor of mortality in patients with coronary artery disease (CAD),13 and sparked interest in the role of amino acid metabolism in atherosclerosis. Today, there is growing interest in amino acid profile and metabolism, not only as potential risk factors and biomarkers for ASCVD, but also as driving factors and interventional targets.

The current review discusses the emerging role of amino acids and the effect of their metabolism on key characteristics of the atherosclerotic plaque, including size, inflammation, and necrotic cores. It focuses on earlier studies on arginine as well as later studies exploring the arginine-related metabolites, homoarginine and polyamines. In addition, the accumulating evidence of branched-chain amino acids (BCAAs), glycine, aromatic amino acids (AAAs), their related metabolites, as well as selenocysteine in ASCVD is discussed.

Arginine and Related Metabolites

The role of arginine metabolism in atherosclerosis has been extensively studied over the past several decades.12,22 Arginine influences physiological signaling through nitric oxide (NO) synthase, which catalyzes the conversion of arginine to citrulline, generating NO as a by-product.23 Metabolism of arginine through this pathway, mediated by endothelial NO synthase, triggers vasodilation,24 prevents platelet aggregation,25 lowers leukocyte adhesion,26 and maintains endothelial cell quiescence.27 Studies have also identified a correlation between increased arginase activity, which synthesizes ornithine from arginine, and diminished levels of NO, establishing an alternate pathway of arginine metabolism that is part of a phenotypic switch between proinflammatory and proresolving macrophages. NO has been extensively reviewed in the literature, therefore this review focuses on the arginine-related metabolites, homoarginine and polyamines.

Synthesized from arginine and lysine by the mitochondrial enzyme arginine:glycine amidinotransferase, homoarginine has emerged as a potential biomarker of CVD, where lower circulating levels predict adverse cardiovascular events and mortality28, 29, 30 (Table 1).14,17,21,28,31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42 Beyond its potential as a biomarker, experimental evidence suggests that homoarginine plays a protective role in CVD and atherosclerosis.30 Homoarginine supplementation to apolipoprotein E–null (Apoe−/−) mice lowered atherosclerosis and the content of lesional CD3+ T cells by inhibiting T-cell proliferation and their migratory capacity (Table 2).15,19,21,43, 44, 45, 46, 47, 48, 49

Table 1.

Association of Circulatory Levels of Amino Acids with CVD and Measured Risk Factors in Humans

Amino acid/related metabolite Blood levels CVD-related parameters
Homoarginine28 Decrease
  • Incidences of myocardial infarction, heart failure, and mortality

BCAAs14,31, 32, 33 Increase
  • Prevalence of myocardial infarction

  • cIMT

  • hsCRP

Glycine17,21,34, 35, 36, 37 Decrease
  • LDL-C, ApoB, obesity, hypertension, and diabetes

  • Atherosclerosis assessed by CCTA

  • Acute myocardial infarction and stenosis

  • Genetic and observational associations with the risk of coronary heart diseases

Tryptophan38 Decrease
  • Hypertension

3-HKYN39 (tryptophan metabolite) Increase
  • hsCRP

  • Prevalence of acute myocardial infarction

Tyrosine40,41 Increase
  • cIMT

  • Hypertension

  • hsCRP

  • Ischemic stroke

Phenylalanine41,42 Increase
  • Systolic blood pressure, LDL-C, triglycerides, and hsCRP

  • Occurrence of myocardial infarction

  • Ischemic stroke

  • Unstable angina

3-HKYN, kynurenine and 3-hydroxykynurenine; ApoB, apolipoprotein B; BCAA, branched-chain amino acid; CCTA, coronary computed tomography angiography; cIMT, carotid intima-media thickness; CVD, cardiovascular disease; hsCRP, high-sensitivity C-reactive protein; LDL-C, low-density lipoprotein cholesterol.

Table 2.

Plaque Characteristics following Amino Acid Treatment in Rodent Models of Atherosclerosis

Treatment Circulating factors Plaque size Necrotic core Plaque features
Homoarginine43 Decrease:
IFN-γ
IL-12p70
Decrease NA Decrease:
CD3c
CD4
Putrescine19,44 NA Decrease Decrease Increase:
IL-10
MerTK
Spermidine45 NA No change Decrease Decrease:
oil red O
Leucine46 Increase:
HDL-C
Decrease:
LDL-C
MCP-1
Decrease NA NA
BCAAs47 Decrease:
MCP-1
TNF-α
IL-1β
IL-6
TC
HDL-C
LDL-C
Decrease NA Decrease:
F4/80
ICAM-1
VCAM-1
BCAAs48 Increase:
TNF-α
IL-1β
Increase NA Increase:
F4/80
iNOS
Glycine21 Decrease:
TC
TG
Decrease NA NA
3,4-DAA15 (tryptophan metabolite) Decrease:
IL-6
TNF-α
GMCSF
IL-10
CXCL1
Decrease NA Decrease:
CD68
3-HAA49 (tryptophan metabolite) Decrease:
TG
Cholesterol
Decrease NA Decrease:
CD4
No change:
CD68
VCAM-1

3,4-DAA, 3,4-dimethoxycinnamoyl anthranilic acid; 3-HAA, 3-hydroxyanthranilic acid; BCAA, branched-chain amino acid; GMCSF, granulocyte-macrophage colony-stimulating factor; HDL-C, high-density lipoprotein cholesterol; ICAM-1, intercellular adhesion molecule-1; IFN-γ, interferon-γ; iNOS, inducible nitric oxide synthase; LDL-C, low-density lipoprotein cholesterol; MCP-1, monocyte chemoattractant protein-1; MerTK, MER proto-oncogene tyrosine kinase; NA, not applicable; TC, total cholesterol; TG, triglycerides; TNF-α, tumor necrosis factor-α; VCAM-1, vascular cell adhesion molecule-1.

Arginine metabolism into polyamines is a more recent field of study in the context of ASCVD. Polyamines are a family of small, linear polycations composed of putrescine, spermidine, and spermine. The polyamine pathway is initiated by the conversion of arginine into ornithine via arginase 1, which is then decarboxylated by ornithine decarboxylase 1 to produce putrescine.50 The conversion of putrescine into spermidine and spermine follows linear reactions performed by spermidine synthase and spermine synthase, respectively. These polyamines play pivotal roles in numerous molecular and cellular signaling events, including proliferation, gene transcription, mRNA stability, and protein translation, some of which are aberrant in the pathophysiology of ASCVD.

Interestingly, proinflammatory and proresolving macrophages (both present in the progression and regression of atherosclerosis, respectively) are defined by their expression of arginine-metabolizing enzymes. In proinflammatory macrophages, arginine is used to produce NO, whereas in proresolving macrophages, it is converted into ornithine. Notably, the conversion of arginine into polyamines, facilitated by arginase 1 and ornithine decarboxylase 1, is necessary for the clearance of dead cells and the secretion of the proresolving mediator IL-10 by macrophages after stroke and during atherosclerosis regression, respectively19,44,51 (Table 2).

Among polyamines, spermidine plays a significant role in scavenging reactive oxygen species, thereby reducing oxidative stress.52,53 Enhanced spermidine bioavailability is also associated with sustained autophagy,54,55 the impairment of which drives atherosclerosis progression.56, 57, 58, 59 In addition to its antioxidant effects, spermidine supplementation reduces necrotic core formation in Apoe−/− mice by promoting cholesterol efflux in a smooth muscle cell autophagy related 7 (Atg7)-dependent manner45 (Table 2), and suppresses inflammation by reducing circulating tumor necrosis factor-α levels, which mitigate leukocyte recruitment.60, 61, 62 Evidence from a prospective community-based cohort study of 829 individuals demonstrated an association between dietary spermidine intake and reduced mortality63; this finding was supported by data from the National Health and Nutrition Examination Survey, which surveyed individuals from 2003 to 2014 and revealed a strong correlation between dietary spermidine intake and decreased mortality rates, both overall and specific to CVD.64 This study had several notable strengths, including the identification of specific dietary sources of spermidine and the inclusion of a large and diverse population consisting of 10,942 men and 12,952 women.

A diverse range of clinical and epidemiologic studies have highlighted the complex interplay between arginine metabolism, dietary supplementation, and cardiovascular outcomes. The impact of arginine-mediated NO synthesis on endothelial function,65 along with the role of arginase in polyamine production and its implications in cellular functions and ASCVD, underlines the multifaceted nature of arginine metabolism. Nevertheless, clinical trials, such as Vascular Interaction With Age in Myocardial Infarction (VINTAGE MI) (L-arginine therapy), demonstrated that arginine supplementation after infarction provided no beneficial effects on vascular stiffness or ejection fraction,66 whereas the findings from the National Health and Nutrition Examination Survey appear to suggest a protective role following spermidine supplementation. Understanding these seemingly contradictory findings requires an understanding of the patient populations participating in the studies. VINTAGE MI studies following establishment of CVD, and the National Health and Nutrition Examination Survey study, focused on dietary habits over time have advanced the field further. However, it is clear the field needs a better molecular understanding of amino acid function to develop more effective clinical therapies.

Branched-Chain Amino Acids

BCAAs include leucine, isoleucine, and valine, all of which are essential amino acids occurring at a high frequency in proteins.67 Beyond their roles in protein structure and function, BCAAs also regulate metabolic health through their catabolism and regulation of multiple signaling pathways.31,68, 69, 70 In this regard, perturbations in BCAA levels have significant associations with atherosclerosis and related cardiometabolic diseases, which are discussed in this section.

There has been a long history of clinical studies demonstrating a positive correlation between BCAAs and atherosclerosis progression as well as risk factors, such as insulin resistance, diabetes, and obesity.14 More specifically, surrogate markers of atherosclerosis and vascular dysfunction, including carotid intima-media thickness, are directly correlated with circulating BCAA levels (Table 1).14,32,33,71,72 The increase in BCAAs likely results from a combination of both lower BCAA uptake by tissues along with reduced activity of the branched-chain keto α-acid dehydrogenase complex, which accounts for the bulk of BCAA degradation, an observation that has been supported by studies in animal models.73, 74, 75 Furthermore, transcriptomic analysis of plaques at various stages of progression, early (classified as intimal thickening) and advanced (classified as thin or thick fibrous cap atheromas), shows an enrichment of gene networks involved in leucine/isoleucine degradation in early-stage plaques compared with advanced-stage lesions,76 suggesting that the accumulation of BCAAs occurs in both the plasma and at the tissue level.

Although the preponderance of evidence demonstrates a direct relationship between BCAAs and CVD, there are notable studies that reveal a negative association. For instance, a cross-sectional study of women found the intake of leucine was associated with lower arterial stiffness.77 In other studies, supplementation of leucine in mouse models led to improved hepatic lipid metabolism and reduced atherosclerotic lesion size, plaque lipid burden, and monocyte chemoattractant protein-1–mediated macrophage infiltration, whereas body weight, food intake, and blood glucose levels remained unchanged.46 In a second study, supplementing all the BCAAs confirmed these findings,47 indicating a protective role in atherosclerosis that can be attributed to supplementation BCAA46,47 (Table 2). Unfortunately, the leucine supplementation study did not measure plasma levels, making any correlation with human studies and circulating BCAAs difficult. However, the second study showed a trend for increasing levels of valine, leucine, and isoleucine that was significant when combined. Both of these studies suggest a protective role of BCAA supplementation during the initiation and progression of atherosclerosis, which raises the question whether commonly observed elevations in circulating BCAA levels in CVD are simply a biomarker for metabolic dysfunction rather than a promoter of disease. However, supplementation of BCAAs following established atherosclerosis in a murine model has not been performed, making direct comparisons between mice and patients difficult. Currently, the mechanisms of BCAA action are unknown; however, several studies have attempted to address this paucity of information.

Accumulating data from human and animal studies have led to several hypotheses being proposed to explain the effects of BCAAs on cardiovascular risk. A prominent concept is the ability of BCAAs to impair insulin signaling, resulting in systemic insulin resistance, which plays a major role in the development of atherosclerosis.78, 79, 80 As direct activators of mammalian target of rapamycin (mTOR) signaling [particularly mTOR complex (mTORC) 2], BCAAs enhance phosphorylation of insulin receptor substrate-1 on inhibitory sites, leading to blunted phosphatidylinositol-3-kinase activation and downstream insulin signaling.81, 82, 83 Although the significance of mTOR has been challenged in other studies,84,85 the overall inhibitory role of BCAAs in insulin signaling is a recurring observation.86 Beyond the regulation of insulin signaling through the mTOR–insulin receptor substrate-1–phosphatidylinositol-3-kinase axis, BCAAs also appear to cause insulin resistance through their catabolic metabolites. For example, the valine metabolite, 3-hydroxyisobutyrate, can activate transendothelial fatty acid transport, causing local tissue lipid accumulation and lipotoxicity, with resultant compromise in insulin signaling.87 In addition, certain BCAAs and their metabolites inhibit mitochondrial metabolic machinery and function in pancreatic islet β cells, contributing to insulin resistance.87, 88, 89 Furthermore, BCAAs lead to selective disruption of mitochondrial pyruvate use through inhibition of pyruvate dehydrogenase complex and decreased glucose oxidation.89 Finally, the role of short-chain acylcarnitines, which are by-products of BCAA metabolism, has a positive correlation with CAD; however, the mechanism remains unknown.72

There have been recent reports defining pro-atherogenic roles for BCAAs directly at the level of the atherosclerotic plaque. In mouse studies using atherogenic Apoe−/− mouse models, Zhang et al18 found that BCAAs, particularly leucine, derived from diets high in protein, are potent activators of mTORC1 signaling in plaque macrophages. Hyperactive mTORC1 leads to inhibition of mitophagy, accumulation of dysfunctional mitochondria, and acceleration of apoptosis and plaque complexity.18 The essential role for mTOR is demonstrated by the observation that the atherogenic effect of high-protein diets is completely abrogated in the absence of macrophage mTOR signaling or autophagy, supporting a specific BCAA-mTORC1-autophagy axis.18 In human clinical studies on peripheral blood mononuclear cells, elevated levels of BCAAs led to increased production of reactive oxygen species via both NADPH oxidase and the mitochondria in an mTOR-dependent manner,90 indicating that this mechanism may be clinically relevant to the link between BCAAs and cardiovascular risk.

A discussion of dietary protein and BCAAs in CVD would not be complete without an evaluation of gut microbiota. Colonization of germ-free mice with gut microbial communities from lean mice increases expression of genes involved in BCAA catabolism with reduced circulating BCAA levels, whereas the reverse happens on colonization with gut microbiota from obese mice.91 Similar phenotypes are detected in mice transplanted with microbiota from lean versus obese human participants.92 In addition, Qiao et al93 identified the gut symbiont, Parabacteroides merdae, as the main effector of atheroprotection in the Apoe−/− mouse model. In particular, enhanced BCAA catabolism by P. merdae promoted the conversion of BCAAs into branched short-chain fatty acids and inactivation of mTORC1 signaling in plaques.93 It is interesting that mTOR signaling is implicated in both metabolic dysfunction and atherosclerosis, albeit by completely different mechanisms.

Although BCAA supplementation has recently been reported to worsen established atherosclerosis in Apoe−/− mice,48 protective roles for BCAAs in atherosclerotic progression have also been reported in the context of macrophage lipid handling and foam cell formation.46,94,95 Studies from Rom et al96 and Grajeda-Iglesias et al97 indicate that leucine decreases macrophage cholesterol and triglyceride content in a dose-dependent manner by attenuating uptake of triglyceride-rich very low-density lipoprotein and inhibiting triglyceride biosynthesis. In addition, peritoneal macrophages isolated from mice supplemented with leucine show decreased cholesterol content because of reduced cholesterol biosynthesis and enhanced cholesterol efflux.97 Similar observations were made in cultured macrophages treated with serum from healthy individuals supplemented with leucine.97 Thus, it is clear that the roles of BCAAs on the immune system and atherosclerosis are complex and context dependent.

Thus far, much of the literature has considered BCAAs in unison when evaluating roles in CVD. However, the distinct roles of leucine, isoleucine, and valine have been largely neglected. Distinct metabolic roles for each of the BCAAs were shown in a recent study of Yu et al,98 who found that a low isoleucine diet provided several metabolic benefits in mice, including improved hepatic insulin sensitivity, elevated ketogenesis, and increased energy expenditure, which may be mediated by the activation of the fibroblast growth factor 21 uncoupling protein 1 axis. In contrast, reducing dietary valine intake showed modest metabolic effects in mice, and a low leucine diet showed no benefits. Such observations imply that the distinctive effects of BCAAs on ASCVD among studies may be due to the BCAA species and their specific roles. Therefore, when considering the use of BCAAs for diagnosis and therapeutics, the exact amount and function of specific BCAAs needs to be taken into consideration.

Glycine

Glycine is a nonessential and the simplest amino acid, which is synthesized from several precursors via interorgan metabolism involving mainly the liver and kidneys.99 These reactions are catalyzed by key enzymes responsible for glycine formation from serine [serine hydroxymethyltransferase (SHMT) 1 and 2], threonine (threonine dehydrogenase), choline via sarcosine (sarcosine dehydrogenase), and alanine and glyoxylate [alanine-glyoxylate aminotransferase (AGXT)].20,99, 100, 101 Glycine degradation occurs through the glycine cleavage system or its reverse conversion to serine via SHMT, sarcosine via glycine N-methyltransferase, or glyoxylate via D-amino acid oxidase.99,102, 103, 104, 105 Emerging studies highlight a key role for glycine and its metabolism in ASCVD and related cardiometabolic diseases, indicating its potential as both a biomarker and an interventional strategy.106

Lower circulating glycine is consistently reported to be associated with a higher risk of CVD and cardiometabolic risk factors, including metabolic syndrome, obesity, hypertension, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD).21,34, 35, 36, 37,107,108 In a meta-analysis of genome-wide associations studies, including over half a million participants with and without coronary heart disease, Wittemans et al17 found that higher circulating glycine is genetically associated with lower coronary heart disease risk. This finding was corroborated through observational studies including 2053 coronary heart disease cases and 11,147 controls. Ding et al37 found that plasma glycine levels are inversely correlated with the risk of acute myocardial infarction in a cohort of >4000 patients with suspected stable angina pectoris (Table 1).

The consistent reports of lower circulating glycine in cardiometabolic diseases have sparked interest in the metabolic mechanisms behind this phenomenon. In MASLD, which is closely related to ASCVD,109,110 the decrease in circulating glycine is attributed to the hepatic suppression of key glycine biosynthetic enzymes, including SHMT1, SHMT2, and AGXT.107,111 Furthermore, lower glycine availability is a limiting factor for de novo synthesis of the potent antioxidant glutathione.107,112 Addressing the inverse association between lower glycine and higher BCAAs in cardiometabolic diseases, White et al113 applied a stable isotope tracing approach, suggesting that the obesity-associated increase in BCAAs activates an interorgan pyruvate-alanine shuttle system that drives glycine depletion. Nevertheless, using comprehensive stable-isotope tracing studies combined with SHMT inhibition, Ghrayeb et al114 reported that enhanced conversion of glycine to serine via reverse SHMT activity, independent of BCAAs, is the major cause for decreased circulating glycine. Although these studies primarily used dietary mouse models, Tan et al115 demonstrated that de novo glycine synthesis is inhibited in patients with morbid obesity and enhanced following bariatric surgery. A decrease in the ratios of glycine, its precursors (serine and threonine), and related metabolites (oxalate) was found in serum from patients with significant CAD, supporting the notion of defective glycine biosynthesis or enhanced glycine use in ASCVD.20 Despite these findings, further metabolic flux studies are warranted to establish whether impaired glycine biosynthesis or enhanced glycine degradation is the cause of reduced circulating glycine in patients with ASCVD, independent of other metabolic risk factors (ie, obesity and MASLD).

Beyond the association between lower circulating glycine, ASCVD, and cardiometabolic risk factors, accumulating animal studies using dietary, genetic, and pharmacologic approaches support the causative role and therapeutic potential of glycine supplementation.11 In Apoe−/− mice, dietary glycine restriction not only increases hepatic steatosis,107 but also accelerates the formation of atherosclerotic plaques measured by plaque area.11,21 Similarly, the loss of AGXT, which catalyze glycine formation from glyoxylate, results in a decreased glycine/oxalate ratio, accelerated MASLD in C57BL/6J mice, and enhanced atherosclerosis in Apoe−/− mice through induction of proinflammatory and proatherogenic chemokines (mainly chemokine C-C motif ligand 5) in the liver, plasma, and atherosclerotic plaque.20, 107 In contrast, the antiatherogenic, anti-inflammatory, and antioxidant properties of glycine and glycine-based treatments are demonstrated in various in vitro, rodent, and nonhuman primate models. In a murine macrophage model system, glycine had the most potent lipid-lowering effects among all amino acids through suppression of very low-density lipoprotein uptake and triglyceride biosynthesis.96 In splenic and alveolar macrophages as well as Kupffer cells, glycine blunts lipopolysaccharide- or peptidoglycan polysaccharide–induced translocation of the p65 subunit of NF-κB into the nucleus and the production of tumor necrosis factor-α and superoxide by increasing chloride influx via a glycine-gated chloride channel.116, 117, 118 Treating with glycine or a glycine-based therapeutic (DT-109) induces de novo glutathione synthesis in macrophages, and attenuates atherosclerosis in Apoe−/− mice concomitant with decreased superoxide21 (Table 2). Similarly, glycine supplementation enhances glutathione formation and lowers superoxide production in a rat model of high-sucrose diet.119 Glycine and DT-109 show additional cardiometabolic benefits by lowering ASCVD risk factors, including obesity, blood pressure, and MASLD, in rodents and nonhuman primates.107,120, 121, 122, 123

Although the antiatherogenic properties and cardiometabolic benefits of glycine-based treatments have been consistently demonstrated in animal studies, the number of clinical studies have been limited. In patients with type 2 diabetes, supplementation with glycine (5 g/day from 3 months) not only improves glycemia, but also decreases proinflammatory cytokines.124 In patients with metabolic syndrome, treatment with glycine (15 g/day from 3 months) lowers circulating lipid peroxidation and systolic blood pressure.125 Moreover, in patients with MASLD, supplementation with 200 mg/kg per day of the glycine precursor, serine, lowers hepatic steatosis and circulating transaminases.112 Despite the success of these studies, the potential benefits of glycine-based treatments in patients with ASCVD have not been systematically addressed and warrant further clinical evaluation.

Aromatic Amino Acids and Related Metabolites

The AAAs, tryptophan, tyrosine, and phenylalanine, are essential nutrients in the human diet and play a role in various physiological processes. They are also precursors for important neurotransmitters and hormones, including serotonin,126 dopamine,127 and thyroid hormones.128 The epidemiology of AAAs in relation to atherosclerosis is complex, with some studies suggesting that elevated levels of specific AAAs are associated with an increased risk of CVD, atherosclerosis progression, and development.38,40,129, 130, 131, 132 One major limitation of the previous studies investigating the link between AAAs and atherosclerosis is that they are mostly observational and cross-sectional, which makes it difficult to establish a causal relationship. Although there is limited evidence of causation, potential mechanisms behind these associations include altered immune function, increased oxidative stress, and endothelial dysfunction. This section explores epidemiologic evidence along with basic research on the relationship and potential causative roles of AAAs and their metabolites in ASCVD. It specifically focuses on the effects of AAAs and their related metabolites on endothelial dysfunction, inflammation, and oxidative stress, key factors in the pathogenesis of atherosclerosis.

Tryptophan is an essential amino acid,133 which can be synthesized by microorganisms via the shikimic acid pathway134 and the activity of tryptophan synthase A (α-subunit) and tryptophan synthase B (β-subunit).135,136 Tryptophan can be metabolized to serotonin and melatonin through hydroxylation and acetylation via tryptophan hydroxylase and arylalkylamine N-acetyltransferase, respectively.137 The liver is responsible for 90% of tryptophan degradation in the kynurenine pathway through the conversion of tryptophan to kynurenine by tryptophan 2,3-dioxygenase. The remaining degradation of tryptophan in the kynurenine pathway occurs through indoleamine 2,3-dioxygenase in the brain, gastrointestinal tract, and liver.138,139 Tryptophan and kynurenine metabolites have emerged as intriguing players associated with atherosclerotic plaque progression.140 Low levels of circulating tryptophan are associated with increased risk of CVD and CAD in various clinical studies.38,40,129 Higher levels of tryptophan metabolites, such as kynurenine and 3-hydroxykynurenine, predict the incidence of cardiovascular risk in patients with end-stage renal disease and patients with angina pectoris, and are correlated with subclinical atherosclerosis and CVD39,141 (Table 1). These observations indicate that tryptophan metabolism may be dysfunctional in ASCVD and provide a rationale for studying the potential causative role of altered tryptophan-kynurenine metabolism in the pathogenesis of atherosclerosis.

Tryptophan metabolites, such as 3-hydroxyanthranilic acid and indoxyl sulfate, are positively associated with proinflammatory and pro-oxidant indexes linked to carotid atherosclerosis in patients with end-stage renal disease.141,142 Indoxyl sulfate activates NADPH oxidase, leading to the generation of reactive oxygen species and NF-κB activation, enhancing the expression of intercellular adhesion molecule-1 and vascular cell adhesion molecule-1143 and activation of endothelial cells.144,145 Cole et al15 found an increase in lesion size and markers of plaque vulnerability, as well as a reduction in IL-10 production, in indoleamine 2,3-dioxygenase–deficient Apoe−/− mice. Moreover, administration of 3,4-dimethoxycinna-moyl anthranilic acid, a synthetic derivative of the tryptophan metabolite anthranilic acid, led to reduced lesion formation and inflammation after collar-induced arterial injury, and reduced cytokine and chemokine production in human atheroma cell cultures15 (Table 2). In contrast, Metghalchi et al16 showed that indoleamine 2,3-dioxygenase deficiency in Ldlr−/− mice protects against atherosclerosis by decreasing the indoleamine 2,3-dioxygenase–derived metabolite kynurenic acid, which reduces IL-10 production through a cAMP-dependent pathway and inhibition of extracellular signal-regulated kinase 1/2 phosphorylation in myeloid cells, leading to enhanced susceptibility to atherosclerosis and colitis. Kynurenic acid levels in the blood predict death and recurrent myocardial infarction in patients with CAD.16 In addition, administration of 3-hydroxyanthranilic acid significantly decreases lesion size in Ldlr−/− mice by lowering plasma cholesterol and triglycerides and inhibiting the uptake of oxidized low-density lipoprotein by macrophages49 (Table 2). Together, although these findings indicate dysregulated tryptophan metabolism in ASCVD, it is likely that there are strain-, cell type–, and tryptophan metabolite–specific effects on atherosclerosis. Further investigation is warranted to clarify these discrepancies and evaluate the therapeutic value of targeting tryptophan metabolism for the treatment of ASCVD.

Tyrosine is obtained from the diet and can also be synthesized via phenylalanine hydroxylase, which catalyzes the addition of a hydroxyl group to the 6-carbon aromatic ring of phenylalanine to yield tyrosine.146 Tyrosine can be metabolized to generate neurotransmitters, such as l-3,4-dihydroxyphenylalanine, dopamine, adrenaline, and noradrenaline, via tyrosine hydroxylase,147 or it can be catalyzed by thyroperoxidase to produce hormones, such as thyroxine and triiodothyronine.148,149 Clinical studies have shown an association between elevated serum levels of tyrosine and an increased risk of atherosclerosis and cardiovascular events40,41 (Table 1). In individuals at risk of developing CVD, elevated tyrosine levels are associated with an increased carotid intima-media thickness, a marker of early atherosclerosis.40 Nuclear magnetic resonance–based metabolomic profiling revealed association of serum concentrations of tyrosine with increased 3-month mortality in patients with acute heart failure.150 Furthermore, reduction in tyrosine aminotransferase, which catalyzes the conversion of tyrosine to 4-hydroxyphenylpyruvate, or administration of tyrosine lowers glutathione and enhances oxidative stress in endothelial cells and the brain.151, 152, 153 Clearly, altered tyrosine metabolism is associated with ASCVD; however, further research is needed to establish a potential causative role.

Phenylalanine is also synthesized through the shikimate pathway.154 Its degradation to tyrosine occurs primarily in the liver by a complex system composed of phenylalanine hydroxylase, the pterin coenzyme tetrahydrobiopterin (BH4), and several enzymes involved in BH4 regeneration, including dihydropteridine reductase and pterin 4α-carbinolamine dehydratase. These can be further metabolized to acetoacetic acid and fumaric acid via hydroxyphenylpyruvic acid.155 Elevated circulating levels of phenylalanine are positively correlated with multiple risk factors for ASCVD, including systolic blood pressure, low-density lipoprotein cholesterol, triglycerides, and high-sensitivity C-reactive protein.42 Furthermore, increased circulating phenylalanine is significantly linked to CAD, ischemic stroke, and CVD events, as well as unexpected cardiac failure41,156 (Table 1). Although phenylalanine, tyrosine, or their ratio was not significantly different between patients with or without CAD, the phenylalanine/tyrosine ratio was associated with higher levels of serum neopterin or C-reactive protein in a large cohort of 3316 patients.157 BH4 preserves vascular function by regulating NO synthase activity and maintaining endothelial function, whereas BH4 deficiency promotes oxidative stress, endothelial dysfunction, reduced NO bioavailability, increased vascular inflammation, and the progression of atherosclerosis. Oral administration of BH4 to Apoe−/− mice restores NO-dependent endothelial function and attenuates the progression of atherosclerosis.158 Interestingly, phenylacetylglutamine, a gut microbial metabolite of phenylalanine, is associated with elevated risk of CVD, particularly in individuals with type 2 diabetes.159 Phenylacetylglutamine enhances platelet activation and increases thrombosis potential in whole blood, isolated platelets, and animal models of arterial injury. The microbial porA gene facilitates the conversion of dietary phenylalanine into phenylacetic acid, leading to the production of phenylacetylglutamine and phenylacetylglycine, which promotes platelet responsiveness and thrombosis potential through adrenergic receptors.159 Taken together, enhanced circulating phenylalanine is consistently elevated in CVD; however, further studies are needed to identify the underlying metabolic mechanisms, and the potential causative role of dysregulated phenylalanine in ASCVD.

Selenocysteine

Selenocysteine, recognized as the 21st protein-encoding amino acid, is notably present in certain enzymes that contribute to oxidation-reduction homeostasis, such as thioredoxin reductases, glutathione peroxidases, and glycine reductases. This amino acid is a variant of cysteine in which selenium, typically obtained from dietary sources, replaces sulfur. Interestingly, selenocysteine possesses a more potent reducing capacity than that of cysteine. This property enables proteins incorporating selenocysteine, referred to as selenoproteins, to perform antioxidant functions.

The glutathione peroxidase family of antioxidant selenoproteins offers protection against lipid peroxidation and oxidative damage.160,161 Genetic strategies targeting glutathione peroxidases have shown that their deficiency plays a role in atherosclerosis progression.162,163 Overexpression of glutathione peroxidase, which augments vascular reactivity, decreases lesion area in Apoe−/− mice.163 Supplementation of selenium and vitamin E decreases lesion size in rabbits fed a high-fat diet, independent of cholesterol levels,164 highlighting the importance of selenoproteins. Human studies have demonstrated similar findings, with a meta-analysis revealing an inverse correlation between serum selenium levels and cardiovascular events.165 This finding was supported by cohort studies reporting high selenium levels are associated with reduced risk of CVD and acute CAD.166 More recently, a meta-analysis found an association between decreased CVD mortality and high selenium intake.167 Despite these associations between selenium levels and cardiovascular health, selenium supplementation has proven insufficient to protect against cardiovascular outcomes in humans,166,168 potentially because of a defect in the incorporation of selenium into selenoproteins. In support of this idea, the Denmark PREvention of Cancer by Intervention with Selenium (PRECISE) trial showed that long-term selenium supplementation increased all-cause mortality.169 Future studies should focus on the role of selenium and selenoprotein synthesis and activity to better inform future therapies for individuals at risk for ASCVD.

Concluding Remarks and Future Directions

This review explored the accumulating evidence over the past several decades on amino acid metabolism in relation to ASCVD, covering epidemiologic, clinical, animal, and in vitro studies on arginine and its metabolites, BCAAs, glycine, AAAs, and selenocysteine. When combining the observational clinical studies, there is a unique signature of an altered amino acid profile associated with ASCVD that includes elevated circulating BCAAs with a concurrent reduction in circulating glycine (Table 1). Although the observational studies are relatively consistent with regard to the altered amino acid profile associated with ASCVD risk, animal studies addressing their causative role in atherosclerosis are often contradictory, specifically those focusing on BCAA and AAA metabolism. Currently, direct comparisons between human data and rodent studies are difficult as there is a wide variation in the rodent models. Most human studies measure amino acids in the circulation of patients with CVD, whereas a significant portion of animal studies center around administering amino acids in conjunction with diets that induce atherosclerosis, hindering direct side-by-side comparisons. In addition, most of the human and animal studies have also relied on cross-sectional designs, limiting the understanding of the temporal relationships between amino acid levels and ASCVD progression.

Another limitation of the research on amino acids and ASCVD so far, which offers new avenues of investigation, is a unifying approach to study the interactions among amino acid metabolism, disease progression, and severity. Most studies have primarily focused on individual amino acids or metabolites, and little is known about their interactions or combined effects in the context of atherosclerosis. So far, such interactions have been addressed in the context of altered glycine metabolism in human ASCVD, where lower ratios of glycine and its precursors, serine and threonine, have been associated with disease severity,21,170 suggesting impaired glycine availability in relation to ASCVD progression. Currently, only two animal studies have explored such interactions, indicating that glycine depletion could be the result of either enhanced BCAAs that activate an interorgan pyruvate-alanine shuttle system113 or the enhanced conversion of glycine to serine via reverse SHMT activity.114 However, these studies were performed in mouse models of obesity and MASLD rather than atherosclerosis. Therefore, to establish a unifying understanding of impaired amino acid metabolism in ASCVD, a comprehensive investigation of the interactions among amino acid metabolism in relation to the pathogenesis of ASCVD is needed and can be achieved through a combination of clinical and animal studies applying metabolomics and tracing techniques.

Finally, the insufficient evidence from human studies is another limitation that warrants further investigation. Longitudinal studies with large and diverse populations are warranted to investigate the association between amino acid levels, their metabolites, and the incidence, progression, and outcomes of ASCVD. Such studies should also consider potential confounding factors, including dietary patterns, lifestyle factors, and coexisting diseases. Moreover, although arginine intervention has been studied in earlier clinical trials of CVD, further clinical evidence on the effects of other amino acids on ASCVD is lacking. Intervention studies are needed to determine the effects of targeted modulation of amino acids or their metabolites on ASCVD progression and outcomes. Randomized controlled trials assessing the efficacy and safety of interventions, such as amino acid supplementation or dietary modifications, are crucial for establishing evidence-based therapeutic strategies. Finally, metabolomics studies have focused on the relationship between the circulating amino acid profile and ASCVD. The emergence of new technologies, including spatial metabolomics and transcriptomics,171 may allow a better understanding of altered amino acid metabolism within different regions and cell types of the human atherosclerotic plaque. Advancements in these fields hold promise in identifying novel diagnostic and prognostic biomarkers as well as therapeutic approaches for ASCVD.

Disclosure Statement

None declared.

Footnotes

Advances in Understanding Cardiovascular Disease Pathogenesis Theme Issue

Supported in part by NIH grants HL145131 and HL167758 (A.Y.), HL139755 (C.B.P), and HL150233, DK134011, and DK136685 (O.R.).

This article is part of a review series highlighting the novel insights provided by next-generation approaches to our understanding of cardiovascular disease pathogenesis and treatment.

Contributor Information

Christopher B. Pattillo, Email: christopher.pattillo@lsuhs.edu.

Oren Rom, Email: oren.rom@lsuhs.edu.

References

  • 1.Weissberg P.L., Bennett M.R. Atherosclerosis--an inflammatory disease. N Engl J Med. 1999;340:1928–1929. [PubMed] [Google Scholar]
  • 2.Otsuka F., Kramer M.C.A., Woudstra P., Yahagi K., Ladich E., Finn A.V., de Winter R.J., Kolodgie F.D., Wight T.N., Davis H.R., Joner M., Virmani R. Natural progression of atherosclerosis from pathologic intimal thickening to late fibroatheroma in human coronary arteries: a pathology study. Atherosclerosis. 2015;241:772–782. doi: 10.1016/j.atherosclerosis.2015.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Libby P., Buring J.E., Badimon L., Hansson G.K., Deanfield J., Bittencourt M.S., Tokgözoğlu L., Lewis E.F. Atherosclerosis. Nat Rev Dis Primers. 2019;5:56. doi: 10.1038/s41572-019-0106-z. [DOI] [PubMed] [Google Scholar]
  • 4.Muller J.E., Tofler G., Stone P. Circadian variation and triggers of onset of acute cardiovascular disease. Circulation. 1989;79:733–743. doi: 10.1161/01.cir.79.4.733. [DOI] [PubMed] [Google Scholar]
  • 5.Newby A.C. Dual role of matrix metalloproteinases (matrixins) in intimal thickening and atherosclerotic plaque rupture. Physiol Rev. 2005;85:1–31. doi: 10.1152/physrev.00048.2003. [DOI] [PubMed] [Google Scholar]
  • 6.Tsao C.W., Aday A.W., Almarzooq Z.I., Anderson C.A., Arora P., Avery C.L., Baker-Smith C.M., Beaton A.Z., Boehme A.K., Buxton A.E. Heart disease and stroke statistics—2023 update: a report from the American Heart Association. Circulation. 2023;147:e93–e621. doi: 10.1161/CIR.0000000000001123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ignatowski A. Influence of animal food on the organsim of rabbits. Izvest Imper Voennomed Akad St Petersburg. 1908;16:154–173. [Google Scholar]
  • 8.Low Wang C.C., Hess C.N., Hiatt W.R., Goldfine A.B. Clinical update: cardiovascular disease in diabetes mellitus: atherosclerotic cardiovascular disease and heart failure in type 2 diabetes mellitus–mechanisms, management, and clinical considerations. Circulation. 2016;133:2459–2502. doi: 10.1161/CIRCULATIONAHA.116.022194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wu G. Amino acids: metabolism, functions, and nutrition. Amino Acids. 2009;37:1–17. doi: 10.1007/s00726-009-0269-0. [DOI] [PubMed] [Google Scholar]
  • 10.FitzPatrick M., Hospelhorn V.D. Changes in amino acid composition of aortic elastin with aging and atherosclerosis. Am Heart J. 1965;69:211–214. doi: 10.1016/0002-8703(65)90039-6. [DOI] [PubMed] [Google Scholar]
  • 11.Katan M.B., Vroomen L.H.M., Hermus R.J.J. Reduction of casein-induced hypercholesterolaemia and atherosclerosis in rabbits and rats by dietary glycine, arginine and alanine. Atherosclerosis. 1982;43:381–391. doi: 10.1016/0021-9150(82)90037-5. [DOI] [PubMed] [Google Scholar]
  • 12.Wang B.Y., Singer A.H., Tsao P.S., Drexler H., Kosek J., Cooke J.P. Dietary arginine prevents atherogenesis in the coronary artery of the hypercholesterolemic rabbit. J Am Coll Cardiol. 1994;23:452–458. doi: 10.1016/0735-1097(94)90433-2. [DOI] [PubMed] [Google Scholar]
  • 13.Nygård O., Nordrehaug J.E., Refsum H., Ueland P.M., Farstad M., Vollset S.E. Plasma homocysteine levels and mortality in patients with coronary artery disease. N Engl J Med. 1997;337:230–237. doi: 10.1056/NEJM199707243370403. [DOI] [PubMed] [Google Scholar]
  • 14.Shah S.H., Bain J.R., Muehlbauer M.J., Stevens R.D., Crosslin D.R., Haynes C., Dungan J., Newby L.K., Hauser E.R., Ginsburg G.S. Association of a peripheral blood metabolic profile with coronary artery disease and risk of subsequent cardiovascular events. Circ Cardiovasc Genet. 2010;3:207–214. doi: 10.1161/CIRCGENETICS.109.852814. [DOI] [PubMed] [Google Scholar]
  • 15.Cole J.E., Astola N., Cribbs A.P., Goddard M.E., Park I., Green P., Davies A.H., Williams R.O., Feldmann M., Monaco C. Indoleamine 2, 3-dioxygenase-1 is protective in atherosclerosis and its metabolites provide new opportunities for drug development. Proc Natl Acad Sci U S A. 2015;112:13033–13038. doi: 10.1073/pnas.1517820112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Metghalchi S., Ponnuswamy P., Simon T., Haddad Y., Laurans L., Clement M., Dalloz M., Romain M., Esposito B., Koropoulis V. Indoleamine 2, 3-dioxygenase fine-tunes immune homeostasis in atherosclerosis and colitis through repression of interleukin-10 production. Cell Metab. 2015;22:460–471. doi: 10.1016/j.cmet.2015.07.004. [DOI] [PubMed] [Google Scholar]
  • 17.Wittemans L.B., Lotta L.A., Oliver-Williams C., Stewart I.D., Surendran P., Karthikeyan S., Day F.R., Koulman A., Imamura F., Zeng L. Assessing the causal association of glycine with risk of cardio-metabolic diseases. Nat Commun. 2019;10:1060. doi: 10.1038/s41467-019-08936-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zhang X., Sergin I., Evans T.D., Jeong S.-J., Rodriguez-Velez A., Kapoor D., Chen S., Song E., Holloway K.B., Crowley J.R. High-protein diets increase cardiovascular risk by activating macrophage mTOR to suppress mitophagy. Nat Metab. 2020;2:110–125. doi: 10.1038/s42255-019-0162-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yurdagul A., Jr., Subramanian M., Wang X., Crown S.B., Ilkayeva O.R., Darville L., Kolluru G.K., Rymond C.C., Gerlach B.D., Zheng Z., Kuriakose G., Kevil C.G., Koomen J.M., Cleveland J.L., Muoio D.M., Tabas I. Macrophage metabolism of apoptotic cell-derived arginine promotes continual efferocytosis and resolution of injury. Cell Metab. 2020;31:518–533.e10. doi: 10.1016/j.cmet.2020.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Liu Y., Zhao Y., Shukha Y., Lu H., Wang L., Liu Z., Liu C., Zhao Y., Wang H., Zhao G., Liang W., Fan Y., Chang L., Yurdagul A., Jr., Pattillo C.B., Orr A.W., Aviram M., Wen B., Garcia-Barrio M.T., Zhang J., Liu W., Sun D., Hayek T., Chen Y.E., Rom O. Dysregulated oxalate metabolism is a driver and therapeutic target in atherosclerosis. Cell Rep. 2021;36 doi: 10.1016/j.celrep.2021.109420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rom O., Liu Y., Finney A.C., Ghrayeb A., Zhao Y., Shukha Y., Wang L., Rajanayake K.K., Das S., Rashdan N.A., Weissman N., Delgadillo L., Wen B., Garcia-Barrio M.T., Aviram M., Kevil C.G., Yurdagul A., Jr., Pattillo C.B., Zhang J., Sun D., Hayek T., Gottlieb E., Mor I., Chen Y.E. Induction of glutathione biosynthesis by glycine-based treatment mitigates atherosclerosis. Redox Biol. 2022;52 doi: 10.1016/j.redox.2022.102313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Nitz K., Lacy M., Atzler D. Amino acids and their metabolism in atherosclerosis. Arterioscler Thromb Vasc Biol. 2019;39:319–330. doi: 10.1161/ATVBAHA.118.311572. [DOI] [PubMed] [Google Scholar]
  • 23.Wu G., Morris S.M., Jr. Arginine metabolism: nitric oxide and beyond. Biochem J. 1998;336(Pt 1):1–17. doi: 10.1042/bj3360001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bode-Boger S.M., Boger R.H., Alfke H., Heinzel D., Tsikas D., Creutzig A., Alexander K., Frolich J.C. L-arginine induces nitric oxide-dependent vasodilation in patients with critical limb ischemia: a randomized, controlled study. Circulation. 1996;93:85–90. doi: 10.1161/01.cir.93.1.85. [DOI] [PubMed] [Google Scholar]
  • 25.Cheung P.Y., Salas E., Schulz R., Radomski M.W. Nitric oxide and platelet function: implications for neonatology. Semin Perinatol. 1997;21:409–417. doi: 10.1016/s0146-0005(97)80006-7. [DOI] [PubMed] [Google Scholar]
  • 26.Kubes P., Suzuki M., Granger D.N. Nitric oxide: an endogenous modulator of leukocyte adhesion. Proc Natl Acad Sci U S A. 1991;88:4651–4655. doi: 10.1073/pnas.88.11.4651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ricard N., Bailly S., Guignabert C., Simons M. The quiescent endothelium: signalling pathways regulating organ-specific endothelial normalcy. Nat Rev Cardiol. 2021;18:565–580. doi: 10.1038/s41569-021-00517-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Pilz S., Meinitzer A., Tomaschitz A., Drechsler C., Ritz E., Krane V., Wanner C., Boehm B.O., Marz W. Low homoarginine concentration is a novel risk factor for heart disease. Heart. 2011;97:1222–1227. doi: 10.1136/hrt.2010.220731. [DOI] [PubMed] [Google Scholar]
  • 29.Atzler D., Baum C., Ojeda F., Keller T., Cordts K., Schnabel R.B., Choe C.U., Lackner K.J., Munzel T., Boger R.H., Blankenberg S., Schwedhelm E., Zeller T. Low homoarginine levels in the prognosis of patients with acute chest pain. J Am Heart Assoc. 2016;5 doi: 10.1161/JAHA.115.002565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Karetnikova E.S., Jarzebska N., Markov A.G., Weiss N., Lentz S.R., Rodionov R.N. Is homoarginine a protective cardiovascular risk factor? Arterioscler Thromb Vasc Biol. 2019;39:869–875. doi: 10.1161/ATVBAHA.118.312218. [DOI] [PubMed] [Google Scholar]
  • 31.Cuomo P., Capparelli R., Iannelli A., Iannelli D. Role of branched-chain amino acid metabolism in type 2 diabetes, obesity, cardiovascular disease and non-alcoholic fatty liver disease. Int J Mol Sci. 2022;23:4325. doi: 10.3390/ijms23084325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mels C., Schutte A., Schutte R., Huisman H., Smith W., Fourie C., Kruger R., Van Rooyen J., Malan N., Malan L. The link between vascular deterioration and branched chain amino acids in a population with high glycated haemoglobin: the SABPA study. Amino Acids. 2013;45:1405–1413. doi: 10.1007/s00726-013-1611-0. [DOI] [PubMed] [Google Scholar]
  • 33.Writing Group Members. Mozaffarian D., Benjamin E.J., Go A., Arnett D., Blaha M., et al. Executive summary: heart disease and stroke statistics--2016 update: a report from the American Heart Association. Circulation. 2016;133:447–454. doi: 10.1161/CIR.0000000000000366. [DOI] [PubMed] [Google Scholar]
  • 34.Ntzouvani A., Nomikos T., Panagiotakos D., Fragopoulou E., Pitsavos C., McCann A., Ueland P., Antonopoulou S. Amino acid profile and metabolic syndrome in a male Mediterranean population: a cross-sectional study. Nutr Metab Cardiovasc Dis. 2017;27:1021–1030. doi: 10.1016/j.numecd.2017.07.006. [DOI] [PubMed] [Google Scholar]
  • 35.Gaggini M., Carli F., Rosso C., Buzzigoli E., Marietti M., Della Latta V., Ciociaro D., Abate M.L., Gambino R., Cassader M. Altered amino acid concentrations in NAFLD: impact of obesity and insulin resistance. Hepatology. 2018;67:145–158. doi: 10.1002/hep.29465. [DOI] [PubMed] [Google Scholar]
  • 36.Li X., Sun L., Zhang W., Li H., Wang S., Mu H., Zhou Q., Zhang Y., Tang Y., Wang Y., Chen W., Yang R. Association of serum glycine levels with metabolic syndrome in an elderly Chinese population. Nutr Metab (Lond) 2018;15:89. doi: 10.1186/s12986-018-0325-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ding Y., Svingen G.F., Pedersen E.R., Gregory J.F., Ueland P.M., Tell G.S., Nygard O.K. Plasma glycine and risk of acute myocardial infarction in patients with suspected stable angina pectoris. J Am Heart Assoc. 2015;5 doi: 10.1161/JAHA.115.002621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Yu E., Ruiz-Canela M., Guasch-Ferré M., Zheng Y., Toledo E., Clish C.B., Salas-Salvadó J., Liang L., Wang D.D., Corella D. Increases in plasma tryptophan are inversely associated with incident cardiovascular disease in the Prevencion con Dieta Mediterranea (PREDIMED) Study. J Nutr. 2017;147:314–322. doi: 10.3945/jn.116.241711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Pedersen E.R., Tuseth N., Eussen S.J., Ueland P.M., Strand E., Svingen G.F.T., Midttun Ø., Meyer K., Mellgren G., Ulvik A. Associations of plasma kynurenines with risk of acute myocardial infarction in patients with stable angina pectoris. Arterioscler Thromb Vasc Biol. 2015;35:455–462. doi: 10.1161/ATVBAHA.114.304674. [DOI] [PubMed] [Google Scholar]
  • 40.Würtz P., Raiko J.R., Magnussen C.G., Soininen P., Kangas A.J., Tynkkynen T., Thomson R., Laatikainen R., Savolainen M.J., Laurikka J. High-throughput quantification of circulating metabolites improves prediction of subclinical atherosclerosis. Eur Heart J. 2012;33:2307–2316. doi: 10.1093/eurheartj/ehs020. [DOI] [PubMed] [Google Scholar]
  • 41.Jauhiainen R., Vangipurapu J., Laakso A., Kuulasmaa T., Kuusisto J., Laakso M. The association of 9 amino acids with cardiovascular events in Finnish men in a 12-year follow-up study. J Clin Endocrinol Metab. 2021;106:3448–3454. doi: 10.1210/clinem/dgab562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Würtz P., Havulinna A.S., Soininen P., Tynkkynen T., Prieto-Merino D., Tillin T., Ghorbani A., Artati A., Wang Q., Tiainen M., Kangas A.J., Kettunen J., Kaikkonen J., Mikkilä V., Jula A., Kähönen M., Lehtimäki T., Lawlor D.A., Gaunt T.R., Hughes A.D., Sattar N., Illig T., Adamski J., Wang T.J., Perola M., Ripatti S., Vasan R.S., Raitakari O.T., Gerszten R.E., Casas J.-P., Chaturvedi N., Ala-Korpela M., Salomaa V. Metabolite profiling and cardiovascular event risk. Circulation. 2015;131:774–785. doi: 10.1161/CIRCULATIONAHA.114.013116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Nitz K., Lacy M., Bianchini M., Wichapong K., Kucukgoze I.A., Bonfiglio C.A., Migheli R., Wu Y., Burger C., Li Y., Forne I., Ammar C., Janjic A., Mohanta S., Duchene J., Heemskerk J.W.M., Megens R.T.A., Schwedhelm E., Huveneers S., Lygate C.A., Santovito D., Zimmer R., Imhof A., Weber C., Lutgens E., Atzler D. The amino acid homoarginine inhibits atherogenesis by modulating T-cell function. Circ Res. 2022;131:701–712. doi: 10.1161/CIRCRESAHA.122.321094. [DOI] [PubMed] [Google Scholar]
  • 44.Yurdagul A., Jr., Kong N., Gerlach B.D., Wang X., Ampomah P., Kuriakose G., Tao W., Shi J., Tabas I. ODC (ornithine decarboxylase)-dependent putrescine synthesis maintains MerTK (MER tyrosine-protein kinase) expression to drive resolution. Arterioscler Thromb Vasc Biol. 2021;41:e144–e159. doi: 10.1161/ATVBAHA.120.315622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Michiels C.F., Kurdi A., Timmermans J.P., De Meyer G.R.Y., Martinet W. Spermidine reduces lipid accumulation and necrotic core formation in atherosclerotic plaques via induction of autophagy. Atherosclerosis. 2016;251:319–327. doi: 10.1016/j.atherosclerosis.2016.07.899. [DOI] [PubMed] [Google Scholar]
  • 46.Zhao Y., Dai X-y, Zhou Z., Zhao G-x, Wang X., Xu M-j. Leucine supplementation via drinking water reduces atherosclerotic lesions in apoE null mice. Acta Pharmacol Sin. 2016;37:196–203. doi: 10.1038/aps.2015.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Li Z., Zhang R., Mu H., Zhang W., Zeng J., Li H., Wang S., Zhao X., Chen W., Dong J. Oral administration of branched-chain amino acids attenuates atherosclerosis by inhibiting the inflammatory response and regulating the gut microbiota in ApoE-deficient mice. Nutrients. 2022;14:5065. doi: 10.3390/nu14235065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zhao S., Zhou L., Wang Q., Cao J.-H., Chen Y., Wang W., Zhu B.-D., Wei Z.-H., Li R., Li C.-Y., Zhou G.-Y., Tan Z.-J., Zhou H.-P., Li C.-X., Gao H.-K., Qin X.-J., Lian K. Elevated branched-chain amino acid promotes atherosclerosis progression by enhancing mitochondrial-to-nuclear H2O2-disulfide HMGB1 in macrophages. Redox Biol. 2023;62 doi: 10.1016/j.redox.2023.102696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhang L., Ovchinnikova O., Jönsson A., Lundberg A.M., Berg M., Hansson G.K., Ketelhuth D.F.J. The tryptophan metabolite 3-hydroxyanthranilic acid lowers plasma lipids and decreases atherosclerosis in hypercholesterolaemic mice. Eur Heart J. 2012;33:2025–2034. doi: 10.1093/eurheartj/ehs175. [DOI] [PubMed] [Google Scholar]
  • 50.Casero R.A., Jr., Murray Stewart T., Pegg A.E. Polyamine metabolism and cancer: treatments, challenges and opportunities. Nat Rev Cancer. 2018;18:681–695. doi: 10.1038/s41568-018-0050-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Cai W., Dai X., Chen J., Zhao J., Xu M., Zhang L., Yang B., Zhang W., Rocha M., Nakao T., Kofler J., Shi Y., Stetler R.A., Hu X., Chen J. STAT6/Arg1 promotes microglia/macrophage efferocytosis and inflammation resolution in stroke mice. JCI Insight. 2019;4 doi: 10.1172/jci.insight.131355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ha H.C., Sirisoma N.S., Kuppusamy P., Zweier J.L., Woster P.M., Casero R.A., Jr. The natural polyamine spermine functions directly as a free radical scavenger. Proc Natl Acad Sci U S A. 1998;95:11140–11145. doi: 10.1073/pnas.95.19.11140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Madeo F., Eisenberg T., Pietrocola F., Kroemer G. Spermidine in health and disease. Science. 2018;359 doi: 10.1126/science.aan2788. [DOI] [PubMed] [Google Scholar]
  • 54.Hofer S.J., Simon A.K., Bergmann M., Eisenberg T., Kroemer G., Madeo F. Mechanisms of spermidine-induced autophagy and geroprotection. Nat Aging. 2022;2:1112–1129. doi: 10.1038/s43587-022-00322-9. [DOI] [PubMed] [Google Scholar]
  • 55.Eisenberg T., Knauer H., Schauer A., Buttner S., Ruckenstuhl C., Carmona-Gutierrez D., Ring J., Schroeder S., Magnes C., Antonacci L., Fussi H., Deszcz L., Hartl R., Schraml E., Criollo A., Megalou E., Weiskopf D., Laun P., Heeren G., Breitenbach M., Grubeck-Loebenstein B., Herker E., Fahrenkrog B., Frohlich K.U., Sinner F., Tavernarakis N., Minois N., Kroemer G., Madeo F. Induction of autophagy by spermidine promotes longevity. Nat Cell Biol. 2009;11:1305–1314. doi: 10.1038/ncb1975. [DOI] [PubMed] [Google Scholar]
  • 56.De Meyer G.R., Grootaert M.O., Michiels C.F., Kurdi A., Schrijvers D.M., Martinet W. Autophagy in vascular disease. Circ Res. 2015;116:468–479. doi: 10.1161/CIRCRESAHA.116.303804. [DOI] [PubMed] [Google Scholar]
  • 57.Liao X., Sluimer J.C., Wang Y., Subramanian M., Brown K., Pattison J.S., Robbins J., Martinez J., Tabas I. Macrophage autophagy plays a protective role in advanced atherosclerosis. Cell Metab. 2012;15:545–553. doi: 10.1016/j.cmet.2012.01.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Razani B., Feng C., Coleman T., Emanuel R., Wen H., Hwang S., Ting J.P., Virgin H.W., Kastan M.B., Semenkovich C.F. Autophagy links inflammasomes to atherosclerotic progression. Cell Metab. 2012;15:534–544. doi: 10.1016/j.cmet.2012.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Sergin I., Razani B. Self-eating in the plaque: what macrophage autophagy reveals about atherosclerosis. Trends Endocrinol Metab. 2014;25:225–234. doi: 10.1016/j.tem.2014.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Jeong J.W., Cha H.J., Han M.H., Hwang S.J., Lee D.S., Yoo J.S., Choi I.W., Kim S., Kim H.S., Kim G.Y., Hong S.H., Park C., Lee H.J., Choi Y.H. Spermidine protects against oxidative stress in inflammation models using macrophages and zebrafish. Biomol Ther (Seoul) 2018;26:146–156. doi: 10.4062/biomolther.2016.272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Paul S., Kang S.C. Natural polyamine inhibits mouse skin inflammation and macrophage activation. Inflamm Res. 2013;62:681–688. doi: 10.1007/s00011-013-0620-5. [DOI] [PubMed] [Google Scholar]
  • 62.Eisenberg T., Abdellatif M., Schroeder S., Primessnig U., Stekovic S., Pendl T., et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat Med. 2016;22:1428–1438. doi: 10.1038/nm.4222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kiechl S., Pechlaner R., Willeit P., Notdurfter M., Paulweber B., Willeit K., Werner P., Ruckenstuhl C., Iglseder B., Weger S., Mairhofer B., Gartner M., Kedenko L., Chmelikova M., Stekovic S., Stuppner H., Oberhollenzer F., Kroemer G., Mayr M., Eisenberg T., Tilg H., Madeo F., Willeit J. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108:371–380. doi: 10.1093/ajcn/nqy102. [DOI] [PubMed] [Google Scholar]
  • 64.Wu H., Wang J., Jiang H., Liu X., Sun X., Chen Y., Hu C., Wang Z., Han T., Sun C., Wei W., Jiang W. The association of dietary spermidine with all-cause mortality and CVD mortality: the U.S. National Health and Nutrition Examination Survey, 2003 to 2014. Front Publ Health. 2022;10 doi: 10.3389/fpubh.2022.949170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Gimbrone M.A., Jr. Garcia-Cardena G: Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ Res. 2016;118:620–636. doi: 10.1161/CIRCRESAHA.115.306301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Schulman S.P., Becker L.C., Kass D.A., Champion H.C., Terrin M.L., Forman S., Ernst K.V., Kelemen M.D., Townsend S.N., Capriotti A., Hare J.M., Gerstenblith G. L-arginine therapy in acute myocardial infarction: the Vascular Interaction with Age in Myocardial Infarction (VINTAGE MI) randomized clinical trial. JAMA. 2006;295:58–64. doi: 10.1001/jama.295.1.58. [DOI] [PubMed] [Google Scholar]
  • 67.Harper A., Miller R., Block K. Branched-chain amino acid metabolism. Annu Rev Nutr. 1984;4:409–454. doi: 10.1146/annurev.nu.04.070184.002205. [DOI] [PubMed] [Google Scholar]
  • 68.Lynch C.J., Adams S.H. Branched-chain amino acids in metabolic signalling and insulin resistance. Nat Rev Endocrinol. 2014;10:723–736. doi: 10.1038/nrendo.2014.171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wolfe R.R. Branched-chain amino acids and muscle protein synthesis in humans: myth or reality? J Int Soc Sports Nutr. 2017;14:30. doi: 10.1186/s12970-017-0184-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.McGarrah R.W., White P.J. Branched-chain amino acids in cardiovascular disease. Nat Rev Cardiol. 2023;20:77–89. doi: 10.1038/s41569-022-00760-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Yang R., Dong J., Zhao H., Li H., Guo H., Wang S., Zhang C., Wang S., Wang M., Yu S. Association of branched-chain amino acids with carotid intima-media thickness and coronary artery disease risk factors. PLoS One. 2014;9 doi: 10.1371/journal.pone.0099598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bhattacharya S., Granger C.B., Craig D., Haynes C., Bain J., Stevens R.D., Hauser E.R., Newgard C.B., Kraus W.E., Newby L.K. Validation of the association between a branched chain amino acid metabolite profile and extremes of coronary artery disease in patients referred for cardiac catheterization. Atherosclerosis. 2014;232:191–196. doi: 10.1016/j.atherosclerosis.2013.10.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Newgard C.B., An J., Bain J.R., Muehlbauer M.J., Stevens R.D., Lien L.F., Haqq A.M., Shah S.H., Arlotto M., Slentz C.A. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009;9:311–326. doi: 10.1016/j.cmet.2009.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hatazawa Y., Tadaishi M., Nagaike Y., Morita A., Ogawa Y., Ezaki O., Takai-Igarashi T., Kitaura Y., Shimomura Y., Kamei Y. PGC-1[alpha]-mediated branched-chain amino acid metabolism in the skeletal muscle. PLoS One. 2014;9 doi: 10.1371/journal.pone.0091006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Burrage L.C., Nagamani S.C., Campeau P.M., Lee B.H. Branched-chain amino acid metabolism: from rare Mendelian diseases to more common disorders. Hum Mol Genet. 2014;23:R1–R8. doi: 10.1093/hmg/ddu123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.He L., Palos-Jasso A., Yi Y., Qin M., Qiu L., Yang X., Zhang Y., Yu J. Bioinformatic analysis revealed the essential regulatory genes and pathways of early and advanced atherosclerotic plaque in humans. Cells. 2022;11:3976. doi: 10.3390/cells11243976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Jennings A., MacGregor A., Welch A., Chowienczyk P., Spector T., Cassidy A. Amino acid intakes are inversely associated with arterial stiffness and central blood pressure in women. J Nutr. 2015;145:2130–2138. doi: 10.3945/jn.115.214700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Adeva M.M., Calviño J., Souto G., Donapetry C. Insulin resistance and the metabolism of branched-chain amino acids in humans. Amino Acids. 2012;43:171–181. doi: 10.1007/s00726-011-1088-7. [DOI] [PubMed] [Google Scholar]
  • 79.Yoon M.-S. The emerging role of branched-chain amino acids in insulin resistance and metabolism. Nutrients. 2016;8:405. doi: 10.3390/nu8070405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.White P.J., McGarrah R.W., Herman M.A., Bain J.R., Shah S.H., Newgard C.B. Insulin action, type 2 diabetes, and branched-chain amino acids: a two-way street. Mol Metab. 2021;52 doi: 10.1016/j.molmet.2021.101261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Tremblay F., Marette A. Amino acid and insulin signaling via the mTOR/p70 S6 kinase pathway: a negative feedback mechanism leading to insulin resistance in skeletal muscle cells. J Biol Chem. 2001;276:38052–38060. doi: 10.1074/jbc.M106703200. [DOI] [PubMed] [Google Scholar]
  • 82.Yoon M.-S. The role of mammalian target of rapamycin (mTOR) in insulin signaling. Nutrients. 2017;9:1176. doi: 10.3390/nu9111176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Khamzina L., Veilleux A., Bergeron S., Marette A. Increased activation of the mammalian target of rapamycin pathway in liver and skeletal muscle of obese rats: possible involvement in obesity-linked insulin resistance. Endocrinology. 2005;146:1473–1481. doi: 10.1210/en.2004-0921. [DOI] [PubMed] [Google Scholar]
  • 84.Leibowitz G., Cerasi E., Ketzinel-Gilad M. The role of mTOR in the adaptation and failure of [beta]-cells in type 2 diabetes. Diabetes Obes Metab. 2008;10:157–169. doi: 10.1111/j.1463-1326.2008.00952.x. [DOI] [PubMed] [Google Scholar]
  • 85.Magkos F., Bradley D., Schweitzer G.G., Finck B.N., Eagon J.C., Ilkayeva O., Newgard C.B., Klein S. Effect of Roux-en-Y gastric bypass and laparoscopic adjustable gastric banding on branched-chain amino acid metabolism. Diabetes. 2013;62:2757–2761. doi: 10.2337/db13-0185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Newgard C.B. Interplay between lipids and branched-chain amino acids in development of insulin resistance. Cell Metab. 2012;15:606–614. doi: 10.1016/j.cmet.2012.01.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Jang C., Oh S.F., Wada S., Rowe G.C., Liu L., Chan M.C., Rhee J., Hoshino A., Kim B., Ibrahim A. A branched-chain amino acid metabolite drives vascular fatty acid transport and causes insulin resistance. Nat Med. 2016;22:421–426. doi: 10.1038/nm.4057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wada E., Kobayashi M., Kohno D., Kikuchi O., Suga T., Matsui S., Yokota-Hashimoto H., Honzawa N., Ikeuchi Y., Tsuneoka H. Disordered branched chain amino acid catabolism in pancreatic islets is associated with postprandial hypersecretion of glucagon in diabetic mice. J Nutr Biochem. 2021;97 doi: 10.1016/j.jnutbio.2021.108811. [DOI] [PubMed] [Google Scholar]
  • 89.Karusheva Y., Strassburger K., Markgraf D.F., Zaharia O.-P., Bódis K., Kössler T., Tura A., Pacini G., Burkart V., Roden M. Branched-chain amino acids associate negatively with postprandial insulin secretion in recent-onset diabetes. J Endocr Soc. 2021;5 doi: 10.1210/jendso/bvab067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zhenyukh O., Civantos E., Ruiz-Ortega M., Sánchez M.S., Vazquez C., Peiro C., Egido J., Mas S. High concentration of branched-chain amino acids promotes oxidative stress, inflammation and migration of human peripheral blood mononuclear cells via mTORC1 activation. Free Radic Biol Med. 2017;104:165–177. doi: 10.1016/j.freeradbiomed.2017.01.009. [DOI] [PubMed] [Google Scholar]
  • 91.Ridaura V.K., Faith J.J., Rey F.E., Cheng J., Duncan A.E., Kau A.L., Griffin N.W., Lombard V., Henrissat B., Bain J.R. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341 doi: 10.1126/science.1241214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Pedersen H.K., Gudmundsdottir V., Nielsen H.B., Hyotylainen T., Nielsen T., Jensen B.A., Forslund K., Hildebrand F., Prifti E., Falony G. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature. 2016;535:376–381. doi: 10.1038/nature18646. [DOI] [PubMed] [Google Scholar]
  • 93.Qiao S., Liu C., Sun L., Wang T., Dai H., Wang K., Bao L., Li H., Wang W., Liu S.-J. Gut Parabacteroides merdae protects against cardiovascular damage by enhancing branched-chain amino acid catabolism. Nat Metab. 2022;4:1271–1286. doi: 10.1038/s42255-022-00649-y. [DOI] [PubMed] [Google Scholar]
  • 94.Grajeda-Iglesias C., Rom O., Aviram M. Branched-chain amino acids and atherosclerosis: friends or foes? Curr Opin Lipidol. 2018;29:166–169. doi: 10.1097/MOL.0000000000000494. [DOI] [PubMed] [Google Scholar]
  • 95.Ifrim S., Amalinei C., Cojocaru E., Matei M.C. Administration of valine, leucine, and isoleucine improved plasma cholesterol and mitigated the preatherosclerotic lesions in rats fed with hypercholesterolemic diet. Rev Romana Med Lab. 2018;26:65–75. [Google Scholar]
  • 96.Rom O., Grajeda-Iglesias C., Najjar M., Abu-Saleh N., Volkova N., Dar D.E., Hayek T., Aviram M. Atherogenicity of amino acids in the lipid-laden macrophage model system in vitro and in atherosclerotic mice: a key role for triglyceride metabolism. J Nutr Biochem. 2017;45:24–38. doi: 10.1016/j.jnutbio.2017.02.023. [DOI] [PubMed] [Google Scholar]
  • 97.Grajeda-Iglesias C., Rom O., Hamoud S., Volkova N., Hayek T., Abu-Saleh N., Aviram M. Leucine supplementation attenuates macrophage foam-cell formation: studies in humans, mice, and cultured macrophages. Biofactors. 2018;44:245–262. doi: 10.1002/biof.1415. [DOI] [PubMed] [Google Scholar]
  • 98.Yu D., Richardson N.E., Green C.L., Spicer A.B., Murphy M.E., Flores V., Jang C., Kasza I., Nikodemova M., Wakai M.H., Tomasiewicz J.L., Yang S.E., Miller B.R., Pak H.H., Brinkman J.A., Rojas J.M., Quinn W.J., 3rd, Cheng E.P., Konon E.N., Haider L.R., Finke M., Sonsalla M., Alexander C.M., Rabinowitz J.D., Baur J.A., Malecki K.C., Lamming D.W. The adverse metabolic effects of branched-chain amino acids are mediated by isoleucine and valine. Cell Metab. 2021;33:905–922.e6. doi: 10.1016/j.cmet.2021.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Wang W., Wu Z., Dai Z., Yang Y., Wang J., Wu G. Glycine metabolism in animals and humans: implications for nutrition and health. Amino Acids. 2013;45:463–477. doi: 10.1007/s00726-013-1493-1. [DOI] [PubMed] [Google Scholar]
  • 100.Meléndez-Hevia E., de Paz-Lugo P., Cornish-Bowden A., Cárdenas M.L. A weak link in metabolism: the metabolic capacity for glycine biosynthesis does not satisfy the need for collagen synthesis. J Biosci. 2009;34:853–872. doi: 10.1007/s12038-009-0100-9. [DOI] [PubMed] [Google Scholar]
  • 101.Nikiforov M.A., Chandriani S., O'Connell B., Petrenko O., Kotenko I., Beavis A., Sedivy J.M., Cole M.D. A functional screen for Myc-responsive genes reveals serine hydroxymethyltransferase, a major source of the one-carbon unit for cell metabolism. Mol Cell Biol. 2002;22:5793–5800. doi: 10.1128/MCB.22.16.5793-5800.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lamers Y., Williamson J., Gilbert L.R., Stacpoole P.W., Gregory J.F., III Glycine turnover and decarboxylation rate quantified in healthy men and women using primed, constant infusions of [1, 2-13C2] glycine and [2H3] leucine. J Nutr. 2007;137:2647–2652. doi: 10.1093/jn/137.12.2647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Kikuchi G., Motokawa Y., Yoshida T., Hiraga K. Glycine cleavage system: reaction mechanism, physiological significance, and hyperglycinemia. Proc Jpn Acad Ser B. 2008;84:246–263. doi: 10.2183/pjab/84.246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Thureen P.J., Narkewicz M.R., Battaglia F.C., Tjoa S., Fennessey P.V. Pathways of serine and glycine metabolism in primary culture of ovine fetal hepatocytes. Pediatr Res. 1995;38:775–782. doi: 10.1203/00006450-199511000-00023. [DOI] [PubMed] [Google Scholar]
  • 105.Yoshida T., Kikuchi G., Tada K., Narisawa K., Arakawa T. Physiological significance of glycine cleavage system in human liver as revealed by the study of a case of hyperglycinemia. Biochem Biophys Res Commun. 1969;35:577–583. doi: 10.1016/0006-291x(69)90387-8. [DOI] [PubMed] [Google Scholar]
  • 106.Rom O., Villacorta L., Zhang J., Chen Y.E., Aviram M. Emerging therapeutic potential of glycine in cardiometabolic diseases: dual benefits in lipid and glucose metabolism. Curr Opin Lipidol. 2018;29:428–432. doi: 10.1097/MOL.0000000000000543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Rom O., Liu Y., Liu Z., Zhao Y., Wu J., Ghrayeb A., Villacorta L., Fan Y., Chang L., Wang L. Glycine-based treatment ameliorates NAFLD by modulating fatty acid oxidation, glutathione synthesis, and the gut microbiome. Sci Transl Med. 2020;12 doi: 10.1126/scitranslmed.aaz2841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Guasch-Ferré M., Hruby A., Toledo E., Clish C.B., Martínez-González M.A., Salas-Salvadó J. Metabolomics in prediabetes and diabetes: a systematic review and meta-analysis. Diabetes Care. 2016;39:833–846. doi: 10.2337/dc15-2251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Duell P.B., Welty F.K., Miller M., Chait A., Hammond G., Ahmad Z., Cohen D.E., Horton J.D., Pressman G.S., Toth P.P. Nonalcoholic fatty liver disease and cardiovascular risk: a scientific statement from the American Heart Association. Arterioscler Thromb Vasc Biol. 2022;42:e168–e185. doi: 10.1161/ATV.0000000000000153. [DOI] [PubMed] [Google Scholar]
  • 110.Finney A.C., Das S., Kumar D., McKinney M.P., Cai B., Yurdagul A., Jr., Rom O. The interplay between nonalcoholic fatty liver disease and atherosclerotic cardiovascular disease. Front Cardiovasc Med. 2023;10:580. doi: 10.3389/fcvm.2023.1116861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Mardinoglu A., Agren R., Kampf C., Asplund A., Uhlen M., Nielsen J. Genome-scale metabolic modelling of hepatocytes reveals serine deficiency in patients with non-alcoholic fatty liver disease. Nat Commun. 2014;5:3083. doi: 10.1038/ncomms4083. [DOI] [PubMed] [Google Scholar]
  • 112.Mardinoglu A., Bjornson E., Zhang C., Klevstig M., Söderlund S., Ståhlman M., Adiels M., Hakkarainen A., Lundbom N., Kilicarslan M. Personal model-assisted identification of NAD+ and glutathione metabolism as intervention target in NAFLD. Mol Syst Biol. 2017;13:916. doi: 10.15252/msb.20167422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.White P.J., Lapworth A.L., McGarrah R.W., Kwee L.C., Crown S.B., Ilkayeva O., An J., Carson M.W., Christopher B.A., Ball J.R. Muscle-liver trafficking of BCAA-derived nitrogen underlies obesity-related glycine depletion. Cell Rep. 2020;33 doi: 10.1016/j.celrep.2020.108375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ghrayeb A., Finney A.C., Agranovich B., Peled D., Anand S.K., McKinney M.P., Sarji M., Yang D., Weissman N., Drucker S., Fernandes S.I., Fernández-García J., Mahan K., Abassi Z., Tan L., Lorenzi P.L., Traylor J., Zhang J., Abramovich I., Chen Y.E., Rom O., Mor I., Gottlieb E. Serine synthesis via reversed SHMT2 activity drives glycine depletion and acetaminophen hepatotoxicity in MASLD. Cell Metab. 2024;36:116–129.e7. doi: 10.1016/j.cmet.2023.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Tan H.C., Hsu J.W., Tai E.S., Chacko S., Wu V., Lee C.F., Kovalik J.-P., Jahoor F. De novo glycine synthesis is reduced in adults with morbid obesity and increases following bariatric surgery. Front Endocrinol. 2022;13:1135. doi: 10.3389/fendo.2022.900343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Li X., Bradford B.U., Wheeler M.D., Stimpson S.A., Pink H.M., Brodie T.A., Schwab J.H., Thurman R.G. Dietary glycine prevents peptidoglycan polysaccharide-induced reactive arthritis in the rat: role for glycine-gated chloride channel. Infect Immun. 2001;69:5883–5891. doi: 10.1128/IAI.69.9.5883-5891.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Wheeler M.D., Thurman R.G. Production of superoxide and TNF-[alpha] from alveolar macrophages is blunted by glycine. Am J Physiol Lung Cell Mol. 1999;277:L952–L959. doi: 10.1152/ajplung.1999.277.5.L952. [DOI] [PubMed] [Google Scholar]
  • 118.Ikejima K., Qu W., Stachlewitz R.F., Thurman R.G. Kupffer cells contain a glycine-gated chloride channel. Am J Physiol Gastrointest Liver Physiol. 1997;272:G1581–G1586. doi: 10.1152/ajpgi.1997.272.6.G1581. [DOI] [PubMed] [Google Scholar]
  • 119.Ruiz-Ramírez A., Ortiz-Balderas E., Cardozo-Saldaña G., Diaz-Diaz E., El-Hafidi M. Glycine restores glutathione and protects against oxidative stress in vascular tissue from sucrose-fed rats. Clin Sci. 2014;126:19–29. doi: 10.1042/CS20130164. [DOI] [PubMed] [Google Scholar]
  • 120.Caldow M.K., Ham D.J., Godeassi D.P., Chee A., Lynch G.S., Koopman R. Glycine supplementation during calorie restriction accelerates fat loss and protects against further muscle loss in obese mice. Clin Nutr. 2016;35:1118–1126. doi: 10.1016/j.clnu.2015.08.013. [DOI] [PubMed] [Google Scholar]
  • 121.Takashima S., Ikejima K., Arai K., Yokokawa J., Kon K., Yamashina S., Watanabe S. Glycine prevents metabolic steatohepatitis in diabetic KK-Ay mice through modulation of hepatic innate immunity. Am J Physiol Gastrointest Liver Physiol. 2016;311:G1105–G1113. doi: 10.1152/ajpgi.00465.2015. [DOI] [PubMed] [Google Scholar]
  • 122.Hafidi M.E., Pérez I., Zamora J., Soto V., Carvajal-Sandoval G., Banos G. Glycine intake decreases plasma free fatty acids, adipose cell size, and blood pressure in sucrose-fed rats. Am J Physiol Regul Integr Comp Physiol. 2004;287:R1387–R1393. doi: 10.1152/ajpregu.00159.2004. [DOI] [PubMed] [Google Scholar]
  • 123.Qu P., Rom O., Li K., Jia L., Gao X., Liu Z., Ding S., Zhao M., Wang H., Chen S. DT-109 ameliorates nonalcoholic steatohepatitis in nonhuman primates. Cell Metab. 2023;35:742–757.e10. doi: 10.1016/j.cmet.2023.03.013. [DOI] [PubMed] [Google Scholar]
  • 124.Cruz M., Maldonado-Bernal C., Mondragon-Gonzalez R., Sanchez-Barrera R., Wacher N., Carvajal-Sandoval G., Kumate J. Glycine treatment decreases proinflammatory cytokines and increases interferon-γ in patients with type 2 diabetes. J Endo Invest. 2008;31:694–699. doi: 10.1007/BF03346417. [DOI] [PubMed] [Google Scholar]
  • 125.Díaz-Flores M., Cruz M., Duran-Reyes G., Munguia-Miranda C., Loza-Rodríguez H., Pulido-Casas E., Torres-Ramírez N., Gaja-Rodriguez O., Kumate J., Baiza-Gutman L.A. Oral supplementation with glycine reduces oxidative stress in patients with metabolic syndrome, improving their systolic blood pressure. Can J Physiol Pharmacol. 2013;91:855–860. doi: 10.1139/cjpp-2012-0341. [DOI] [PubMed] [Google Scholar]
  • 126.Heine W., Radke M., Wutzke K.-D. The significance of tryptophan in human nutrition. Amino Acids. 1995;9:91–205. doi: 10.1007/BF00805951. [DOI] [PubMed] [Google Scholar]
  • 127.Fernstrom J.D. Effects of the diet on brain neurotransmitters. Metabolism. 1977;26:207–223. doi: 10.1016/0026-0495(77)90057-9. [DOI] [PubMed] [Google Scholar]
  • 128.Dalgliesh C. Metabolism of the aromatic amino acids. Adv Prot Chem. 1955;10:31–150. doi: 10.1016/s0065-3233(08)60104-7. [DOI] [PubMed] [Google Scholar]
  • 129.Wirleitner B., Rudzite V., Neurauter G., Murr C., Kalnins U., Erglis A., Trusinskis K., Fuchs D. Immune activation and degradation of tryptophan in coronary heart disease. Eur J Clin Invest. 2003;33:550–554. doi: 10.1046/j.1365-2362.2003.01186.x. [DOI] [PubMed] [Google Scholar]
  • 130.Gündüz M., Çakar S., Kuyum P., Makay B., Arslan N. Comparison of atherogenic risk factors among poorly controlled and well-controlled adolescent phenylketonuria patients. Cardiol Young. 2016;26:901–908. doi: 10.1017/S104795111500150X. [DOI] [PubMed] [Google Scholar]
  • 131.de Mello V.D., Sehgal R., Männistö V., Klåvus A., Nilsson E., Perfilyev A., Kaminska D., Miao Z., Pajukanta P., Ling C. Serum aromatic and branched-chain amino acids associated with NASH demonstrate divergent associations with serum lipids. Liver Int. 2021;41:754–763. doi: 10.1111/liv.14743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Xu D., Xu Y., Zhang B., Wang Y., Han L., Sun J., Sun H. Higher dietary intake of aromatic amino acids was associated with lower risk of cardiovascular disease mortality in adult participants in NHANES III. Nutr Res. 2023;113:39–48. doi: 10.1016/j.nutres.2023.03.004. [DOI] [PubMed] [Google Scholar]
  • 133.Galili G., Amir R., Fernie A.R. The regulation of essential amino acid synthesis and accumulation in plants. Annu Rev Plant Biol. 2016;67:153–178. doi: 10.1146/annurev-arplant-043015-112213. [DOI] [PubMed] [Google Scholar]
  • 134.Weinstein L., Porter C., Laurencot H. Role of the shikimic acid pathway in the formation of tryptophan in higher plants: evidence for an alternative pathway in the bean. Nature. 1962;194:205–206. [Google Scholar]
  • 135.Spraggon G., Kim C., Nguyen-Huu X., Yee M.-C., Yanofsky C., Mills S.E. The structures of anthranilate synthase of Serratia marcescens crystallized in the presence of (i) its substrates, chorismate and glutamine, and a product, glutamate, and (ii) its end-product inhibitor, L-tryptophan. Proc Natl Acad Sci U S A. 2001;98:6021–6026. doi: 10.1073/pnas.111150298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Radwanski E.R., Last R.L. Tryptophan biosynthesis and metabolism: biochemical and molecular genetics. Plant Cell. 1995;7:921. doi: 10.1105/tpc.7.7.921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Huether G., Hajak G., Reimer A., Poeggeler B., Blömer M., Rodenbeck A., Rüther E. The metabolic fate of infused L-tryptophan in men: possible clinical implications of the accumulation of circulating tryptophan and tryptophan metabolites. Psychopharmacology. 1992;109:422–432. doi: 10.1007/BF02247718. [DOI] [PubMed] [Google Scholar]
  • 138.Gál E.M., Sherman A.D. l-Kynurenine its synthesis and possible regulatory function in brain. Neurochem Res. 1980;5:223–239. doi: 10.1007/BF00964611. [DOI] [PubMed] [Google Scholar]
  • 139.O'Mahony S.M., Clarke G., Borre Y., Dinan T.G., Cryan J. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res. 2015;277:32–48. doi: 10.1016/j.bbr.2014.07.027. [DOI] [PubMed] [Google Scholar]
  • 140.Qi Q., Hua S., Clish C.B., Scott J.M., Hanna D.B., Wang T., Haberlen S.A., Shah S.J., Glesby M.J., Lazar J.M. Plasma tryptophan-kynurenine metabolites are altered in HIV infection and associated with progression of carotid artery atherosclerosis. Clin Infect Dis. 2018;67:235–242. doi: 10.1093/cid/ciy053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Pawlak K., Domaniewski T., Mysliwiec M., Pawlak D. The kynurenines are associated with oxidative stress, inflammation and the prevalence of cardiovascular disease in patients with end-stage renal disease. Atherosclerosis. 2009;204:309–314. doi: 10.1016/j.atherosclerosis.2008.08.014. [DOI] [PubMed] [Google Scholar]
  • 142.Pawlak K., Brzosko S., Mysliwiec M., Pawlak D. Kynurenine, quinolinic acid—the new factors linked to carotid atherosclerosis in patients with end-stage renal disease. Atherosclerosis. 2009;204:561–566. doi: 10.1016/j.atherosclerosis.2008.10.002. [DOI] [PubMed] [Google Scholar]
  • 143.Shimizu H., Yisireyili M., Higashiyama Y., Nishijima F., Niwa T. Indoxyl sulfate upregulates renal expression of ICAM-1 via production of ROS and activation of NF-[kappa]B and p53 in proximal tubular cells. Life Sci. 2013;92:143–148. doi: 10.1016/j.lfs.2012.11.012. [DOI] [PubMed] [Google Scholar]
  • 144.Shen W.-C., Liang C.-J., Huang T.-M., Liu C.-W., Wang S.-H., Young G.-H., Tsai J.-S., Tseng Y.-C., Peng Y.-S., Wu V.-C., Chen Y.-L. Indoxyl sulfate enhances IL-1[beta]-induced E-selectin expression in endothelial cells in acute kidney injury by the ROS/MAPKs/NF[kappa]B/AP-1 pathway. Arch Toxicol. 2016;90:2779–2792. doi: 10.1007/s00204-015-1652-0. [DOI] [PubMed] [Google Scholar]
  • 145.Six I., Gross P., Rémond M.C., Chillon J.M., Poirot S., Drueke T.B., Massy Z.A. Deleterious vascular effects of indoxyl sulfate and reversal by oral adsorbent AST-120. Atherosclerosis. 2015;243:248–256. doi: 10.1016/j.atherosclerosis.2015.09.019. [DOI] [PubMed] [Google Scholar]
  • 146.Jorgensen G., Larson B. Conversion of phenylalanine to tyrosine in the bovine mammary secretory cell. Biochim Biophys Acta Gen Subj. 1968;165:121–126. doi: 10.1016/0304-4165(68)90196-7. [DOI] [PubMed] [Google Scholar]
  • 147.Daubner S.C., Le T., Wang S. Tyrosine hydroxylase and regulation of dopamine synthesis. Arch Biochem Biophys. 2011;508:1–12. doi: 10.1016/j.abb.2010.12.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Shiman R., Gray D.W. Formation and fate of tyrosine: intracellular partitioning of newly synthesized tyrosine in mammalian liver. J Biol Chem. 1998;273:34760–34769. doi: 10.1074/jbc.273.52.34760. [DOI] [PubMed] [Google Scholar]
  • 149.Rousset B., Dupuy C., Miot F., Dumont J. Endotext [Internet] Copyright MDText.com, Inc.; 2000–2024. Chapter 2: thyroid hormone synthesis and secretion.https://www.ncbi.nlm.nih.gov/books/NBK285550 Available at: (last revised September 2, 2015) [Google Scholar]
  • 150.Stryeck S., Gastrager M., Degoricija V., Trbušić M., Potočnjak I., Radulović B., Pregartner G., Berghold A., Madl T., Frank S. Serum concentrations of citrate, tyrosine, 2- and 3- hydroxybutyrate are associated with increased 3-month mortality in acute heart failure patients. Sci Rep. 2019;9:6743. doi: 10.1038/s41598-019-42937-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Incalza M.A., D'Oria R., Natalicchio A., Perrini S., Laviola L., Giorgino F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vasc Pharm. 2018;100:1–19. doi: 10.1016/j.vph.2017.05.005. [DOI] [PubMed] [Google Scholar]
  • 152.Sgaravatti Â.M., Magnusson A.S., de Oliveira A.S., Rosa A.P., Mescka C.P., Zanin F.R., Pederzolli C.D., Wyse A.T., Wannmacher C.M., Wajner M. Tyrosine administration decreases glutathione and stimulates lipid and protein oxidation in rat cerebral cortex. Metab Brain Dis. 2009;24:415–425. doi: 10.1007/s11011-009-9153-6. [DOI] [PubMed] [Google Scholar]
  • 153.Macêdo L.G., Carvalho-Silva M., Ferreira G.K., Vieira J.S., Olegário N., Gonçalves R.C., Vuolo F.S., Ferreira G.C., Schuck P.F., Dal-Pizzol F. Effect of acute administration of L-tyrosine on oxidative stress parameters in brain of young rats. Neurochem Res. 2013;38:2625–2630. doi: 10.1007/s11064-013-1180-3. [DOI] [PubMed] [Google Scholar]
  • 154.Herrmann K.M. The shikimate pathway: early steps in the biosynthesis of aromatic compounds. Plant Cell. 1995;7:907. doi: 10.1105/tpc.7.7.907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Kaufman S. The phenylalanine hydroxylating system. Adv Enzymol Relat Areas Mol Biol. 1993;67:77–264. doi: 10.1002/9780470123133.ch2. [DOI] [PubMed] [Google Scholar]
  • 156.Delles C., Rankin N.J., Boachie C., McConnachie A., Ford I., Kangas A., Soininen P., Trompet S., Mooijaart S.P., Jukema J.W., Zannad F., Ala-Korpela M., Salomaa V., Havulinna A.S., Welsh P., Würtz P., Sattar N. Nuclear magnetic resonance-based metabolomics identifies phenylalanine as a novel predictor of incident heart failure hospitalisation: results from PROSPER and FINRISK 1997. Eur J Heart Fail. 2018;20:663–673. doi: 10.1002/ejhf.1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Murr C., Grammer T.B., Meinitzer A., Kleber M.E., März W., Fuchs D. Immune activation and inflammation in patients with cardiovascular disease are associated with higher phenylalanine to tyrosine ratios: the ludwigshafen risk and cardiovascular health study. J Amino Acids. 2014;2014 doi: 10.1155/2014/783730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Hattori Y., Hattori S., Wang X., Satoh H., Nakanishi N., Kasai K. Oral administration of tetrahydrobiopterin slows the progression of atherosclerosis in apolipoprotein E-knockout mice. Arterioscler Thromb Vasc Biol. 2007;27:865–870. doi: 10.1161/01.ATV.0000258946.55438.0e. [DOI] [PubMed] [Google Scholar]
  • 159.Nemet I., Saha P.P., Gupta N., Zhu W., Romano K.A., Skye S.M., Cajka T., Mohan M.L., Li L., Wu Y., Funabashi M., Ramer-Tait A.E., Naga Prasad S.V., Fiehn O., Rey F.E., Tang W.H.W., Fischbach M.A., DiDonato J.A., Hazen S.L. A cardiovascular disease-linked gut microbial metabolite acts via adrenergic receptors. Cell. 2020;180:862–877.e22. doi: 10.1016/j.cell.2020.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Steinbrenner H., Bilgic E., Alili L., Sies H., Brenneisen P. Selenoprotein P protects endothelial cells from oxidative damage by stimulation of glutathione peroxidase expression and activity. Free Radic Res. 2006;40:936–943. doi: 10.1080/10715760600806248. [DOI] [PubMed] [Google Scholar]
  • 161.Maiorino M., Conrad M., Ursini F. GPx4, lipid peroxidation, and cell death: discoveries, rediscoveries, and open issues. Antioxid Redox Signal. 2018;29:61–74. doi: 10.1089/ars.2017.7115. [DOI] [PubMed] [Google Scholar]
  • 162.Torzewski M., Ochsenhirt V., Kleschyov A.L., Oelze M., Daiber A., Li H., Rossmann H., Tsimikas S., Reifenberg K., Cheng F., Lehr H.A., Blankenberg S., Forstermann U., Munzel T., Lackner K.J. Deficiency of glutathione peroxidase-1 accelerates the progression of atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2007;27:850–857. doi: 10.1161/01.ATV.0000258809.47285.07. [DOI] [PubMed] [Google Scholar]
  • 163.Guo Z., Ran Q., Roberts L.J., 2nd, Zhou L., Richardson A., Sharan C., Wu D., Yang H. Suppression of atherogenesis by overexpression of glutathione peroxidase-4 in apolipoprotein E-deficient mice. Free Radic Biol Med. 2008;44:343–352. doi: 10.1016/j.freeradbiomed.2007.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Schwenke D.C., Behr S.R. Vitamin E combined with selenium inhibits atherosclerosis in hypercholesterolemic rabbits independently of effects on plasma cholesterol concentrations. Circ Res. 1998;83:366–377. doi: 10.1161/01.res.83.4.366. [DOI] [PubMed] [Google Scholar]
  • 165.Flores-Mateo G., Navas-Acien A., Pastor-Barriuso R., Guallar E. Selenium and coronary heart disease: a meta-analysis. Am J Clin Nutr. 2006;84:762–773. doi: 10.1093/ajcn/84.4.762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Zhang X., Liu C., Guo J., Song Y. Selenium status and cardiovascular diseases: meta-analysis of prospective observational studies and randomized controlled trials. Eur J Clin Nutr. 2016;70:162–169. doi: 10.1038/ejcn.2015.78. [DOI] [PubMed] [Google Scholar]
  • 167.Xiang S., Dai Z., Man C., Fan Y. Circulating selenium and cardiovascular or all-cause mortality in the general population: a meta-analysis. Biol Trace Elem Res. 2020;195:55–62. doi: 10.1007/s12011-019-01847-8. [DOI] [PubMed] [Google Scholar]
  • 168.Ju W., Li X., Li Z., Wu G.R., Fu X.F., Yang X.M., Zhang X.Q., Gao X.B. The effect of selenium supplementation on coronary heart disease: a systematic review and meta-analysis of randomized controlled trials. J Trace Elem Med Biol. 2017;44:8–16. doi: 10.1016/j.jtemb.2017.04.009. [DOI] [PubMed] [Google Scholar]
  • 169.Rayman M.P., Winther K.H., Pastor-Barriuso R., Cold F., Thvilum M., Stranges S., Guallar E., Cold S. Effect of long-term selenium supplementation on mortality: results from a multiple-dose, randomised controlled trial. Free Radic Biol Med. 2018;127:46–54. doi: 10.1016/j.freeradbiomed.2018.02.015. [DOI] [PubMed] [Google Scholar]
  • 170.Hitzel J., Lee E., Zhang Y., Bibli S.I., Li X., Zukunft S., Pfluger B., Hu J., Schurmann C., Vasconez A.E., Oo J.A., Kratzer A., Kumar S., Rezende F., Josipovic I., Thomas D., Giral H., Schreiber Y., Geisslinger G., Fork C., Yang X., Sigala F., Romanoski C.E., Kroll J., Jo H., Landmesser U., Lusis A.J., Namgaladze D., Fleming I., Leisegang M.S., Zhu J., Brandes R.P. Oxidized phospholipids regulate amino acid metabolism through MTHFD2 to facilitate nucleotide release in endothelial cells. Nat Commun. 2018;9:2292. doi: 10.1038/s41467-018-04602-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Seeley E.H., Liu Z., Yuan S., Stroope C., Cockerham E., Rashdan N.A., Delgadillo L.F., Finney A.C., Kumar D., Das S., Razani B., Liu W., Traylor J., Orr A.W., Rom O., Pattillo C.B., Yurdagul A., Jr. Spatially resolved metabolites in stable and unstable human atherosclerotic plaques identified by mass spectrometry imaging. Arterioscler Thromb Vasc Biol. 2023;43:1626–1635. doi: 10.1161/ATVBAHA.122.318684. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The American Journal of Pathology are provided here courtesy of American Society for Investigative Pathology

RESOURCES