Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Int J Mol Sci ; 24(19)2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37834229

ABSTRACT

A restoration of low homoarginine (hArg) levels in obese ZSF1 rats (O-ZSF1) before (S1-ZSF1) and after (S2-ZSF1) the manifestation of heart failure with preserved ejection fraction (HFpEF) did not affect the worsening of cardiac HFpEF characteristics. Here, potential regulation of key enzymes of arginine metabolism in other organs was analyzed. Arginase 2 (ARG2) was reduced >35% in the kidney and small intestine of hArg-supplemented rats compared to O-ZSF1. Glycine amidinotransferase (GATM) was 29% upregulated in the kidneys of S1-ZSF1. Dimethylarginine dimethylaminohydrolase 1 (DDAH1) levels were reduced >50% in the livers of O-ZSF1 but restored in S2-ZSF1 compared to healthy rats (L-ZSF1). In the skeletal muscle, iNOS was lower in O-ZSF1 and further decreased in S1-ZSF1 and S2-ZSF1 compared to L-ZSF1. iNOS levels were lower in the liver of the S2-ZSF1 group but higher in the kidneys of S1-ZSF1 compared to L-ZSF1. Supplementation with hArg in an in vivo HFpEF model resulted in the inhibition of renal ARG2 and an increase in GATM expression. This supplementation might contribute to the stabilization of intestinal iNOS and ARG2 imbalances, thereby enhancing barrier function. Additionally, it may offer protective effects in skeletal muscle by downregulating iNOS. In the conceptualization of hArg supplementation studies, the current disease progression stage as well as organ-specific enzyme regulation should be considered.


Subject(s)
Heart Failure , Rats , Animals , Heart Failure/drug therapy , Homoarginine/metabolism , Arginine/metabolism , Stroke Volume/physiology , Dietary Supplements
2.
Curr Opin Clin Nutr Metab Care ; 26(1): 42-49, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36330857

ABSTRACT

PURPOSE OF REVIEW: Homoarginine (hArg) is an endogenous, nonproteinogenic amino acid. It is enzymatically synthesized from L-arginine and L-lysine. Low hArg concentrations appear to be a risk factor in the renal and cardiovascular systems. This review discusses advances in-vitro and in-vivo experimental and clinical research on hArg in health and disease. RECENT FINDINGS: Recent studies indicate that low circulating and low urinary concentrations of hArg are associated with morbidity and worse outcome. Although the biological activities of hArg remain still unexplored, hArg supplementation is intensely investigated as a strategy to increase hArg concentration to reach normal levels in cases of low hArg concentrations. The greatest changes in circulating hArg concentrations are observed during pregnancy and after delivery. In healthy adults, a daily dose of 125 mg hArg seems to be optimum to normalize circulating levels. Short-term supplementation of inorganic nitrate enhances hArg biosynthesis in healthy young men. Apart from hArg supplementation, dietary L-arginine and L-citrulline appear to be a promising alternative. SUMMARY: Considerable progress has been made in recent years, but hArg remains still enigmatic. Further research is required to explore the biological activities of hArg. Supplementation of hArg or its precursors L-citrulline/L-arginine seem to be promising strategies to prevent and overcome altered hArg synthesis.


Subject(s)
Cardiovascular System , Homoarginine , Male , Adult , Humans , Homoarginine/metabolism , Citrulline , Arginine/metabolism , Cardiovascular System/metabolism , Kidney/metabolism
3.
Chem Commun (Camb) ; 58(86): 12054-12057, 2022 Oct 27.
Article in English | MEDLINE | ID: mdl-36193595

ABSTRACT

Cyanobactins are linear and cyclic post-translationally modified peptides. Here we show that the prenyl-D-Arg-containing autumnalamide A is a member of the cyanobactin family. Biochemical assays demonstrate that the AutF prenyltransferase targets the guanidinium moiety in arginine and homoarginine and is a useful tool for biotechnological applications.


Subject(s)
Biosynthetic Pathways , Dimethylallyltranstransferase , Dimethylallyltranstransferase/chemistry , Dimethylallyltranstransferase/metabolism , Arginine/metabolism , Homoarginine/metabolism , Guanidine , Peptides, Cyclic/chemistry
4.
Int J Mol Sci ; 23(18)2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36142560

ABSTRACT

The aim of the study was to investigate the effects of short-term oral administration of inorganic nitrate (NaNO3; n = 8) or placebo (NaCl; n = 9) (each 0.1 mmol/kg body weight/d for 9 days) on plasma amino acids, creatinine, and oxidative stress in healthy young men. At baseline, the plasma concentrations of amino acids did not differ between the groups. At the end of the study, the plasma concentrations of homoarginine (hArg; by 24%, p = 0.0001), citrulline and ornithine (Cit/Orn; by 16%, p = 0.015), and glutamine/glutamate (Gln/Glu; by 6%, p = 0.0003) were higher in the NaNO3 group compared to the NaCl group. The plasma concentrations of sarcosine (Sarc; by 28%, p < 0.0001), tyrosine (by 14%, p = 0.0051), phenylalanine (by 8%, p = 0.0026), and tryptophan (by 8%, p = 0.0047) were lower in the NaNO3 group compared to the NaCl group. These results suggest that nitrate administration affects amino-acid metabolism. The arginine/glycine amidinotransferase (AGAT) catalyzes two reactions: (1) the formation of l-homoarginine (hArg) and l-ornithine (Orn) from l-arginine (Arg) and l-lysine (Lys): Arg + Lys <−> hArg + Orn, with equilibrium constant Kharg; (2) the formation of guanidinoacetate (GAA) and Orn from Arg and glycine (Gly): Arg + Gly <−> GAA + Orn, with equilibrium constant Kgaa. The plasma Kgaa/KhArg ratio was lower in the NaNO3 group compared to the NaCl group (1.57 vs. 2.02, p = 0.0034). Our study suggests that supplementation of inorganic nitrate increases the AGAT-catalyzed synthesis of hArg and decreases the N-methyltransferase-catalyzed synthesis of GAA, the precursor of creatine. To our knowledge, this is the first study to demonstrate elevation of hArg synthesis by inorganic nitrate supplementation. Remarkably, an increase of 24% corresponds to the synthesis capacity of one kidney in healthy humans. Differences in the association between plasma concentrations of amino acids in the NaNO3 and NaCl groups suggest changes in amino-acid homeostasis. Plasma concentrations of the oxidative stress marker malondialdehyde (MDA) did not change after supplementation of NaNO3 or NaCl over the whole exercise time range. Plasma nitrite concentration turned out to be a more discriminant marker of NaNO3 ingestion than plasma nitrate (area under the receiver operating characteristic curve: 0.951 vs. 0.866, p < 0.0001 each).


Subject(s)
Homoarginine , Nitrates , Arginine/metabolism , Citrulline , Creatine , Creatinine , Dietary Supplements , Glutamates , Glutamine , Glycine , Homoarginine/metabolism , Humans , Lysine , Male , Malondialdehyde , Methyltransferases , Nitrites , Ornithine , Phenylalanine , Sarcosine , Sodium Chloride , Tryptophan , Tyrosine
5.
Biomark Med ; 16(13): 993-1004, 2022 09.
Article in English | MEDLINE | ID: mdl-36052727

ABSTRACT

Aim: The aim of this study was to measure serum levels of methylarginine derivatives and related metabolites in patients with gout. Materials & methods: This study enrolled 100 patients with gout and 80 patients in the control group. Serum asymmetric dimethylarginine, symmetric dimethylarginine, L-N-monomethylarginine, arginine, homoarginine, citrulline and ornithine levels were measured with tandem mass spectrometry. Results: Serum ornithine, citrulline and total methylated arginine load levels were statistically significantly higher in patients with gout compared with the control group, while serum arginine and homoarginine levels and global arginine bioavailability ratio were statistically significantly lower. Conclusion: There may be an association between gout, methylarginine levels and hyperuricemia and increased risk of cardiovascular disease.


Subject(s)
Gout , Homoarginine , Arginine/metabolism , Citrulline/metabolism , Homoarginine/metabolism , Humans , Nitric Oxide/metabolism , Ornithine
6.
Thromb Res ; 217: 96-103, 2022 09.
Article in English | MEDLINE | ID: mdl-35926348

ABSTRACT

Eptifibatide is an αIIbß3 inhibitor that is currently used in the clinic. More than 10 scientific communications indicate that eptifibatide has a Lys-Gly-Asp or Arg-Gly-Asp sequence, while it actually has a hArg-Gly-Asp sequence. We aimed to unravel the importance of the homoarginine residue in eptifibatide in platelet activation and aggregation. Arg- and Lys-eptifibatide were synthesized by solid-phase peptide synthesis and measured in light transmission aggregometry, flow cytometry and whole blood thrombus formation under flow. Interactions of eptifibatide and its variants with αIIbß3 integrin were studied using molecular dynamics simulations. Eptifibatide showed inhibition of collagen- and ADP-induced platelet aggregation, while Arg- and Lys-eptifibatide did not. Multiparameter assessment of thrombus formation showed suppressed platelet aggregate and fibrin formation upon eptifibatide treatment, in contrast to the other variants. Molecular dynamics simulations revealed that the hArg residue in eptifibatide is crucial to its activity, since the substitution of the hArg to Arg or Lys resulted in the inability to form double H-bonds with Asp224 in the αIIb chain of the αIIbß3 receptor. The hArg is pivotal for the interaction of eptifibatide for the αIIbß3 receptor and efficient inhibition of platelet aggregation.


Subject(s)
Platelet Aggregation Inhibitors , Thrombosis , Blood Platelets/metabolism , Eptifibatide/pharmacology , Homoarginine/metabolism , Homoarginine/pharmacology , Humans , Peptides/metabolism , Peptides/pharmacology , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombosis/drug therapy , Thrombosis/metabolism
7.
Fundam Clin Pharmacol ; 36(6): 992-1004, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35697514

ABSTRACT

Low serum concentrations of the amino acid homoarginine (HA) are associated with increased cardiovascular mortality by incompletely understood mechanisms. This study sought to assess the influence of HA on cardiac remodeling in rats undergoing either transaortic banding or inhibition of nitric oxide synthesis by Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME). Male Wistar rats (n = 136) underwent sham operation (SH) or aortic banding (AB). Both groups were equally divided into 14 subgroups, receiving different doses of HA alone or in combination with lisinopril, spironolactone, or L-NAME for 4 weeks. HA treatment in AB animals resulted in a dose-dependent improvement of cardiac function up to a concentration of 800 mg·kg-1 ·day-1 . Combining 800 mg·kg-1 ·day-1 HA with spironolactone or lisinopril yielded additional effects, showing a positive correlation with LV ejection fraction (+33%, p = 0.0002) and fractional shortening (+41%, p = 0.0014). An inverse association was observed with collagen area fraction (-41%, p < 0.0001), myocyte cross-sectional area (-22%, p < 0.0001) and the molecular markers atrial natriuretic factor (-74%, p = 0.0091), brain natriuretic peptide (-42%, p = 0.0298), beta-myosin heavy chain (-46%, p = 0.0411), and collagen type V alpha 1 chain (-73%, p = 0.0257) compared to placebo-treated AB animals. Co-administration of HA and L-NAME was found to attenuate cardiac remodeling and prevent NO-deficient hypertension following AB. HA treatment has led to a dose-dependent improvement of myocardial function and marked histological and molecular changes in cardiac remodeling following AB. Combining HA with standard heart failure medication resulted in additional beneficial effects boosting its direct impact on heart failure pathophysiology.


Subject(s)
Heart Failure , Hypertension , Rats , Male , Animals , NG-Nitroarginine Methyl Ester/pharmacology , Spironolactone/metabolism , Spironolactone/pharmacology , Spironolactone/therapeutic use , Homoarginine/metabolism , Homoarginine/pharmacology , Homoarginine/therapeutic use , Lisinopril/metabolism , Lisinopril/pharmacology , Lisinopril/therapeutic use , Ventricular Remodeling , Hypertension/drug therapy , Rats, Wistar , Myocardium/metabolism , Heart Failure/drug therapy , Blood Pressure
8.
Int J Mol Sci ; 23(9)2022 Apr 25.
Article in English | MEDLINE | ID: mdl-35563125

ABSTRACT

L-Arginine:glycine amidinotransferase (AGAT) catalyzes the formation of L-homoarginine (hArg) and L-ornithine (Orn) from L-arginine (Arg) and L-lysine (Lys): Arg + Lys ↔ hArg + Orn; equilibrium constant KhArg. AGAT also catalyzes the formation of guanidinoacetate (GAA) and Orn from Arg and glycine (Gly): Arg + Gly ↔ GAA + Orn; equilibrium constant KGAA. In humans, pharmacological hArg is metabolized to Lys. Low circulating and low excretory concentrations of hArg are associated with worse outcomes and mortality in the renal and cardiovascular systems. The metabolism and pharmacology of hArg have been little investigated. In the present study, we investigated the effects of pharmacological hArg (i.p., 0, 20, 220, 440 mg/kg at time point 0 min) on amino acids homeostasis in a rat model of isoprenaline-induced takotsubo cardiomyopathy (i.p., 50 mg/kg at time point 15 min). We measured by gas chromatography-mass spectrometry free and proteinic amino acids, as well as the polyamines putrescine and spermidine in the heart, lung, kidney, and liver of ten rats sacrificed at various time points (range, 0 to 126 min). hArg administration resulted in multiple changes in the tissue contents of several free and proteinic amino acids, as well as in the putrescine-spermidine molar ratio, an indicator of polyamines catabolism. Our results suggest that Lys and Arg are major metabolites of pharmacological hArg. Kidneys and heart seem to play a major metabolic role for hArg. Circulating Lys does not change over time, yet there is a considerable interchange of free Lys between organs, notably kidney and heart, during the presence of isoprenaline in the rats (time range, 15 to 90 min). Antidromic changes were observed for KhArg and KGAA, notably in the heart in this time window. Our study shows for the first time that free hArg and sarcosine (N-methylglycine) are positively associated with each other. The acute effects of high-dosed hArg administration and isoprenaline on various amino acids and on AGAT-catalyzed reaction in the heart, lung, kidney, and liver are detailed and discussed.


Subject(s)
Homoarginine , Takotsubo Cardiomyopathy , Amino Acids , Animals , Arginine/metabolism , Homeostasis , Homoarginine/metabolism , Isoproterenol , Lysine , Pilot Projects , Putrescine , Rats , Spermidine
9.
Nitric Oxide ; 122-123: 47-53, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35367633

ABSTRACT

BACKGROUND: Arginine metabolites are associated with cardiovascular and all-cause mortality in several patient groups. We investigated whether arginine metabolites are associated with mortality in patients with critical illness and whether associations are independent of other factors affecting prognosis in an Intensive Care Unit (ICU). METHODS: 1155 acutely unwell adult patients admitted to a mixed medical-surgical ICU were studied. Arginine, asymmetric dimethyl-l-arginine (ADMA), monomethyl-l-arginine (MMA), symmetric dimethyl-l-arginine (SDMA) and l-homoarginine were measured in a plasma sample collected at admission to ICU by liquid chromatography tandem mass spectrometry. Risk of death score was calculated using data submitted to the Australia and New Zealand Intensive Care Society. RESULTS: In this cohort, 163 patients (14.1%) died. ADMA (odds ratio = 1.159 (1.033-1.300) per 0.1 µmol/L increment, p = 0.012), homoarginine (odds ratio = 0.963 (0.934-0.992), p = 0.013) and risk of death score (odds ratio = 1.045 (1.037-1.053) per 1% increment, p < 0.001) were independently associated with mortality in ICU patients. The area under the receiver operator characteristic curve for risk of death score, ADMA and homoarginine combined for mortality was greater than for risk of death score alone (0.815 (95% CI 0.790-0.837) vs 0.796 (95% CI 0.781-0.820), p = 0.019). Other arginine metabolites were not independently associated with mortality. CONCLUSIONS: ADMA is positively and homoarginine negatively associated with mortality in ICU patients, independent of other clinical factors. Measuring ADMA and homoarginine may refine models to predict ICU mortality. Reducing ADMA and increasing homoarginine are potential therapeutic targets to reduce mortality in critically ill patients.


Subject(s)
Cardiovascular System , Homoarginine , Adult , Arginine/metabolism , Biomarkers/metabolism , Cardiovascular System/metabolism , Cohort Studies , Critical Illness , Homoarginine/metabolism , Humans
10.
Amino Acids ; 54(7): 1101-1108, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35377022

ABSTRACT

The cationic amino acid transporter 1 (CAT1/SLC7A1) plays a key role in the cellular uptake or export of L-arginine and some of its derivatives. This study investigated the effect of 113 chemically diverse and commonly used drugs (at 20 and 200 µM) on the CAT1-mediated cellular uptake of L-arginine, L-homoarginine, and asymmetric dimethylarginine (ADMA). Twenty-three (20%) of the tested substances showed weak inhibitory or stimulatory effects, but only verapamil showed consistent inhibitory effects on CAT1-mediated transport of all tested substrates.


Subject(s)
Arginine , Cationic Amino Acid Transporter 1 , Biological Transport , Cationic Amino Acid Transporter 1/genetics , Cationic Amino Acid Transporter 1/metabolism , Homoarginine/metabolism
11.
Sci Rep ; 12(1): 5108, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35332188

ABSTRACT

In humans and mice, L-arginine:glycine amidinotransferase (AGAT) and its metabolites homoarginine (hArg) and creatine have been linked to cardiovascular disease (CVD), specifically myocardial infarction (MI) and heart failure (HF). The underlying molecular and regulatory mechanisms, however, remain unclear. To identify potential pathways of cardiac AGAT metabolism, we sequenced microRNA (miRNA) in left ventricles of wild-type (wt) compared to AGAT-deficient (AGAT-/-) mice. Using literature search and validation by qPCR, we identified eight significantly regulated miRNAs in AGAT-/- mice linked to atherosclerosis, MI and HF: miR-30b, miR-31, miR-130a, miR-135a, miR-148a, miR-204, miR-298, and let-7i. Analysis of Gene Expression Omnibus (GEO) data confirmed deregulation of these miRNAs in mouse models of MI and HF. Quantification of miRNA expression by qPCR in AGAT-/- mice supplemented with creatine or hArg revealed that miR-30b, miR-31, miR-130a, miR-148a, and miR-204 were regulated by creatine, while miR-135a and miR-298 showed a trend of regulation by hArg. Finally, bioinformatics-based target prediction showed that numerous AGAT-dependent genes previously linked to CVD are likely to be regulated by the identified miRNAs. Taken together, AGAT deficiency and hArg/creatine supplementation are associated with cardiac miRNA expression which may influence cardiac (dys)function and CVD.


Subject(s)
Heart Failure , MicroRNAs , Myocardial Infarction , Amidinotransferases , Animals , Arginine/metabolism , Creatine/metabolism , Homoarginine/metabolism , Mice , MicroRNAs/genetics , Myocardial Infarction/genetics
12.
Int J Mol Sci ; 23(4)2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35216250

ABSTRACT

INTRODUCTION: L-Arginine (Arg) is a semi-essential amino acid. Constitutive and inducible nitric oxide synthase (NOS) isoforms convert Arg to nitric oxide (NO), a potent vaso- and bronchodilator with multiple biological functions. Atopic dermatitis (AD) and bronchial asthma (BA) are atopic diseases affecting many children globally. Several studies analyzed NO in airways, yet the systemic synthesis of NO in AD and BA in children with BA, AD or both is elusive. METHODS: In a multicenter study, blood and urine were obtained from 130 of 302 participating children for the measurement of metabolites of the Arg/NO pathway (BA 31.5%; AD 5.4%; AD + BA 36.1%; attention deficit hyperactivity disorder (ADHD) 12.3%). In plasma and urine amino acids Arg and homoarginine (hArg), both substrates of NOS, asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA), both inhibitors of NOS, dimethylamine (DMA), and nitrite and nitrate, were measured by gas chromatography-mass spectrometry. Malondialdehyde (MDA) was measured in plasma and urine samples to evaluate possible effects of oxidative stress. RESULTS: There were no differences in the Arg/NO pathway between the groups of children with different atopic diseases. In comparison to children with ADHD, children with AD, BA or AD and BA had higher plasma nitrite (p < 0.001) and nitrate (p < 0.001) concentrations, suggesting higher systemic NO synthesis in AD and BA. Urinary excretion of DMA was also higher (p = 0.028) in AD and BA compared to patients with ADHD, suggesting elevated ADMA metabolization. DISCUSSION/CONCLUSION: The Arg/NO pathway is activated in atopic diseases independent of severity. Systemic NO synthesis is increased in children with an atopic disease. Plasma and urinary MDA levels did not differ between the groups, suggesting no effect of oxidative stress on the Arg/NO pathway in atopic diseases.


Subject(s)
Arginine/metabolism , Dermatitis, Atopic/metabolism , Nitric Oxide/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology , Arginine/analogs & derivatives , Arginine/blood , Asthma/blood , Asthma/metabolism , Child , Dermatitis, Atopic/blood , Female , Homoarginine/blood , Homoarginine/metabolism , Humans , Male , Malondialdehyde/blood , Malondialdehyde/metabolism , Nitrates/blood , Nitrates/metabolism , Nitric Oxide/blood , Nitrites/blood , Nitrites/metabolism
13.
Res Vet Sci ; 144: 175-180, 2022 May.
Article in English | MEDLINE | ID: mdl-34823870

ABSTRACT

A striking increase in homoarginine concentrations, about more than 100-fold that observed in humans, was recently reported during pregnancy in a nutritionally induced model of intra-uterine growth restriction in ewes. To determine whether this phenomenon is at least partially related to the nutritional regimen, estrus synchronization, or analytical method, thirty-four one-year-old primiparous, non-synchronized, and well-fed Sarda breed ewes were exposed to fertile rams allowing those who came into estrus to naturally mate. Plasma arginine, homoarginine, asymmetric dimethylarginine, symmetric dimethylarginine, mono methylarginine, and citrulline concentrations were measured in each sample using LC-MS/MS. Homoarginine concentrations showed a 44-fold variation between the highest and the lowest values while the fluctuations of arginine and its analogues and metabolites were much smaller, between 1.1 and 1.6-fold. Repeated-measures correlation analysis showed a significant negative correlation between homoarginine/arginine and arginine/asymmetric dimethylarginine ratios (Rm = -0.40; P < 0.000001). Furthermore, median homoarginine concentrations significantly increased with the number of fetuses. The marked increase in homoarginine concentrations with advancing gestational age is genuine and independent of mating, feeding, diet, and hormone treatment. The higher homoarginine concentrations found in ewes bearing multiple fetuses suggest the presence of a physiological link between this arginine analog and energy metabolism in pregnancy that warrants further investigation.


Subject(s)
Homoarginine , Tandem Mass Spectrometry , Animals , Chromatography, Liquid/veterinary , Female , Fetus/metabolism , Gestational Age , Homoarginine/metabolism , Male , Pregnancy , Sheep , Tandem Mass Spectrometry/veterinary
14.
Physiol Rep ; 9(5): e14766, 2021 03.
Article in English | MEDLINE | ID: mdl-33713581

ABSTRACT

Recently we showed that homoarginine supplementation confers kidney protection in diabetic mouse models. In this study we tested whether the protective effect of homoarginine is nitric oxide synthase-3 (NOS3)-independent in diabetic nephropathy (DN). Experiments were conducted in NOS3 deficient (NOS3-/- ) mice and their wild type littermate using multiple low doses of vehicle or streptozotocin and treated with homoarginine via drinking water for 24 weeks. Homoarginine supplementation for 24 weeks in diabetic NOS3-/- mice significantly attenuated albuminuria, increased blood urea nitrogen, histopathological changes and kidney fibrosis, kidney fibrotic markers, and kidney macrophage recruitment compared with vehicle-treated diabetic NOS3-/- mice. Furthermore, homoarginine supplementation restored kidney mitochondrial function following diabetes. Importantly, there were no significant changes in kidney NOS1 or NOS2 mRNA expression between all groups. In addition, homoarginine supplementation improved cardiac function and reduced cardiac fibrosis following diabetes. These data demonstrate that the protective effect of homoarginine is independent of NOS3, which will ultimately change our understanding of the mechanism(s) by which homoarginine induce renal and cardiac protection in DN. Homoarginine protective effect in DN could be mediated via improving mitochondrial function.


Subject(s)
Diabetic Nephropathies/drug therapy , Homoarginine/pharmacology , Nitric Oxide Synthase Type III/drug effects , Oxidative Stress/drug effects , Streptozocin/pharmacology , Albuminuria/metabolism , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Homoarginine/metabolism , Kidney/drug effects , Kidney/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout
15.
Circ Heart Fail ; 14(1): e007275, 2021 01.
Article in English | MEDLINE | ID: mdl-33464957

ABSTRACT

BACKGROUND: Heart failure (HF) is a heterogeneous disease characterized by significant metabolic disturbances; however, the breadth of metabolic dysfunction before the onset of overt disease is not well understood. The purpose of this study was to determine the association of circulating metabolites with incident HF to uncover novel metabolic pathways to disease. METHODS: We performed targeted plasma metabolomic profiling in a deeply phenotyped group of Black adults from the JHS (Jackson Heart Study; n=2199). We related metabolites associated with incident HF to established etiological mechanisms, including increased left ventricular mass index and incident coronary heart disease. Furthermore, we evaluated differential associations of metabolites with HF with preserved ejection fraction versus HF with reduced ejection fraction. RESULTS: Metabolites associated with incident HF included products of posttranscriptional modifications of RNA, as well as polyamine and nitric oxide metabolism. A subset of metabolite-HF associations was independent of well-established HF pathways such as increased left ventricular mass index and incident coronary heart disease and included homoarginine (per 1 SD increase in metabolite level, hazard ratio, 0.77; P=1.2×10-3), diacetylspermine (hazard ratio, 1.34; P=3.4×10-3), and uridine (hazard ratio, 0.79; P, 3×10-4). Furthermore, metabolites involved in pyrimidine metabolism (orotic acid) and collagen turnover (N-methylproline) among others were part of a distinct metabolic signature that differentiated individuals with HF with preserved ejection fraction versus HF with reduced ejection fraction. CONCLUSIONS: The integration of clinical phenotyping with plasma metabolomic profiling uncovered novel metabolic processes in nontraditional disease pathways underlying the heterogeneity of HF development in Black adults.


Subject(s)
Black or African American , Coronary Disease/metabolism , Heart Failure/metabolism , Hypertrophy, Left Ventricular/metabolism , Metabolomics , Adult , Aged , Case-Control Studies , Collagen/metabolism , Coronary Disease/epidemiology , Effect Modifier, Epidemiologic , Female , Heart Disease Risk Factors , Heart Failure/epidemiology , Heart Failure/physiopathology , Homoarginine/metabolism , Humans , Hypertrophy, Left Ventricular/epidemiology , Incidence , Longitudinal Studies , Male , Middle Aged , Nitric Oxide/metabolism , Orotic Acid/metabolism , Polyamines/metabolism , Proline/analogs & derivatives , Proline/metabolism , Proportional Hazards Models , Pyrimidines/metabolism , RNA Processing, Post-Transcriptional , Risk , Spermine/analogs & derivatives , Spermine/metabolism , Stroke Volume/physiology , Uridine/metabolism , White People
16.
Amino Acids ; 52(6-7): 975-985, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32642843

ABSTRACT

Elevated plasma concentrations of the uremic toxin asymmetric dimethylarginine (ADMA) and low plasma concentrations of L-homoarginine are independently associated with cardiovascular events and mortality. Key enzymes involved in the homeostasis of both arginine derivatives are expressed in proximal tubule cells of the kidney. To get access to these enzymes, transport proteins are important. One of the transporters mediating the transport of ADMA and L-homoarginine is the solute carrier superfamily (SLC) member OATP4C1, located in the basolateral membrane of proximal tubule cells. To gain insights into the role of export pumps in the transport of both substances, we established a double-transfected MDCK cell line expressing OATP4C1 and the export pump P-glycoprotein (P-gp). Using MDCK cell monolayers, we demonstrated in time-dependent and concentration-dependent vectorial transport experiments that ADMA and L-homoarginine are transported from the basolateral to the apical compartment of MDCK-OATP4C1-P-gp cells with significantly higher transport rates compared to single-transfected MDCK-OATP4C1, MDCK-P-gp and MDCK-VC (control) cells (e.g. transport ratio MDCK-OATP4C1-P-gp/MDCK-VC: for 50 µM ADMA = 2.0-fold, for 50 µM L-homoarginine = 3.4-fold). These results indicate that both OATP4C1 and P-gp transport the arginine derivatives ADMA and L-homoarginine and are, therefore, important for the homoeostasis of both substances.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Arginine/analogs & derivatives , Homoarginine/metabolism , Organic Anion Transporters/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Arginine/metabolism , Dogs , Humans , Madin Darby Canine Kidney Cells , Organic Anion Transporters/genetics , Transcytosis , Transfection
17.
Amino Acids ; 52(6-7): 965-973, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32613307

ABSTRACT

BACKGROUND AND AIMS: The L-arginine derivatives asymmetric (ADMA) and symmetric dimethylarginine (SDMA), as well as L-homoarginine may have opposing effects in the pathogenesis of atherosclerosis. We aimed to investigate (i) 5-year changes in arginine derivatives, and (ii) the association between baseline arginine derivatives and follow-up measures of carotid wall thickness in South Africans. METHODS AND RESULTS: This study included men (n = 187) and women (n = 396) who took part in the 2010 and 2015 data collections of the South African arm of the Prospective Urban and Rural Epidemiology (PURE) study. Arginine derivatives were determined in plasma with liquid chromatography-tandem mass spectrometry. Carotid intima-media thickness (cIMT) and cross-sectional wall area (CSWA) were determined with B-mode ultrasonography. RESULTS: Mean values of arginine derivatives did not change over time. In the study group, follow-up cIMT (ß = - 0.10 p = 0.018) and CSWA (ß = - 0.12; p = 0.004) inversely associated with baseline L-homoarginine, and cIMT inversely associated with ADMA (ß = - 0.09; p = 0.033). In women, CSWA inversely associated with both ADMA (ß = - 0.11; p = 0.034) and L-homoarginine (ß = - 0.11; p = 0.024). No such associations were found in men. CONCLUSION: These results suggest that higher levels of L-homoarginine may play a protective role against vascular injury and delay progression of carotid wall thickening in this cohort. The role of ADMA in atherosclerosis deserves further investigation in this population.


Subject(s)
Arginine/analogs & derivatives , Carotid Intima-Media Thickness , Homoarginine/metabolism , Aged , Arginine/metabolism , Black People , Blood Pressure , Cohort Studies , Female , Humans , Male , Middle Aged , Nitric Oxide/metabolism , Prospective Studies , Ultrasonography
18.
J Mol Med (Berl) ; 98(7): 999-1008, 2020 07.
Article in English | MEDLINE | ID: mdl-32504111

ABSTRACT

Arginine, homoarginine (hArg), asymmetric dimethylarginine (ADMA), and symmetric dimethylarginine (SDMA) affect nitric oxide metabolism and altered concentrations are associated with cardiovascular morbidity and mortality. We analyzed these metabolites using liquid chromatography-tandem mass spectrometry in patients with atrial fibrillation (AF) (n = 241) with a focus on heart rhythm at blood withdrawal, AF progression phenotypes, and successful sinus rhythm (SR) restoration (n = 22). AF progression phenotypes were defined as paroxysmal AF with/without low voltage areas (LVA) and persistent AF with/without LVA. While arginine, ADMA, and hArg were within reference limits for healthy controls, SDMA was higher in the AF cohort (0.57 ± 0.12 vs. 0.53 µmol/L (97.5th percentile in reference cohort)). SR restoration in AF patients resulted in normalization of SDMA concentrations (0.465 ± 0.082 vs. 0.570 ± 0.134 µmol/L at baseline, p < 0.001). Patients with AF at the time of blood sampling had significantly lower hArg (1.65 ± 0.51 vs. 1.85 ± 0.60 µmol/L, p = 0.006) and higher ADMA concentrations (0.526 ± 0.08 vs. 0.477 ± 0.08 µmol/L, p < 0.001) compared with AF patients in SR. hArg concentrations were lower in patients with advanced AF progression phenotypes (persistent AF with LVA (p = 0.046)) independent of heart rhythm at blood sampling. Summarizing, arginine metabolism imbalance is associated with AF in general and AF progression and may contribute to associated risk. KEY MESSAGES: • Heart rhythm at blood withdrawal affects ADMA and hArg level in AF patients. • SDMA is higher in AF patients. • SDMA levels normalize after sinus rhythm restoration. • hArg levels decrease in advanced AF progression phenotypes.


Subject(s)
Arginine/metabolism , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Aged , Arginine/analogs & derivatives , Arginine/blood , Atrial Fibrillation/blood , Biomarkers/blood , Biomarkers/metabolism , Cohort Studies , Disease Progression , Female , Homoarginine/blood , Homoarginine/metabolism , Humans , Male , Middle Aged , Phenotype
19.
Amino Acids ; 52(6-7): 1067-1069, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32594255

ABSTRACT

Our study evaluated the effect of creatine and homoarginine in AGAT- and GAMT-deficient mice after simvastatin exposure. Balestrino and Adriano suggest that guanidinoacetate might explain the difference between AGAT- and GAMT-deficient mice in simvastatin-induced myopathy. We agree with Balestrino and Adriano that our data shows that (1) creatine possesses a protective potential to ameliorate statin-induced myopathy in humans and mice and (2) homoarginine did not reveal a beneficial effect in statin-induced myopathy. Third, we agree that guanidinoacetate can be phosphorylated and partially compensate for phosphocreatine. In our study, simvastatin-induced damage showed a trend to be less pronounced in GAMT-deficient mice compared with wildtype mice. Therefore, (phospo) guanidinoacetate cannot completely explain the milder phenotype of GAMT-deficient mice, but we agree that it might contribute to ameliorate statin-induced myopathy in GAMT-deficient mice compared with AGAT-deficient mice. Finally, we agree with Balestino and Adriano that AGAT metabolites should further be evaluated as potential treatments in statin-induced myopathy.


Subject(s)
Creatine/metabolism , Glycine/analogs & derivatives , Homoarginine/metabolism , Muscular Diseases/metabolism , Amidinotransferases/deficiency , Amino Acid Metabolism, Inborn Errors , Animals , Creatine/pharmacology , Developmental Disabilities , Glycine/metabolism , Guanidinoacetate N-Methyltransferase/deficiency , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Intellectual Disability , Mice , Muscular Diseases/chemically induced , Phosphocreatine/metabolism , Speech Disorders
20.
Int J Mol Sci ; 21(5)2020 Mar 05.
Article in English | MEDLINE | ID: mdl-32150996

ABSTRACT

The amino acid L-arginine serves as substrate for the nitric oxide synthase which is crucial in vascular function and disease. Derivatives of arginine, such as asymmetric (ADMA) and symmetric dimethylarginine (SDMA), are regarded as markers of endothelial dysfunction and have been implicated in vascular disorders. While there is a variety of studies consolidating ADMA as biomarker of cerebrovascular risk, morbidity and mortality, SDMA is currently emerging as an interesting metabolite with distinct characteristics in ischemic stroke. In contrast to dimethylarginines, homoarginine is inversely associated with adverse events and mortality in cerebrovascular diseases and might constitute a modifiable protective risk factor. This review aims to provide an overview of the current evidence for the pathophysiological role of arginine derivatives in cerebrovascular ischemic diseases. We discuss the complex mechanisms of arginine metabolism in health and disease and its potential clinical implications in diverse aspects of ischemic stroke.


Subject(s)
Arginine/analogs & derivatives , Cerebrovascular Disorders/physiopathology , Endothelium, Vascular/pathology , Nitric Oxide Synthase/metabolism , Arginine/metabolism , Biomarkers , Cerebrovascular Disorders/metabolism , Endothelium, Vascular/metabolism , Homoarginine/metabolism , Humans , Nitric Oxide Synthase/antagonists & inhibitors , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...