1
|
Anand SK, Governale TA, Zhang X, Razani B, Yurdagul A, Pattillo CB, Rom O. Amino Acid Metabolism and Atherosclerotic Cardiovascular Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:510-524. [PMID: 38171450 PMCID: PMC10988767 DOI: 10.1016/j.ajpath.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/09/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024]
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.
Collapse
Affiliation(s)
- Sumit Kumar Anand
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Theresea-Anne Governale
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Xiangyu Zhang
- Division of Cardiology and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Babak Razani
- Division of Cardiology and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Christopher B Pattillo
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana.
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana.
| |
Collapse
|
2
|
Seeley EH, Liu Z, Yuan S, Stroope C, Cockerham E, Rashdan NA, Delgadillo L, Finney AC, Kumar D, Das S, Razani B, Liu W, Traylor J, Orr AW, Rom O, Pattillo CB, Yurdagul A. Spatially Resolved Metabolites in Stable and Unstable Human Atherosclerotic Plaques Identified by Mass Spectrometry Imaging. Arterioscler Thromb Vasc Biol 2023; 43:1626-1635. [PMID: 37381983 PMCID: PMC10527524 DOI: 10.1161/atvbaha.122.318684] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/14/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Impairments in carbohydrate, lipid, and amino acid metabolism drive features of plaque instability. However, where these impairments occur within the atheroma remains largely unknown. Therefore, we sought to characterize the spatial distribution of metabolites within stable and unstable atherosclerosis in both the fibrous cap and necrotic core. METHODS Atherosclerotic tissue specimens from 9 unmatched individuals were scored based on the Stary classification scale and subdivided into stable and unstable atheromas. After performing mass spectrometry imaging on these samples, we identified over 850 metabolite-related peaks. Using MetaboScape, METASPACE, and Human Metabolome Database, we confidently annotated 170 of these metabolites and found over 60 of these were different between stable and unstable atheromas. We then integrated these results with an RNA-sequencing data set comparing stable and unstable human atherosclerosis. RESULTS Upon integrating our mass spectrometry imaging results with the RNA-sequencing data set, we discovered that pathways related to lipid metabolism and long-chain fatty acids were enriched in stable plaques, whereas reactive oxygen species, aromatic amino acid, and tryptophan metabolism were increased in unstable plaques. In addition, acylcarnitines and acylglycines were increased in stable plaques whereas tryptophan metabolites were enriched in unstable plaques. Evaluating spatial differences in stable plaques revealed lactic acid in the necrotic core, whereas pyruvic acid was elevated in the fibrous cap. In unstable plaques, 5-hydroxyindoleacetic acid was enriched in the fibrous cap. CONCLUSIONS Our work here represents the first step to defining an atlas of metabolic pathways involved in plaque destabilization in human atherosclerosis. We anticipate this will be a valuable resource and open new avenues of research in cardiovascular disease.
Collapse
Affiliation(s)
- Erin H. Seeley
- Department of Chemistry, University of Texas at Austin, TX, USA
| | - Zhipeng Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, IN, USA
| | - Shuai Yuan
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA, USA
| | - Chad Stroope
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Elizabeth Cockerham
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Nabil A Rashdan
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Luisa Delgadillo
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Dhananjay Kumar
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Sandeep Das
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Babak Razani
- Cardiovascular Division, Department of Medicine and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
- John Cochran VA Medical Center, St. Louis, MO, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences and Department of Pharmacology, Wayne State University, MI, USA
| | - James Traylor
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - A Wayne Orr
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Christopher B Pattillo
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center at Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center at Shreveport, LA, USA
| |
Collapse
|
3
|
Schwieren L, Jensen M, Schulz R, Lezius S, Laxy E, Milatz M, Thomalla G, Böger R, Gerloff C, Magnus T, Schwedhelm E, Choe CU. Homoarginine Associates with Carotid Intima-Media Thickness and Atrial Fibrillation and Predicts Adverse Events after Stroke. Life (Basel) 2023; 13:1590. [PMID: 37511965 PMCID: PMC10381763 DOI: 10.3390/life13071590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Homoarginine is associated with cardio- and cerebrovascular morbidity and mortality. However, the underlying pathomechanisms remain elusive. Here, we evaluated the association of homoarginine with adverse events (i.e., death, stroke, and myocardial infarction) and carotid intima-media thickness (cIMT) in stroke patients. In the prospective bioMARKers in STROKE (MARK-STROKE) cohort, patients with acute ischemic stroke or transient ischemic attack (TIA) were enrolled. Plasma homoarginine concentrations were analyzed and associated with clinical phenotypes in cross-sectional (374 patients) and prospective (273 patients) analyses. Adjustments for possible confounders were evaluated. A two-fold increase in homoarginine was inversely associated with the National Institutes of Health Stroke Scale (NIHSS) score at admission, cIMT, and prevalent atrial fibrillation (mean factor -0.68 [95% confidence interval (CI): -1.30, -0.07], -0.14 [95% CI: -0.22, -0.05]; and odds ratio 0.57 [95% CI: 0.33, 0.96], respectively). During the follow-up (median 284 [25th, 75th percentile: 198, 431] days), individuals with homoarginine levels in the highest tertile had fewer incident events compared with patients in the lowest homoarginine tertile independent of traditional risk factors (hazard ratio 0.22 [95% CI: 0.08, 0.63]). A lower prevalence of atrial fibrillation and a reduced cIMT pinpointed potential underlying pathomechanisms.
Collapse
Affiliation(s)
- Laura Schwieren
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
- Institute of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Märit Jensen
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Robert Schulz
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Susanne Lezius
- Institute of Medical Biometry and Epidemiology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Elena Laxy
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Magalie Milatz
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Götz Thomalla
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christian Gerloff
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK e.V.) Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Chi-Un Choe
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
4
|
Koch V, Gruenewald LD, Gruber-Rouh T, Eichler K, Leistner DM, Mahmoudi S, Booz C, Bernatz S, D'Angelo T, Albrecht MH, Alizadeh LS, Nour-Eldin NEA, Scholtz JE, Yel I, Vogl TJ, März W, Hardt SE, Martin SS. Homoarginine in the cardiovascular system: Pathophysiology and recent developments. Fundam Clin Pharmacol 2022; 37:519-529. [PMID: 36509694 DOI: 10.1111/fcp.12858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Upcoming experimental and epidemiological data have identified the endogenous non-proteinogenic amino acid L-homoarginine (L-hArg) not only as a novel biomarker for cardiovascular disease but also as being directly involved in the pathogenesis of cardiac dysfunction. The association of low L-hArg levels with adverse cardiovascular events and mortality has proposed the idea of nutritional supplementation to rescue pathways inversely associated with cardiovascular health. Subsequent clinical and experimental studies contributed significantly to our knowledge of potential effects on the cardiorenal axis, acting either as a biomarker or a cardiovascular active agent. In this review article, we provide a comprehensive summary of the L-hArg metabolism, pathophysiological aspects, and current developments in the field of experimental and clinical evidence in favor of protective cardiovascular effects. Establishing a reliable biomarker to identify patients at high risk to die of cardiovascular disease represents one of the main goals for tackling this disease and providing individual therapeutic guidance.
Collapse
Affiliation(s)
- Vitali Koch
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany.,Department of Cardiology, Angiology, and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | - Katrin Eichler
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - David M Leistner
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Christian Booz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Simon Bernatz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Tommaso D'Angelo
- Department of Biomedical Sciences and Morphological and Functional Imaging, University Hospital Messina, Messina, Italy
| | | | - Leona S Alizadeh
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Jan-Erik Scholtz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Ibrahim Yel
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Thomas J Vogl
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Winfried März
- Fifth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan E Hardt
- Department of Cardiology, Angiology, and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | - Simon S Martin
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
5
|
Schwedhelm E, Cordts K, Moritz E, Wesemann R, Choe CU, Böger R, Ittermann T, Dörr M, Friedrich N, Bahls M. Reference Interval for Serum L-Homoarginine Determined with Enzyme-Linked Immunosorbent Assay in the Population-Based Study of Health in Pomerania. J Appl Lab Med 2022; 7:1272-1282. [DOI: 10.1093/jalm/jfac074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/12/2022] [Indexed: 11/12/2022]
Abstract
Abstract
Background
Low levels of the endogenous amino acid L-homoarginine are a risk factor for cardiovascular morbidity and mortality. For individual risk prediction, commercially available test systems are mandatory. This study aims at formulating sex- and age-specific reference intervals of serum L-homoarginine determined with an ELISA.
Methods
We determined reference intervals for serum L-homoarginine stratified by age and sex in a sample of 1285 healthy participants of the Study of Health in Pomerania (SHIP)-TREND cohort after exclusion of participants with cardiovascular diseases, diabetes mellitus, hypertension, metabolic syndrome, elevated liver enzymes, chronic kidney disease stages III or IV, or body mass index >25 kg/m2. Serum L-homoarginine was determined applying a commercially available ELISA.
Results
The reference cohort included 836 women (median age 41, 25th and 75th percentiles are 32 and 50 years) and 449 men (median age 38, 25th, and 75th percentiles are 30 and 49 years). The median serum concentration of L-homoarginine was 1.93 (25th 1.49; 75th 2.60) µmol/L in women and 2.02 (25th 1.63; 75th 2.61) µmol/L in men (P = 0.04). The reference intervals (2.5th to 97.5th percentile) were 0.89–5.29 µmol/L for women and 1.09–3.76 µmol/L for men. The L-homoarginine serum concentration declined over age decades in both sexes and a notable interaction with sex hormone intake in women was observed.
Conclusions
The novelty of our study is that we determined reference intervals specific for the L-isomer being lower than those previously reported for homoarginine in SHIP and thus might be helpful in identifying individuals suitable for oral L-homoarginine supplementation.
Collapse
Affiliation(s)
- Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck , Hamburg , Germany
| | - Kathrin Cordts
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Eileen Moritz
- Institute of Pharmacology, University Medicine Greifswald , Greifswald , Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald , Greifswald , Germany
| | | | - Chi un Choe
- Department of Neurology, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Till Ittermann
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald , Greifswald , Germany
- Institute for Community Medicine, University Medicine Greifswald , Greifswald , Germany
| | - Marcus Dörr
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald , Greifswald , Germany
- Department of Internal Medicine B, University Medicine Greifswald , Greifswald , Germany
| | - Nele Friedrich
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald , Greifswald , Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald , Greifswald , Germany
| | - Martin Bahls
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald , Greifswald , Germany
- Department of Internal Medicine B, University Medicine Greifswald , Greifswald , Germany
| |
Collapse
|
6
|
Koch V, Gruenewald LD, Gruber‐Rouh T, Martin S, Eichler K, Booz C, Yel I, Vogl TJ, Buchner K, Hagenmueller M, März W, Frey N, Hardt SE, Riffel JH. Homoarginine treatment of rats improves cardiac function and remodeling in response to pressure overload. Fundam Clin Pharmacol 2022; 36:992-1004. [DOI: 10.1111/fcp.12808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Vitali Koch
- Goethe University Frankfurt Frankfurt am Main Germany
- Department of Cardiology, Angiology, and Pulmonology Heidelberg University Hospital Heidelberg Germany
| | | | | | - Simon Martin
- Goethe University Frankfurt Frankfurt am Main Germany
| | | | | | - Ibrahim Yel
- Goethe University Frankfurt Frankfurt am Main Germany
| | | | - Kristina Buchner
- Institute of Human Genetics, Section for Developmental Genetics University of Heidelberg Heidelberg Germany
| | - Marco Hagenmueller
- Department of Cardiology, Angiology, and Pulmonology Heidelberg University Hospital Heidelberg Germany
| | - Winfried März
- Synlab Academy Synlab Holding Deutschland GmbH Augsburg Germany
| | - Norbert Frey
- Department of Cardiology, Angiology, and Pulmonology Heidelberg University Hospital Heidelberg Germany
| | - Stefan E. Hardt
- Department of Cardiology, Angiology, and Pulmonology Heidelberg University Hospital Heidelberg Germany
| | - Johannes H. Riffel
- Department of Cardiology, Angiology, and Pulmonology Heidelberg University Hospital Heidelberg Germany
| |
Collapse
|
7
|
Kleist CJ, Choe CU, Atzler D, Schönhoff M, Böger R, Schwedhelm E, Wicha SG. Population kinetics of homoarginine and optimized supplementation for cardiovascular risk reduction. Amino Acids 2022; 54:889-896. [PMID: 35618975 PMCID: PMC9213336 DOI: 10.1007/s00726-022-03169-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/01/2022] [Indexed: 11/24/2022]
Abstract
Homoarginine is an endogenous amino acid whose levels are reduced in patients with renal, cardio- and cerebrovascular disease. Moreover, low homoarginine concentrations independently predict morbidity and mortality in these patients. Besides endogenous synthesis, homoarginine is also a constituent of the human diet. The objective of the present study was to analyze the kinetics of orally supplemented homoarginine in human plasma by means of a pharmacometric approach. We developed a pharmacometric model to evaluate different dosing regimens, especially the regimen of 125 mg once weekly, based on a previous clinical study (n = 20). The model was adapted to account for differences in baseline homoarginine plasma concentrations between healthy and diseased individuals. A novel dosing regimen of 25 mg once daily led to higher attainment of homoarginine reference concentrations using clinical trial simulations. With 25 mg/day, the trough concentration of only 6% of the older and 3.8% of the younger population was predicted to be below the target concentration of 2.0-4.1 µmol/L. In synopsis, the new dosing regimen recapitulates the kinetics of homoarginine in healthy individuals optimally.
Collapse
Affiliation(s)
- Christine J Kleist
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Bundesstraße 45, 20146, Hamburg, Germany
| | - Chi-Un Choe
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mirjam Schönhoff
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Bundesstraße 45, 20146, Hamburg, Germany.
| |
Collapse
|
8
|
Goni L, Razquin C, Toledo E, Guasch-Ferré M, Clish CB, Babio N, Wittenbecher C, Atzeni A, Li J, Liang L, Dennis C, Alonso-Gómez Á, Fitó M, Corella D, Gómez-Gracia E, Estruch R, Fiol M, Lapetra J, Serra-Majem L, Ros E, Arós F, Salas-Salvadó J, Hu FB, Martínez-González MA, Ruiz-Canela M. Arginine catabolism metabolites and atrial fibrillation or heart failure risk: 2 case-control studies within the Prevención con Dieta Mediterránea (PREDIMED) trial. Am J Clin Nutr 2022; 116:653-662. [PMID: 35575609 PMCID: PMC9437981 DOI: 10.1093/ajcn/nqac139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/25/2022] [Accepted: 05/11/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Arginine-derived metabolites are involved in oxidative and inflammatory processes related to endothelial functions and cardiovascular risks. OBJECTIVES We prospectively examined the associations of arginine catabolism metabolites with the risks of atrial fibrillation (AF) or heart failure (HF), and evaluated the potential modifications of these associations through Mediterranean diet (MedDiet) interventions in a large, primary-prevention trial. METHODS Two nested, matched, case-control studies were designed within the Prevención con Dieta Mediterránea (PREDIMED) trial. We selected 509 incident cases and 547 matched controls for the AF case-control study and 326 cases and 402 matched controls for the HF case-control study using incidence density sampling. Fasting blood samples were collected at baseline and arginine catabolism metabolites were measured using LC-tandem MS. Multivariable conditional logistic regression models were applied to test the associations between the metabolites and incident AF or HF. Interactions between metabolites and intervention groups (MedDiet groups compared with control group) were analyzed with the likelihood ratio test. RESULTS Inverse association with incident AF was observed for arginine (OR per 1 SD, 0.83; 95% CI: 0.73-0.94), whereas a positive association was found for N1-acetylspermidine (OR for Q4 compared with Q1 1.58; 95% CI: 1.13-2.25). For HF, inverse associations were found for arginine (OR per 1 SD, 0.82; 95% CI: 0.69-0.97) and homoarginine (OR per 1 SD, 0.81; 95% CI: 0.68-0.96), and positive associations were found for the asymmetric dimethylarginine (ADMA) and symmetric dimethlyarginine (SDMA) ratio (OR per 1 SD, 1.19; 95% CI: 1.02-1.41), N1-acetylspermidine (OR per 1 SD, 1.34; 95% CI: 1.12-1.60), and diacetylspermine (OR per 1 SD, 1.20; 95% CI: 1.02-1.41). In the stratified analysis according to the dietary intervention, the lower HF risk associated with arginine was restricted to participants in the MedDiet groups (P-interaction = 0.044). CONCLUSIONS Our results suggest that arginine catabolism metabolites could be involved in AF and HF. Interventions with the MedDiet may contribute to strengthen the inverse association between arginine and the risk of HF. This trial was registered at controlled-trials.com as ISRCTN35739639.
Collapse
Affiliation(s)
- Leticia Goni
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain,IdiSNA (Instituto de Investigación Sanitaria de Navarra), Pamplona, Spain,Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Razquin
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain,IdiSNA (Instituto de Investigación Sanitaria de Navarra), Pamplona, Spain,Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Estefanía Toledo
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain,IdiSNA (Instituto de Investigación Sanitaria de Navarra), Pamplona, Spain,Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA,Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, MA, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nancy Babio
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain,Institut d'Investigació Sanitària Pere i Virgili, Hospital Universitari Sant Joan de Reus, Reus, Spain
| | - Clemens Wittenbecher
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA,Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Alessandro Atzeni
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain,Institut d'Investigació Sanitària Pere i Virgili, Hospital Universitari Sant Joan de Reus, Reus, Spain
| | - Jun Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Ángel Alonso-Gómez
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Bioaraba Health Research Institute, Osakidetza Basque Health Service, Araba University Hospital, Vitoria-Gasteiz, Spain,University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Montserrat Fitó
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Dolores Corella
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Enrique Gómez-Gracia
- Department of Preventive Medicine, University of Malaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Ramón Estruch
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Department of Internal Medicine, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Miquel Fiol
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Plataforma de Ensayos Clínicos, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - Jose Lapetra
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, Sevilla, Spain
| | - Lluis Serra-Majem
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Nutrition Research Group, Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Emilio Ros
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Lipid Clinic, Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Fernando Arós
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Bioaraba Health Research Institute, Osakidetza Basque Health Service, Araba University Hospital, Vitoria-Gasteiz, Spain,University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Jordi Salas-Salvadó
- Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain,Institut d'Investigació Sanitària Pere i Virgili, Hospital Universitari Sant Joan de Reus, Reus, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA,Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, MA, USA
| | - Miguel A Martínez-González
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain,IdiSNA (Instituto de Investigación Sanitaria de Navarra), Pamplona, Spain,Centro de Investigacion Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | |
Collapse
|
9
|
Expression of cardiovascular-related microRNAs is altered in L-arginine:glycine amidinotransferase deficient mice. Sci Rep 2022; 12:5108. [PMID: 35332188 PMCID: PMC8948300 DOI: 10.1038/s41598-022-08846-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/09/2022] [Indexed: 11/09/2022] Open
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.
Collapse
|
10
|
ADMA and homoarginine independently predict mortality in critically ill patients. Nitric Oxide 2022; 122-123:47-53. [DOI: 10.1016/j.niox.2022.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/20/2022] [Accepted: 03/07/2022] [Indexed: 12/23/2022]
|
11
|
Porro B, Eligini S, Conte E, Cosentino N, Capra N, Cavalca V, Banfi C. An Optimized MRM-Based Workflow of the l-Arginine/Nitric Oxide Pathway Metabolites Revealed Disease- and Sex-Related Differences in the Cardiovascular Field. Int J Mol Sci 2022; 23:ijms23031136. [PMID: 35163055 PMCID: PMC8835333 DOI: 10.3390/ijms23031136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 11/29/2022] Open
Abstract
Clinical data indicate that low circulating l-homoarginine (HArg) concentrations are associated with cardiovascular (CV) disease, CV mortality, and all-cause mortality. A high number of LC-based analytical methods for the quantification of HArg, in combination with the l-arginine (Arg)-related pathway metabolites, have been reported. However, these methods usually consider a limited panel of analytes. Thus, in order to achieve a comprehensive picture of the Arg metabolism, we described an improved targeted metabolomic approach based on a multiple reaction monitoring (MRM) mass spectrometry method for the simultaneous quantification of the Arg/nitric oxide (NO) pathway metabolites. This methodology was then employed to quantify the plasma concentrations of these analytes in a cohort of individuals with different grades/types of coronary artery disease (CAD) in order to increase knowledge about the role of HArg and its associated metabolites in the CV field. Our results showed that the MRM method here implemented is suitable for the simultaneous assessment of a wide panel of amino acids involved in the Arg/NO metabolic pathway in plasma samples from patients with CV disease. Further, our findings highlighted an impairment of the Arg/NO metabolic pathway, and suggest a sex-dependent regulation of this metabolic route.
Collapse
|
12
|
Zhloba AA, Subbotina TF. Homoarginine test for evaluation of metabolic renal dysfunction. Klin Lab Diagn 2021; 66:709-717. [PMID: 35020282 DOI: 10.51620/0869-2084-2021-66-12-709-717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Low plasma L-homoarginine (hArg) concentration is an independent predictor of adverse cardiovascular outcomes and overall mortality, as well as the progression of chronic kidney disease (CKD). The enzyme L-arginine:glycinamidinotransferase (AGAT, EC 2.1.4.1) acts in the mitochondrial membrane of the renal tubular epithelium, forming the precursor of creatine, guanidinoacetic acid, and additionnaly by-product hArg. As it was shown recently, there is a decreased level of hArg in the late stages of CKD, however, the the level of hArg in the early stages of CKD remained unexplored. The aim of this study was to determine the diagnostic threshold levels of hArg in the blood of patients with stages 1 and 2 of CKD. In patients with the initial stages of CKD (n = 44) at the age of 58 (45-67) years, compared with the group of donors of 55 (42-58) years (n = 30), a significant decrease of hArg level was found. In the subgroup with stage CKD 2, the cut-off point of 1.59 μM threshold was characterized by greater sensitivity and specificity than in the subgroup with stage CKD 1 with 1.66 μM threshold level of hArg. For the full group, the hArg cut-off threshold was 1.60 μM, which is about to 0.2 μM lower than the lower limit of the reference interval for healthy individuals. It can be assumed that even before the formation of symptoms of proteinuria and albuminuria, a significant part of individuals from population cohort develops a state of decreased AGAT activity, since the expression of this enzyme is associated with a certain regulatory feedback inhibition at the body level. As a result of the study, it can be noted that in patients with early stages of CKD in the age group 45-67 years, there is a disturbance of the kidneys metabolic function. These metabolic changes can be detected by testing the level of hArg.
Collapse
Affiliation(s)
- A A Zhloba
- Pavlov First Saint Petersburg State Medical University of Minzdrav of Russia
| | - T F Subbotina
- Pavlov First Saint Petersburg State Medical University of Minzdrav of Russia
| |
Collapse
|
13
|
Hsu WH, Han DS, Ku WC, Chao YM, Chen CC, Lin YL. Metabolomic and proteomic characterization of sng and pain phenotypes in fibromyalgia. Eur J Pain 2021; 26:445-462. [PMID: 34608709 PMCID: PMC9298249 DOI: 10.1002/ejp.1871] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/16/2021] [Accepted: 10/03/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Fibromyalgia (FM) is characterized by chronic widespread pain. Its pathophysiological mechanisms remain poorly understood, and effective diagnosis and treatments are lacking. This study aimed to identify significantly changed biosignatures in FM and propose a novel classification for FM based on pain and soreness (sng) symptoms. METHODS Urine and serum samples from 30 FM patients and 25 controls underwent metabolomic and proteomic profiling. RESULTS Compared with controls, FM patients showed significant differential expression of three metabolites in urine and five metabolites and eight proteins in serum. Of them, DETP, 4-guanidinobutanoic acid, SM(d18:1/18:0), PC(20:1(11Z)/18:0), S100A7, SERPINB3, galectin-7 and LYVE1 were first reported as potential biomarkers for FM. Furthermore, lactate, 2-methylmaleate and cotinine in urine and lactate, SM(d18:1/25:1), SM(d18:1/26:1) and prostaglandin D2 (PGD2) and PCYOX1, ITIH4, PFN1, LRG1, C8G, C8A, CP, CDH5 and DBH in serum could differentiate pain- (PG) and sng-dominant groups (SG). Lactate, 2-methylmaleate, cotinine, PCYOX1, ITIH4, PFN1 and DBH have a higher level in SG. SM(d18:1/25:1), SM(d18:1/26:1), PGD2, LRG1, C8G, C8A, CP and CDH5 in SG are lower than PG. The omics results indicated disordered free radical scavenging, and lipid and amino acid metabolism networks and resulting NF-κB-dependent cytokine generation in FM. Lactate level was altered simultaneously in urine and serum and significantly higher in sng-dominant patients than others. CONCLUSIONS In this study, we identified potential biomarkers from FM patients. The selected biomarkers could discriminate sng and pain phenotypes in FM patients. These results could help elucidate the underlying pathological mechanisms for more effective diagnosis and therapy for FM.
Collapse
Affiliation(s)
- Wei-Hsiang Hsu
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Der-Sheng Han
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan.,Community and Geriatric Medicine Research Center, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan.,Department of Physical Medicine and Rehabilitation, National Taiwan University College of Medicine, Taipei, Taiwan.,Health Science and Wellness Center, National Taiwan University, Taipei, Taiwan
| | - Wei-Chi Ku
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
| | - Yen-Ming Chao
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, Taiwan.,Taiwan Mouse Clinic, Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Yun-Lian Lin
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan.,Department of Pharmacy, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
14
|
Association of Lower Plasma Homoarginine Concentrations with Greater Risk of All-Cause Mortality in the Community: The Framingham Offspring Study. J Clin Med 2020; 9:jcm9062016. [PMID: 32604958 PMCID: PMC7356383 DOI: 10.3390/jcm9062016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/03/2020] [Accepted: 06/24/2020] [Indexed: 02/06/2023] Open
Abstract
Lower circulating homoarginine concentrations have been associated with morbidity and mortality in patients with established cardiovascular disease (CVD). We assayed plasma homoarginine concentrations in 3331 Framingham Offspring Study participants attending examination cycle six (mean age 58.6 years, 53% women). We evaluated correlates of plasma homoarginine and related homoarginine to incident CVD and death. We also classified participants as having higher (upper quartile) versus lower (lower three quartiles) homoarginine and previously assayed asymmetric dimethylarginine (ADMA) concentrations, and created cross-classification groups. We observed 630 incident CVD events and 940 deaths during a median follow-up of 18 years. In multivariable regression analysis, homoarginine was associated positively with male sex, body mass index, anti-hypertensive medication use and systolic blood pressure, but inversely with age and smoking. Higher homoarginine levels were associated with a lower mortality risk (hazard ratio (HR) per SD increment, 0.83, 95% CI: 0.74–0.93) adjusting for standard CVD risk factors, and ADMA. Among the cross-classification groups, participants with higher homoarginine and lower ADMA had a lower mortality risk (HR, 0.81, 95% CI: 0.67–0.98) compared to those with low levels of both. Further studies are needed to dissect the mechanisms of the association of homoarginine and mortality over decades in the community.
Collapse
|
15
|
Analysis of L-arginine:glycine amidinotransferase-, creatine- and homoarginine-dependent gene regulation in the murine heart. Sci Rep 2020; 10:4821. [PMID: 32179820 PMCID: PMC7076046 DOI: 10.1038/s41598-020-61638-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/06/2020] [Indexed: 02/04/2023] Open
Abstract
L-arginine:glycine amidinotransferase (AGAT) and its metabolites creatine and homoarginine (HA) have been linked to cardiovascular pathologies in both human and murine studies, but the underlying molecular mechanisms are poorly understood. Here, we report the first analysis of heart transcriptome variation using microarrays in an AGAT-deficient (AGAT−/−) mouse model to evaluate AGAT-, creatine- and HA-dependent gene regulation. Our data revealed significant differences of gene expression between AGAT−/− and wild-type (WT) mice, affecting cardiac energy metabolism (Fbp2, Ucp2), cardiac hypertrophy and fibrosis (Nppa, Ctgf), immune response (Fgl2), and the conduction system of the heart (Dsc2, Ehd4, Hcn2, Hcn4, Scn4a, Scn4b). All of these genes being expressed on WT level in creatine-supplemented mice. Using in silico analysis based on the GEO database we found that most of these candidate genes (Ctgf, Dsc2, Fbp2, Fgl2, Hcn2, Nppa) revealed significant alterations in a WT mouse model of myocardial infarction underlining a pathophysiological relationship between AGAT metabolism and cardiovascular disease.
Collapse
|
16
|
Karetnikova ES, Jarzebska N, Markov AG, Weiss N, Lentz SR, Rodionov RN. Is Homoarginine a Protective Cardiovascular Risk Factor? Arterioscler Thromb Vasc Biol 2020; 39:869-875. [PMID: 30866658 DOI: 10.1161/atvbaha.118.312218] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A series of recent epidemiological studies have implicated the endogenous nonproteinogenic amino acid l-homoarginine as a novel candidate cardiovascular risk factor. The association between homoarginine levels and the risk of adverse cardiovascular outcomes is inverse (ie, high cardiovascular risk is predicted by low rather than high homoarginine levels), which makes it plausible to normalize systemic homoarginine levels via oral supplementation. The emergence of homoarginine as a potentially treatable protective cardiovascular risk factor has generated a wave of hope in the field of cardiovascular prevention. Herein, we review the biochemistry, physiology, and metabolism of homoarginine, summarize the strengths and weaknesses of the epidemiological evidence linking homoarginine to cardiovascular disease and its potential protective cardiovascular effects, and identify priorities for future research needed to define the clinical utility of homoarginine as a prognostic factor and therapeutic target in cardiovascular disease.
Collapse
Affiliation(s)
- Ekaterina S Karetnikova
- From the Department of Physiology, Saint-Petersburg State University, Russia (E.S.K., A.G.M.)
| | - Natalia Jarzebska
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital "Carl Gustav Carus", Technische Universität Dresden, Germany (N.J., N.W., R.N.R.)
| | - Alexander G Markov
- From the Department of Physiology, Saint-Petersburg State University, Russia (E.S.K., A.G.M.)
| | - Norbert Weiss
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital "Carl Gustav Carus", Technische Universität Dresden, Germany (N.J., N.W., R.N.R.)
| | - Steven R Lentz
- Department of Internal Medicine, University of Iowa Carver College of Medicine (S.R.L.)
| | - Roman N Rodionov
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital "Carl Gustav Carus", Technische Universität Dresden, Germany (N.J., N.W., R.N.R.).,Flinders University, Adelaide, Australia (R.N.R.)
| |
Collapse
|
17
|
Zhloba AA, Subbotina TF, Molchan NS, Polushin YS. [Homoarginine level and methionine-homocysteine balance in patients with ischemic heart disease.]. Klin Lab Diagn 2019; 64:516-524. [PMID: 31610102 DOI: 10.18821/0869-2084-2019-64-9-516-524] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 06/25/2019] [Indexed: 11/17/2022]
Abstract
The level of homoarginine (hArg) in terms of prognostic significance may exceed the natriuretic peptides and other well-known markers according to the latest data about the progression of cardiovascular diseases. The lack of data on the association of hArg levels with levels of other metabolites makes it difficult to understand its role in the pathogenesis of cardiovascular diseases. Relationships of hArg and other amino acids, including methionine (Met) and total homocysteine (tHcy), and their ratio in patients with ischemic heart disease were evaluated. The study included 74 patients with coronary heart disease (57 men and 17 women) aged 62 (57 - 67) years before coronary artery bypass surgery and 27 healthy people of similar age. In patients, the level of hArg was almost 2 times lower (p < 0.05) than in healthy individuals and rates lower than 1.4 μM were in half of them. The statistically significant decrease (p = 0.0025) of the Met/tHcy ratio corresponded to a decrease in the level of hArg. This ratio did not correlate with glucose level or body mass index. Less statistical significance of hArg correlation with levels of Met or tHcy separately was observed. In the subgroup of patients with hAarg level above 2.1 μM, a lower incidence of myocardial infarction was noted. Thus, a low hArg level is associated with impaired metabolism of sulfur-containing amino acids involved in transmethylation reactions, in patients with ischemic heart disease. The Met/tHcy ratio, closely correlating with the level of hArg, apparently reveals a link between the reactions of creatine formation and transmethylation, highlighting a cohort of patients with the most profound and dangerous changes in tissue metabolism.
Collapse
Affiliation(s)
- A A Zhloba
- Federal State Budget Educational Institution of Higher Eduction "The Pavlov First Saint-Petersburg State Medical University" of Minzdrav of Russia, 197022, St. Petersburg, Russia
| | - T F Subbotina
- Federal State Budget Educational Institution of Higher Eduction "The Pavlov First Saint-Petersburg State Medical University" of Minzdrav of Russia, 197022, St. Petersburg, Russia
| | - N S Molchan
- Federal State Budget Educational Institution of Higher Eduction "The Pavlov First Saint-Petersburg State Medical University" of Minzdrav of Russia, 197022, St. Petersburg, Russia
| | - Yu S Polushin
- Federal State Budget Educational Institution of Higher Eduction "The Pavlov First Saint-Petersburg State Medical University" of Minzdrav of Russia, 197022, St. Petersburg, Russia
| |
Collapse
|
18
|
Guľašová Z, Guerreiro SG, Link R, Soares R, Tomečková V. Tackling endothelium remodeling in cardiovascular disease. J Cell Biochem 2019; 121:938-945. [DOI: 10.1002/jcb.29379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/15/2019] [Indexed: 01/19/2023]
Affiliation(s)
- Zuzana Guľašová
- Department of Experimental Medicine, Faculty of Medicine University of Pavol Jozef Šafárik in Košice Košice Slovakia
| | - Susana G. Guerreiro
- Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine University of Porto Porto Portugal
- I3S, Instituto de Investigação e Inovação em Saúde University of Porto Porto Portugal
- Faculdade de Ciências da Nutrição e Alimentação University of Porto Porto Portugal
| | - Rene Link
- Department of Experimental Medicine, Faculty of Medicine University of Pavol Jozef Šafárik in Košice Košice Slovakia
| | - Raquel Soares
- Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine University of Porto Porto Portugal
- I3S, Instituto de Investigação e Inovação em Saúde University of Porto Porto Portugal
| | - Vladimíra Tomečková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine University of Pavol Jozef Šafárik in Košice Košice Slovakia
| |
Collapse
|
19
|
Association of Asymmetric Dimethylarginine and Diastolic Dysfunction in Patients with Hypertrophic Cardiomyopathy. Biomolecules 2019; 9:biom9070277. [PMID: 31337005 PMCID: PMC6681289 DOI: 10.3390/biom9070277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/29/2022] Open
Abstract
Despite genetic heterogeneity, early manifestation of diastolic dysfunction (DD) is common in hypertrophic cardiomyopathy (HCM). Nitric oxide (NO) may contribute to myocardial relaxation. NO synthases (NOS) use l-arginine (Arg) as a substrate, as asymmetric dimethylarginine (ADMA) is a direct endogenous inhibitor of NOS. This study aimed to analyze the association of Arg and its derivates, i.e., l-homoarginine (hArg), ADMA and symmetric dimethylarginine (SDMA), with DD in HCM patients. In 215 HCM patients (mean age 54 ± 15 years, 58% male) transmitral and mitral annulus velocities were echocardiographically analyzed. Plasma concentrations of Arg derivatives were measured by liquid chromatography tandem-mass spectrometry. In 143 (70%) patients suffering from DD, ADMA showed the strongest association with DD (0.66 ± 0.16, 0.72 ± 0.24, and 0.76 ± 0.26 µmol/L, p < 0.01 for trend). In linear regression analyses, positive association per standard deviation increase of ADMA was found with E-wave (beta coefficient (95% confidence interval): 4.72 (0.43–9.01); p < 0.05) and mean E/E’ (1.76 (0.73–2.79) p < 0.001). Associations were adjusted for age, sex, body mass index (BMI), diabetes mellitus, coronary artery disease, and arterial hypertension. Elevated ADMA is associated with the severity of DD in HCM. Higher ADMA level might lead to decreased NO production and thus an impaired myocardial relaxation pattern.
Collapse
|
20
|
Schwemer TF, Deutscher N, Diermann N, Böger R, Schwedhelm E, Blankenberg S, Friedrich FW. Effect of ranolazine on plasma arginine derivatives and urinary isoprostane 8-iso-PGF 2α in patients with myocardial infarction in the randomized RIMINI-Trial. Sci Rep 2019; 9:5708. [PMID: 30952941 PMCID: PMC6450888 DOI: 10.1038/s41598-019-42239-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 03/13/2019] [Indexed: 12/20/2022] Open
Abstract
The purpose of the present study was to assess whether 6-week ranolazine application on top of guideline-based treatment impacts on the arginine/NO pathway and urinary isoprostane 8-iso-PGF2α as marker of oxidative stress in patients directly after a myocardial infarction. 20 patients with unstable angina pectoris and proof of acute cardiac ischemia entered the study. 10 subjects received the study drug ranolazine in addition to standard treatment, the others received only standard treatment. Urine and venous blood were collected before and after treatment. At the end of the study and compared to baseline, homoarginine levels had increased in the control group. This was not the case in ranolazine-patients. Interestingly, in ranolazine-treated-patients arginine plasma levels were significantly higher at the end of the study than at baseline (difference +26 µmol/L, 95% CI 8.6 to 44 µmol/L). ADMA and SDMA levels were not different. Urine levels of the oxidative stress marker 8-iso-PGF2α tended to be lower in ranolazine-treated patients (−144 pmol/mg creatinine). Findings of this hypothesis-driven study give evidence that ranolazine treatment enhances arginine plasma levels and lowers oxidative stress.
Collapse
Affiliation(s)
| | | | | | - Rainer Böger
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.,Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edzard Schwedhelm
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.,Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Blankenberg
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.,University Heart Center Hamburg, Hamburg, Germany
| | - Felix W Friedrich
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
21
|
Maas R, Mieth M, Titze SI, Hübner S, Fromm MF, Kielstein JT, Schmid M, Köttgen A, Kronenberg F, Krane V, Hausknecht B, Eckardt KU, Schneider MP. Drugs linked to plasma homoarginine in chronic kidney disease patients—a cross-sectional analysis of the German Chronic Kidney Disease cohort. Nephrol Dial Transplant 2018; 35:1187-1195. [DOI: 10.1093/ndt/gfy342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/25/2018] [Indexed: 01/16/2023] Open
Abstract
Abstract
Background
Elevated plasma concentrations of symmetric and asymmetric dimethylarginine (SDMA and ADMA, respectively) and a lower plasma concentration of the structurally related homoarginine are commonly observed in patients with chronic kidney disease (CKD) and independently predict total mortality as well as progression of renal disease. We aimed to identify drugs that may alter this adverse metabolite pattern in a favourable fashion.
Methods
Plasma ADMA, SDMA, homoarginine and l-arginine were determined by liquid chromatography–tandem mass spectrometry in 4756 CKD patients ages 18–74 years with an estimated glomerular filtration rate (eGFR) of 30–60 mL/min/1.73 m2 or an eGFR >60 mL/min/1.73 m2 and overt proteinuria who were enrolled in the German Chronic Kidney Disease (GCKD) study. Associations between laboratory, clinical and medication data were assessed.
Results
Intake of several commonly used drugs was independently associated with plasma concentrations of homoarginine and/or related metabolites. Among these, the peroxisome proliferator-activated receptor alpha (PPAR-α) agonist fenofibrate was associated with the most profound differences in ADMA, SDMA and homoarginine plasma concentrations: 66 patients taking fenofibrate had a multivariable adjusted odds ratio (OR) of 5.83 [95% confidence interval (CI) 2.82–12.03, P < 0.001] to have a plasma homoarginine concentration above the median. The median homoarginine plasma concentration in patients taking fenofibrate was 2.30 µmol/L versus 1.55 in patients not taking the drug (P < 0.001). In addition, fibrates were significantly associated with lower plasma SDMA and higher l-arginine concentrations. In contrast, glucocorticoids were associated with lower plasma homoarginine, with adjusted ORs of 0.52 (95% CI 0.40–0.67, P < 0.001) and 0.53 (95% CI 0.31–0.90, P = 0.018) for prednisolone and methylprednisolone, respectively.
Conclusions
In a large cohort of CKD patients, intake of fenofibrate and glucocorticoids were independently associated with higher and lower plasma homoarginine concentrations, respectively. Effects on plasma homoarginine and methylarginines warrant further investigation as potential mechanisms mediating beneficial or adverse drug effects.
Collapse
Affiliation(s)
- Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Maren Mieth
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stephanie I Titze
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Silvia Hübner
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jan T Kielstein
- Divison of Nephrology, Medical School Hannover, Hannover, Germany
- Medical Clinic V Nephrology Rheumatology Blood Purification, Klinikum Braunschweig, Braunschweig, Germany
| | - Matthias Schmid
- Department of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Centre, University of Freiburg, Freiburg, Germany
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Vera Krane
- Department of Medicine I, Division of Nephrology, University Hospital Würzburg, Würzburg, Germany
| | - Birgit Hausknecht
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Markus P Schneider
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
22
|
Homoarginine/ADMA ratio and homoarginine/SDMA ratio as independent predictors of cardiovascular mortality and cardiovascular events in lower extremity arterial disease. Sci Rep 2018; 8:14197. [PMID: 30242192 PMCID: PMC6155043 DOI: 10.1038/s41598-018-32607-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/10/2018] [Indexed: 12/20/2022] Open
Abstract
Endothelial dysfunction plays a key role in development of atherosclerosis and lower extremity arterial disease (LEAD). Homoarginine, asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA) are sensitive markers for endothelial dysfunction and independent risk factors for cardiovascular death. However, homoarginine may influence the proatherogenic effects of ADMA and SDMA suggesting homoarginine/ADMA ratio or homoarginine/SDMA ratio as further predictors for cardiovascular mortality. Therefore, we investigated the predictive value of homoarginine/ADMA ratio and homoarginine/SDMA ratio related to cardiovascular mortality and cardiovascular events in claudicant patients with LEAD. 151 patients with intermittent claudication were included in a prospective observational study (observation time 7.7 ± 2.5 years) with cardiovascular mortality as main outcome parameter and the occurrence of cardiovascular events as secondary outcome parameter. Homoarginine, ADMA and SDMA were measured by high-performance liquid chromatography at baseline. Low homoarginine/ADMA ratio and homoarginine/SDMA ratio were independently associated with higher cardiovascular mortality (HR 2.803 [95% CI 1.178–6.674], p = 0.020; HR 2.782 [95% CI 1.061–7.290], p = 0.037, respectively) and higher incidence of cardiovascular events (HR 1.938 [95% CI 1.015–3.700], p = 0.045; HR 2.397 [95% CI 1.243–4.623], p = 0.009, respectively). We observed that homoarginine/ADMA ratio and homoarginine/SDMA ratio are independent predictors for long-term cardiovascular mortality and events in claudicant patients with LEAD.
Collapse
|
23
|
Cross-Sectional Associations between Homoarginine, Intermediate Phenotypes, and Atrial Fibrillation in the Community-The Gutenberg Health Study. Biomolecules 2018; 8:biom8030086. [PMID: 30200232 PMCID: PMC6165554 DOI: 10.3390/biom8030086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/26/2018] [Accepted: 08/27/2018] [Indexed: 01/19/2023] Open
Abstract
Homoarginine has come into the focus of interest as a biomarker for cardiovascular disease. Atrial fibrillation (AF) causes a substantial increase in morbidity and mortality. Whether circulating homoarginine is associated with occurrence or persistence of AF and may serve as a new predictive biomarker remains unknown. We measured plasma levels of homoarginine in the population-based Gutenberg health study (3761 patients included, of them 51.7% males), mean age 55.6 ± 10.9 years-old. Associations between homoarginine and intermediate electrocardiographic and echocardiographic phenotypes and manifest AF were examined. Patients with AF (124 patients, of them 73.4% males) had a mean age 64.8 ± 8.6 years-old compared to a mean age of 55.3 ± 10.9 in the population without AF (p-value < 0.001) and showed a less beneficial risk factor profile. The median homoarginine levels in individuals with and without AF were 1.9 μmol/L (interquartile range (IQR) 1.5–2.5) and 2.0 μmol/L (IQR 1.5–2.5), respectively, p = 0.56. In multivariable-adjusted regression analyses homoarginine was not statistically significantly related to electrocardiographic variables. Among echocardiographic variables beta per standard deviation increase was −0.12 (95% confidence interval (CI) −0.23–(−0.02); p = 0.024) for left atrial area and −0.01 (95% CI −0.02–(−0.003); p = 0.013) for E/A ratio. The odds ratio between homoarginine and AF was 0.91 (95% CI 0.70–1.16; p = 0.45). In our large, population-based cross-sectional study, we did not find statistically significant correlations between lower homoarginine levels and occurrence or persistence of AF or most standard electrocardiographic phenotypes, but some moderate inverse associations with echocardiographic left atrial size and E/A. Homoarginine may not represent a strong biomarker to identify individuals at increased risk for AF. Further investigations will be needed to elucidate the role of homoarginine and cardiac function.
Collapse
|
24
|
The biomarker and causal roles of homoarginine in the development of cardiometabolic diseases: an observational and Mendelian randomization analysis. Sci Rep 2017; 7:1130. [PMID: 28442717 PMCID: PMC5430630 DOI: 10.1038/s41598-017-01274-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 03/28/2017] [Indexed: 02/05/2023] Open
Abstract
High L-homoarginine (hArg) levels are directly associated with several risk factors for cardiometabolic diseases whereas low levels predict increased mortality in prospective studies. The biomarker role of hArg in young adults remains unknown. To study the predictive value of hArg in the development of cardiometabolic risk factors and diseases, we utilized data on high-pressure liquid chromatography-measured hArg, cardiovascular risk factors, ultrasound markers of preclinical atherosclerosis and type 2 diabetes from the population-based Young Finns Study involving 2,106 young adults (54.6% females, aged 24–39). We used a Mendelian randomization approach involving tens to hundreds of thousands of individuals to test causal associations. In our 10-year follow-up analysis, hArg served as an independent predictor for future hyperglycaemia (OR 1.31, 95% CI 1.06–1.63) and abdominal obesity (OR 1.60, 95% 1.14–2.30) in men and type 2 diabetes in women (OR 1.55, 95% CI 1.02–2.41). The MR analysis revealed no evidence of causal associations between serum hArg and any of the studied cardiometabolic outcomes. In conclusion, lifetime exposure to higher levels of circulating hArg does not seem to alter cardiometabolic disease risk. Whether hArg could be used as a biomarker for identification of individuals at risk developing cardiometabolic abnormalities merits further investigation.
Collapse
|
25
|
Atzler D, Schönhoff M, Cordts K, Ortland I, Hoppe J, Hummel FC, Gerloff C, Jaehde U, Jagodzinski A, Böger RH, Choe CU, Schwedhelm E. Oral supplementation with L-homoarginine in young volunteers. Br J Clin Pharmacol 2016; 82:1477-1485. [PMID: 27434056 DOI: 10.1111/bcp.13068] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/14/2016] [Accepted: 07/17/2016] [Indexed: 12/17/2022] Open
Abstract
AIMS Low blood concentrations of the naturally occurring amino acid L-homoarginine (L-hArg) are related to impaired cardiovascular outcome and mortality in humans and animals. L-hArg is a weak substrate of nitric oxide synthase and an inhibitor of arginases in vitro. The aim of our study was to obtain kinetic and dynamic data after oral L-hArg supplementation. METHODS In a double-blind, randomized, placebo-controlled crossover study, 20 young volunteers received 125 mg L-hArg once daily for 4 weeks. Kinetic parameters (Cmax , Tmax and AUC0-24h ) were calculated after ingestion of single and multiple doses of oral supplementation as primary endpoint. Secondary endpoints that were evaluated were routine laboratory, L-arginine, asymmetric dimethylarginine (ADMA), pulse wave velocity (PWV), augmentation index (AIx), flow-mediated vasodilatation (FMD), corticospinal excitability, i.e. motor threshold (MT), and cortical excitability, i.e. intracortical inhibition (ICI) and facilitation (ICF). RESULTS One hour after ingestion (Tmax ), L-hArg increased the baseline L-hArg plasma concentration (2.87 ± 0.91 μmol l-1 , mean ± SD) by 8.74 ± 4.46 [95% confidence intervals 6.65; 10.9] and 17.3 ± 4.97 [14.9; 19.6] μmol l-1 (Cmax ), after single and multiple doses, respectively. Once-only and 4 weeks of supplementation resulted in AUCs0-24h of 63.5 ± 28.8 [50.0; 76.9] and 225 ± 78.5 [188; 2624] μmol l-1 *h, for single and multiple doses, respectively. Routine laboratory parameters, L-arginine, ADMA, PWV, AIx, FMD, MT, ICI and ICF did not change by L-hArg supplementation compared to baseline. CONCLUSION Once daily orally applied 125 mg L-hArg raises plasma L-hArg four- and sevenfold after single dose and 4 weeks of supplementation, respectively, and is safe and well tolerated in young volunteers.
Collapse
Affiliation(s)
- Dorothee Atzler
- Department of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Hamburg/Kiel/Lübeck, Germany.,Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München Ludwig Maximilians-University of Munich, Munich, Germany
| | - Mirjam Schönhoff
- Department of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Kathrin Cordts
- Department of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Imke Ortland
- Institute of Pharmacy Department of Clinical Pharmacy, University of Bonn, Bonn, Germany
| | - Julia Hoppe
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedhelm C Hummel
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Gerloff
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Jaehde
- Institute of Pharmacy Department of Clinical Pharmacy, University of Bonn, Bonn, Germany
| | - Annika Jagodzinski
- DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Hamburg/Kiel/Lübeck, Germany.,Department of General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rainer H Böger
- Department of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Chi-Un Choe
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edzard Schwedhelm
- Department of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| |
Collapse
|
26
|
Atzler D, Baum C, Ojeda F, Keller T, Cordts K, Schnabel RB, Choe CU, Lackner KJ, Münzel T, Böger RH, Blankenberg S, Schwedhelm E, Zeller T. Low Homoarginine Levels in the Prognosis of Patients With Acute Chest Pain. J Am Heart Assoc 2016; 5:e002565. [PMID: 27076564 PMCID: PMC4859271 DOI: 10.1161/jaha.115.002565] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND The endogenous amino acid homoarginine predicts mortality in cerebro- and cardiovascular disease. The objective was to explore whether homoarginine is associated with atrial fibrillation (AF) and outcome in patients with acute chest pain. METHODS AND RESULTS One thousand six hundred forty-nine patients with acute chest pain were consecutively enrolled in this study, of whom 589 were diagnosed acute coronary syndrome (ACS). On admission, plasma concentrations of homoarginine as well as brain natriuretic peptide (BNP), and high-sensitivity assayed troponin I (hsTnI) were determined along with electrocardiography (ECG) variables. During a median follow-up of 183 days, 60 major adverse cardiovascular events (MACEs; 3.8%), including all-cause death, myocardial infarction, or stroke, were registered in the overall study population and 43 MACEs (7.5%) in the ACS subgroup. Adjusted multivariable Cox regression analyses revealed that an increase of 1 SD of plasma log-transformed homoarginine (0.37) was associated with a hazard reduction of 26% (hazard ratio [HR], 0.74; 95% CI, 0.57-0.96) for incident MACE and likewise of 35% (HR, 0.65; 95% CI, 0.49-0.88) in ACS patients. In Kaplan-Meier survival curves, homoarginine was predictive for patients with high-sensitivity assayed troponin I (hsTnI) above 27 ng/L (P<0.05). Last, homoarginine was inversely associated with QTc duration (P<0.001) and prevalent AF (OR, 0.83; 95% CI, 0.71-0.95). CONCLUSION Low plasma homoarginine was identified as a risk marker for incident MACEs in patients with acute chest pain, in particular, in those with elevated hsTnI. Impaired homoarginine was associated with prevalent AF. Further studies are needed to investigate the link to AF and evaluate homoarginine as a therapeutic option for these patients.
Collapse
Affiliation(s)
- Dorothee Atzler
- Department of Cardiovascular Medicine, University of Oxford, United Kingdom German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Christina Baum
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany Department of General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francisco Ojeda
- Department of General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Keller
- Division of Cardiology, Department of Medicine III, Goethe University Frankfurt, Frankfurt, Germany German Center for Cardiovascular Research (DZHK), Partner Site Rhein/Main, Germany
| | - Kathrin Cordts
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Renate B Schnabel
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany Department of General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Chi-un Choe
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karl J Lackner
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein/Main, Germany Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thomas Münzel
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein/Main, Germany Department of Medicine II, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Rainer H Böger
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Blankenberg
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany Department of General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edzard Schwedhelm
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Zeller
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany Department of General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|