1
|
Zhao Z, Wei TT, Zhang WX, Zhang SS, Wu R, Li F, Yang H, Zhang Q, Xi J, Zhou Y, Wang T, Du J, Lu QB, Ge Q. Association of homoarginine with arginine and disease severity in COVID-19 patients. Amino Acids 2025; 57:24. [PMID: 40332615 PMCID: PMC12058869 DOI: 10.1007/s00726-025-03453-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025]
Abstract
This study explored the relationship between the concentrations of homoarginine and arginine and between homoarginine concentration and laboratory parameters in coronavirus disease 2019 (COVID-19) patients with different severity to demonstrate the role of homoarginine in the progress of COVID-19. The laboratory-confirmed COVID-19 patients were included from Peking University Third Hospital during December 2022 to January 2023. Serum, urine, and stool samples were collected from the patients and detected by liquid chromatography-mass spectrometry. Totally 46 patients were recruited, including 18 in the mild group, 19 in the severe group, and 9 fatal. The concentration of homoarginine was positively correlated with the concentration of arginine in serum (r = 0.50), urine (r = 0.55), and stool samples (r = 0.39), respectively (all P < 0.001). The serum concentration and urine concentration of homoarginine were lower in severe patients than in mild patients (both P < 0.05). 13 indicators reflecting immunity and coagulation, including but not limited to T cell, white blood cell, natural killer cell, interleukin 6 (IL-6), and IL-8, had statistically significant correlations with both disease severity and the homoarginine concentration. Patients with hypertension were significantly associated with the decreased serum homoarginine (odds ratio 10.905, 95% confidence interval 1.454 - 137.144). Our results suggest that the homoarginine plays a role in the progress of COVID-19, which may be achieved by influencing arginine metabolism.
Collapse
Affiliation(s)
- Zhiling Zhao
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Ting-Ting Wei
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Wan-Xue Zhang
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Shan-Shan Zhang
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Rui Wu
- Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Fei Li
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Han Yang
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Qiang Zhang
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Jingjing Xi
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Yiguo Zhou
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China
| | - Tiehua Wang
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Juan Du
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Qing-Bin Lu
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China.
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China.
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China.
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
- Center for Infectious Disease and Policy Research and Department of Laboratorial of Science and Technology, School of Public Health, Peking University, No 38 Xue-Yuan Rd, Haidian District, Beijing, 100191, China.
| | - Qinggang Ge
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China.
| |
Collapse
|
2
|
Li J, Wu X, Fu Y, Liu J, Liu Y, Li J, Qing B, Zhang Y, Gao J, He X, Wang J, Li G. Transcriptomic and metabolomic insights into neutrophil activity in COPD complicated by metabolic syndrome. Biomed Eng Online 2025; 24:43. [PMID: 40234868 PMCID: PMC11998468 DOI: 10.1186/s12938-025-01378-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
OBJECTIVES Chronic obstructive pulmonary disease (COPD) frequently coexists with metabolic syndrome (MS), compounding its impact on patients' health and quality of life. This study aimed to elucidate the immune and metabolic response characteristics in COPD patients with and without MS. METHODS A total of 11,315 COPD patients admitted to the Department of Respiratory and Critical Care Medicine at the Third People's Hospital of Chengdu between January 1, 2013, and May 1, 2023, were selected. Multivariate logistic regression was conducted to identify the risk factors for acute exacerbation of chronic obstructive pulmonary disease. Moreover, from this cohort, 30 patients (18 with COPD and 12 with COPD-MS) were recruited for a further study to investigate the underlying mechanisms of COPD and COPD-MS. Blood samples were collected from these participants to perform transcriptomic and metabolomic analyses, aiming to explore the differences in immune responses and metabolic alterations between the two groups. RESULTS Our findings indicate a significant enhancement of neutrophil-mediated immune responses in COPD-MS patients. Transcriptomic analysis revealed 327 differentially expressed genes (DEGs) significantly involved in neutrophil-mediated immunity. Key metabolic pathways were disrupted, with 39 differential metabolites identified. Notably, metabolites, such as L-homoarginine and diethanolamine, which were elevated in COPD-MS patients, showed strong correlations with DEGs involved in neutrophil pathways and immune checkpoint regulation. The study also found decreased levels of IL4 and IL5RA in COPD-MS patients, suggesting a shift from Th2 to Th1 inflammatory responses, potentially contributing to glucocorticoid resistance. CONCLUSIONS COPD patients with metabolic syndrome exhibit a heightened neutrophil-mediated inflammatory response and significant metabolic disturbances, which underscores the need for precise therapeutic strategies targeting both metabolic and inflammatory pathways to improve patient outcomes and manage COPD-MS complexities effectively.
Collapse
Affiliation(s)
- Juan Li
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China
| | - Xue Wu
- Clinical Medicine Department, North Sichuan Medical College, Nanchong, China
- Department of Respiratory and Critical Care Medicine, Hospital of Shimian County, Ya'an, China
| | - Yufen Fu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Respiratory and Critical Care Medicine, Longchang People's Hospital, Neijiang, China
| | - Jiliu Liu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China
| | - Yao Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Jiahuan Li
- Clinical Medicine Department, North Sichuan Medical College, Nanchong, China
| | - Bomiao Qing
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China
| | - Yi Zhang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China
| | - Jie Gao
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China
| | - Xiang He
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China.
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China.
| | - Junyi Wang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China.
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China.
- Clinical Medicine Department, North Sichuan Medical College, Nanchong, China.
| | - Guoping Li
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China.
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China.
- Clinical Medicine Department, North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
3
|
Dinges SMT, Schwedhelm E, Schoenfeld J, Gevaert AB, Winzer EB, Haller B, Baldassarri F, Pressler A, Duvinage A, Böger R, Linke A, Adams V, Pieske B, Edelmann F, Dalen H, Hole T, Larsen AI, Feiereisen P, Karlsen T, Prescott E, Ellingsen Ø, Van Craenenbroeck EM, Halle M, Mueller S. Effects of exercise training on nitric oxide metabolites in heart failure with reduced or preserved ejection fraction: a secondary analysis of the SMARTEX-HF and OptimEx-Clin trials. Eur J Prev Cardiol 2025:zwaf142. [PMID: 40083304 DOI: 10.1093/eurjpc/zwaf142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/29/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
AIMS Exercise has been shown to affect the nitric oxide (NO) pathway, which is involved in the pathophysiology of endothelial dysfunction in heart failure (HF) with reduced (HFrEF) and preserved ejection fraction (HFpEF). However, the effects of different exercise modes on NO metabolites in patients with HF are uncertain. METHODS Blood samples from two randomized controlled HF trials evaluating 1.) high-intensity-interval-training (HIIT), 2.) moderate-continuous-training (MCT) or 3.) a control group (CG) in HFrEF (SMARTEX-HF) and HFpEF (OptimEx-Clin) were analysed for NO metabolites L-arginine, homoarginine (hArg), asymmetric and symmetric dimethylarginine (ADMA; SDMA). Metabolite plasma concentrations were compared between HFrEF and HFpEF at baseline and within each HF type after 3 months of supervised exercise training and 12 month-follow-up. RESULTS Overall, 206 patients with HFrEF (61±12 years, 18.9% females) and 160 with HFpEF (70±8 years, 65.6% females) were investigated. Baseline hArg (1.74±0.78 vs. 1.31±0.69 µmol/l) and ADMA (0.68±0.15 vs. 0.62±0.09 µmol/l) were significantly higher in HFrEF (p<0.001). NO metabolites showed several significant associations with markers of HF severity like exercise capacity (VO2peak) and NT-proBNP, but not with measures of endothelial function (reactive hyperaemia index, flow-mediated dilation). After 3 months of exercise and 12-month-follow-up, changes in metabolite plasma levels were not significantly different between study groups (HIIT, MCT or CG) (pgroup*time >0.05), neither in HFrEF nor HFpEF. CONCLUSION Baseline NO metabolite profile was unfavourable in patients with HF and lower VO2peak or higher NT-proBNP. We did not find a significant influence of HIIT or MCT on NO metabolites at 3 and 12 months.
Collapse
Affiliation(s)
- Sophia Marie-Theres Dinges
- Technical University of Munich, School of Medicine and Health, Department for Preventive Sports Medicine and Sports Cardiology, TUM University Hospital, Georg-Brauchle-Ring 56, 80992 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Potsdamer Str. 58, 10785 Berlin, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Potsdamer Str. 58, 10785 Berlin, Germany
| | - Julia Schoenfeld
- Technical University of Munich, School of Medicine and Health, Department for Preventive Sports Medicine and Sports Cardiology, TUM University Hospital, Georg-Brauchle-Ring 56, 80992 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Potsdamer Str. 58, 10785 Berlin, Germany
| | - Andreas B Gevaert
- Department of Cardiology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium
- Research Group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Ephraim B Winzer
- Department of Internal Medicine/Cardiology, University Clinic, Heart Center, Technische Universität Dresden, Fetscherstr. 76, 01307 Dresden, Germany
| | - Bernhard Haller
- Technical University of Munich, School of Medicine and Health, Institute for AI and Informatics in Medicine, TUM University Hospital, Ismaninger Str. 22, 81675 Munich, Germany
| | - Flavia Baldassarri
- Technical University of Munich, School of Medicine and Health, Department for Preventive Sports Medicine and Sports Cardiology, TUM University Hospital, Georg-Brauchle-Ring 56, 80992 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Potsdamer Str. 58, 10785 Berlin, Germany
| | - Axel Pressler
- Technical University of Munich, School of Medicine and Health, Department for Preventive Sports Medicine and Sports Cardiology, TUM University Hospital, Georg-Brauchle-Ring 56, 80992 Munich, Germany
- Private Center for Sports & Preventive Cardiology, Törringstraße 6, 81675 Munich, Germany
| | - André Duvinage
- Technical University of Munich, School of Medicine and Health, Department for Preventive Sports Medicine and Sports Cardiology, TUM University Hospital, Georg-Brauchle-Ring 56, 80992 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Potsdamer Str. 58, 10785 Berlin, Germany
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Potsdamer Str. 58, 10785 Berlin, Germany
| | - Axel Linke
- Department of Internal Medicine/Cardiology, University Clinic, Heart Center, Technische Universität Dresden, Fetscherstr. 76, 01307 Dresden, Germany
| | - Volker Adams
- Department of Internal Medicine/Cardiology, University Clinic, Heart Center, Technische Universität Dresden, Fetscherstr. 76, 01307 Dresden, Germany
| | - Burkert Pieske
- Division of Cardiology, Department of Internal Medicine, University Medicine Rostock, Schillingallee 35, 18057 Rostock, Germany
| | - Frank Edelmann
- Department of Cardiology, Angiology and Intensive Care Medicine, Campus Virchow Klinikum, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Potsdamer Str. 58, 10785 Berlin, Germany
| | - Håvard Dalen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postbox 8905, 7491 Trondheim, Norway
- Clinic of Cardiology, St. Olavs University Hospital, Postbox 3250 Torgarden, 7006 Trondheim, Norway
- Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Postbox 333, 7601 Levanger, Norway
| | - Torstein Hole
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postbox 8905, 7491 Trondheim, Norway
- Department of Medicine, Ålesund Hospital, Møre og Romsdal Hospital Trust, Postbox 1600, 6026 Ålesund, Norway
| | - Alf Inge Larsen
- Department of Cardiology, Stavanger University Hospital, Gerd Ragna Bloch Thorsens gate 8, 4011 Stavanger, Norway
- Institute of Clinical Science, University of Bergen, Jonas Lies vei 87, 5021 Bergen, Norway
| | - Patrick Feiereisen
- Department of Cardiology, Centre Hospitalier de Luxembourg, 4, Rue Nicolas Ernest Barblé, 1210 Luxembourg, Luxembourg
| | - Trine Karlsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postbox 8905, 7491 Trondheim, Norway
- Faculty of Nursing and Health Sciences, Nord University, Universitetsalléen 1, 8026 Bodø, Norway
| | - Eva Prescott
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Bispebjerg Bakke 23, Copenhagen 2400, Denmark
| | - Øyvind Ellingsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postbox 8905, 7491 Trondheim, Norway
- Clinic of Cardiology, St. Olavs University Hospital, Postbox 3250 Torgarden, 7006 Trondheim, Norway
| | - Emeline M Van Craenenbroeck
- Department of Cardiology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium
- Research Group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Martin Halle
- Technical University of Munich, School of Medicine and Health, Department for Preventive Sports Medicine and Sports Cardiology, TUM University Hospital, Georg-Brauchle-Ring 56, 80992 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Potsdamer Str. 58, 10785 Berlin, Germany
| | - Stephan Mueller
- Technical University of Munich, School of Medicine and Health, Department for Preventive Sports Medicine and Sports Cardiology, TUM University Hospital, Georg-Brauchle-Ring 56, 80992 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Potsdamer Str. 58, 10785 Berlin, Germany
| |
Collapse
|
4
|
Kuukkanen I, Pietikäinen A, Rissanen T, Hurme S, Kortela E, Kanerva MJ, Oksi J, Hytönen J, Karonen M. UHPLC-MS/MS-based untargeted metabolite profiling of Lyme neuroborreliosis. Sci Rep 2025; 15:8442. [PMID: 40069240 PMCID: PMC11897164 DOI: 10.1038/s41598-025-92189-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
The diagnosis of Lyme neuroborreliosis (LNB) requires the demonstration of intrathecal synthesis of Borrelia antibodies in a patient's cerebrospinal fluid (CSF), which involves the invasive procedure of a lumbar puncture. This study serves as a feasibility study aimed at exploring the potential of using serum samples, which are easily obtainable routine clinical samples, for LNB diagnostics via advanced metabolomics techniques. Serum samples were collected from confirmed LNB patients before and after treatment, with post-treatment samples serving as controls. The objective of the study was to find stable biomarkers for acute LNB through untargeted metabolomics analysis using ultrahigh-performance liquid chromatography coupled with tandem mass spectrometry (UHPLC-MS/MS). The study focused on biomarkers that could be reliably detected in serum samples stored under typical clinical conditions, without the need for special handling, ensuring consistent detection over time. The analysis revealed 26,978 molecular features (MFs), of which 1,746 were statistically significant (p < 0.001). Further manual investigation into 91 of the most prominent MFs revealed 53 potential biomarkers for LNB, individually or in combination. The workflow developed provides a comprehensive platform for biomarker detection, with potential applications in both research and clinical settings for LNB and other infections. This minimally invasive diagnostic approach is promising, and additional validation and future studies are needed for it to be considered as a practical alternative or a complement to CSF-based diagnostics of LNB in everyday clinical practice.
Collapse
Affiliation(s)
- Ilari Kuukkanen
- Department of Chemistry, University of Turku, Turku, Finland.
- TBD Turku, University of Turku, Turku, Finland.
| | - Annukka Pietikäinen
- TBD Turku, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Tyks Laboratories, Turku University Hospital, Turku, Finland
| | - Tiia Rissanen
- Department of Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | - Saija Hurme
- Department of Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | | | - Mari J Kanerva
- Department of Infectious Diseases, Turku University Hospital and University of Turku, Turku, Finland
| | - Jarmo Oksi
- Department of Infectious Diseases, Turku University Hospital and University of Turku, Turku, Finland
| | - Jukka Hytönen
- TBD Turku, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Tyks Laboratories, Turku University Hospital, Turku, Finland
| | - Maarit Karonen
- Department of Chemistry, University of Turku, Turku, Finland
- TBD Turku, University of Turku, Turku, Finland
| |
Collapse
|
5
|
Bingöl G, Huraıbat A, Ayduk Gövdeli E, Ser ÖS, Ünlü S, Çelik M, Bulut L, Özden Ö, Özmen E, Kılıçkesmez K. Effect of Homoarginine on Coronary Artery Complexity and Atherosclerotic Burden in Patients with STEMI. J Clin Med 2025; 14:1501. [PMID: 40094973 PMCID: PMC11900044 DOI: 10.3390/jcm14051501] [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: 01/13/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
Objective: Homoarginine is a cationic amino acid derived from lysine. Evidence indicates that low-circulating homoarginine concentration is a risk factor for cardiovascular disease and all-cause mortality. A reduction in homoarginine concentrations has been observed in patients with ischemic heart disease, ischemic stroke, ischemic heart disease, and heart failure. The SYNTAX score (SS), an angiographic scoring system, defines the grade and complexity of coronary artery disease (CAD). The objective of this study was to evaluate the relationship between homoarginine level and the severity of CAD according to the SYNTAX score in patients with ST-segment elevation myocardial infarction (STEMI). Methods: A total of 67 subjects were enrolled into the study with the diagnosis of STEMI of those who underwent coronary angiography. STEMI patients were divided into two groups: low-medium SYNTAX score ≤ 14 (35 patients) and high SYNTAX score > 14 (32 patients). Results: Within the high SS group, serum homoarginine levels were markedly lower (2 ± 0.9 vs. 1.3 ± 0.7; p = 0.001). Homoarginine levels and SS showed a significant negative correlation in entire study cohort In multivariate regression analysis, serum homoarginine levels along with serum urea levels were significantly associated with having higher SS (OR 1.073 p = 0.049 and OR 0.346, p = 0.012, respectively). Conclusions: In conclusion, the diminished plasma homoarginine level emerges as an independent predictor of high atherosclerotic burden among STEMI patients. To the best of our knowledge, this is the first study to demonstrate the relationship between homoarginine and coronary artery complexity.
Collapse
Affiliation(s)
- Gülsüm Bingöl
- Department of Cardiology, Arel University Medicine Faculty, 34010 Istanbul, Turkey
- Department of Cardiology, Memorial Bahcelievler Hospital, 34180 Istanbul, Turkey
| | - Ahmad Huraıbat
- Department of Cardiology, Prof. Dr. Cemil Tascıoğlu City Hospital, 34384 Istanbul, Turkey
| | - Elif Ayduk Gövdeli
- Royal Brompton Hospital Guy's and St Thomas' NHS Foundation Trust, London SW3 6PY, UK
| | - Özgür Selim Ser
- Department of Cardiology, Prof. Dr. Cemil Tascıoğlu City Hospital, 34384 Istanbul, Turkey
| | - Serkan Ünlü
- Department of Cardiology, Gazi University Medical Faculty, 06500 Ankara, Turkey
| | - Murat Çelik
- Department of Biochemistry, Yıldız Teknik University, 34220 Istanbul, Turkey
| | - Leyla Bulut
- Department of Biochemistry, Prof. Dr. Suleyman Yalcın City Hospital, 34722 Istanbul, Turkey
| | - Özge Özden
- Department of Cardiology, Memorial Bahcelievler Hospital, 34180 Istanbul, Turkey
| | - Emre Özmen
- Department of Cardiology, Memorial Bahcelievler Hospital, 34180 Istanbul, Turkey
| | - Kadriye Kılıçkesmez
- Department of Cardiology, Prof. Dr. Cemil Tascıoğlu City Hospital, 34384 Istanbul, Turkey
| |
Collapse
|
6
|
Mu Y, Wang Y, Wang S, Zhang X, Gu S, Zuo H. Associations of plasma arginine, homoarginine, and ADMA/SDMA levels with risk of ischemic stroke: A nested case-control study. Nutr Metab Cardiovasc Dis 2025; 35:103711. [PMID: 39277534 DOI: 10.1016/j.numecd.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/28/2024] [Accepted: 08/05/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND AND AIMS Previous studies have linked aberrant nitric oxide (NO) metabolism with vascular diseases. Although arginine, homoarginine, asymmetric dimethylarginine (ADMA), and symmetric dimethylarginine (SDMA) are involved in NO metabolic pathways, their associations with ischemic stroke (IS) remain unclear. METHODS AND RESULTS We conducted a case-control study nested within the Prospective Follow-up Study on Cardiovascular Morbidity and Mortality in China (PFS-CMMC) (2013-2018, n = 16,457; median follow-up time: 5.3 y), which included 321 incident cases of IS and 321 controls matched by age and sex. Plasma arginine, homoarginine, ADMA/SDMA were measured by ultrahigh performance liquid chromatography-tandem mass spectrometry. Conditional logistic regression analyses were used to calculate odds ratios (ORs) and their 95% confidence intervals (CIs) for the association between the plasma metabolites and IS risk. After adjustment for body mass index, educational attainment, smoking, hypertension, hyperlipidemia, diabetes, and family history of stroke, the OR of IS risk for the highest versus the lowest quartile was 2.46 (95% CI: 1.39-4.35, P trend = 0.004) for homoarginine and 2.22 (95% CI: 1.24-3.97, P trend = 0.003) for ADMA/SDMA. Spline regression analyses indicated positive dose-response relationships of homoarginine and ADMA/SDMA with the IS risk (both P for linearity <0.05). No significant association was observed between plasma arginine and IS risk. CONCLUSIONS Elevated plasma levels of homoarginine and ADMA/SDMA were associated with a higher risk of IS. Our novel findings suggest a role of NO metabolism in the pathogenesis of IS.
Collapse
Affiliation(s)
- Yingjun Mu
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yiqing Wang
- Suzhou Medical College of Soochow University, Suzhou, China
| | - Shujie Wang
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Xinyi Zhang
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Shujun Gu
- Department of Chronic Disease Control and Prevention, Changshu Center for Disease Control and Prevention, Suzhou, China
| | - Hui Zuo
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, Suzhou Medical College of Soochow University, Suzhou, China; MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
7
|
Zubkowski A, Sferruzzi‐Perri AN, Wishart DS. Mechanisms of Homoarginine: Looking Beyond Clinical Outcomes. Acta Physiol (Oxf) 2025; 241:e14273. [PMID: 39817883 PMCID: PMC11737358 DOI: 10.1111/apha.14273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/31/2024] [Accepted: 01/01/2025] [Indexed: 01/18/2025]
Abstract
PURPOSE Homoarginine (hArg) is an arginine metabolite that has been known for years, but its physiological role in the body remains poorly understood. For instance, it is well known that high hArg concentrations in the blood are protective against several disease states, yet the mechanisms behind these health benefits are unclear. This review compiles what is known about hArg, namely its synthetic pathways, its role in different diseases and conditions, and its proposed mechanisms of action in humans and experimental animals. FINDINGS Previous work has identified multiple pathways that control hArg synthesis and degradation in the body. Furthermore, endogenous hArg can modulate the cardiovascular system, with decreased hArg being associated with cardiovascular complications and increased mortality. Studies also suggest that hArg could serve as a diagnostic biomarker for a variety of immune, pancreatic, renal, and hepatic dysfunctions. Finally, in women, hArg concentrations rapidly increase throughout pregnancy and there are suggestions that alterations in hArg could indicate pregnancy complications like pre-eclampsia. SUMMARY Homoarginine is an under-appreciated amino acid with potential wide-ranging roles in systemic health, pregnancy, and pathophysiology. Although recent research has focused on its health or disease associations, there is a need for more investigations into understanding the mechanistic pathways by which hArg may operate. This could be aided using metabolomics, which provides a comprehensive approach to correlating multiple metabolites and metabolic pathways with physiological effects. Increasing our knowledge of hArg's roles in the body could pave the way for its routine use as both a diagnostic and therapeutic molecule.
Collapse
Affiliation(s)
- Ashley Zubkowski
- Department of Biological SciencesUniversity of AlbertaEdmontonAlbertaCanada
| | - Amanda N. Sferruzzi‐Perri
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - David S. Wishart
- Department of Biological SciencesUniversity of AlbertaEdmontonAlbertaCanada
- Department of Computer SciencesUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
8
|
Baranyi A, Meinitzer A, von Lewinski D, Wagner-Skacel J, Garcia SL, Rothenhäusler HB, Amouzadeh-Ghadikolai O, Harpf L, Schweinzer M, Enko D. Homoarginine concentrations correlate with early depressive symptoms and the reduction in physical functioning within the first days after myocardial infarction. Sci Rep 2025; 15:1520. [PMID: 39789030 PMCID: PMC11718095 DOI: 10.1038/s41598-024-84930-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Early depressive symptoms within the first days after acute myocardial infarction (AMI) are mainly manifested with performance parameters (lack of energy, concentration difficulties, reduction in physical functioning). Homoarginine (hArg), a non-proteinogenic amino acid, might increase the availability of nitric oxide (NO). NO controls vasodilatation, blood flow, mitochondrial respiration and improves performance. Therefore, low plasma hArg levels after an AMI might impact performance-related early depressive symptoms. This longitudinal study aims to determine the course of plasma hArg concentrations immediately, on the fourth day and 6 months after AMI and investigates the associations between hArg and early depressive symptoms. A decrease in hArg levels, as observed in AMI patients on the fourth day after AMI, was independent of gender, age, body-mass-index and AMI type. After six months, hArg concentrations no longer differed significantly from baseline values. Females had lower hArg concentrations shortly after and also four days after the AMI compared to males. Within the first days after AMI HAMD-17 and BDI-II total depression scores and performance-related early depressive symptoms such as lack of energy, concentration difficulties and reduction in physical functioning correlated with low hArg concentrations.
Collapse
Affiliation(s)
- Andreas Baranyi
- Department of Psychiatry, Psychosomatics and Psychotherapeutic Medicine, Division of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, 8036, Graz, Austria.
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036, Graz, Austria
| | - Dirk von Lewinski
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8036, Graz, Austria.
| | - Jolana Wagner-Skacel
- Department of Psychiatry, Psychosomatics and Psychotherapeutic Medicine, Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, 8036, Graz, Austria
| | - Sabrina Leal Garcia
- Department of Psychiatry, Psychosomatics and Psychotherapeutic Medicine, Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, 8036, Graz, Austria
| | - Hans-Bernd Rothenhäusler
- Department of Psychiatry, Psychosomatics and Psychotherapeutic Medicine, Division of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, 8036, Graz, Austria
| | | | | | - Melanie Schweinzer
- Department of Psychiatry, Psychosomatics and Psychotherapeutic Medicine, Division of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, 8036, Graz, Austria
| | - Dietmar Enko
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036, Graz, Austria
| |
Collapse
|
9
|
Wu Y, Avcilar-Kücükgöze I, Santovito D, Atzler D. Amino Acid Metabolism and Autophagy in Atherosclerotic Cardiovascular Disease. Biomolecules 2024; 14:1557. [PMID: 39766264 PMCID: PMC11673637 DOI: 10.3390/biom14121557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Cardiovascular disease is the most common cause of mortality globally, accounting for approximately one out of three deaths. The main underlying pathology is atherosclerosis, a dyslipidemia-driven, chronic inflammatory disease. The interplay between immune cells and non-immune cells is of great importance in the complex process of atherogenesis. During atheroprogression, intracellular metabolic pathways, such as amino acid metabolism, are master switches of immune cell function. Autophagy, an important stress survival mechanism involved in maintaining (immune) cell homeostasis, is crucial during the development of atherosclerosis and is strongly regulated by the availability of amino acids. In this review, we focus on the interplay between amino acids, especially L-leucine, L-arginine, and L-glutamine, and autophagy during atherosclerosis development and progression, highlighting potential therapeutic perspectives.
Collapse
Affiliation(s)
- Yuting Wu
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, 80336 Munich, Germany; (Y.W.); (I.A.-K.)
| | - Irem Avcilar-Kücükgöze
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, 80336 Munich, Germany; (Y.W.); (I.A.-K.)
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Donato Santovito
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, 80336 Munich, Germany; (Y.W.); (I.A.-K.)
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
- Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, 20133 Milan, Italy
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, 80336 Munich, Germany; (Y.W.); (I.A.-K.)
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
- Walter Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| |
Collapse
|
10
|
Smith BI, Vásquez-Hidalgo MA, Li X, Vonnahme KA, Grazul-Bilska AT, Swanson KC, Moore TE, Reed SA, Govoni KE. The Effects of Maternal Nutrient Restriction during Mid to Late Gestation with Realimentation on Fetal Metabolic Profiles in the Liver, Skeletal Muscle, and Blood in Sheep. Metabolites 2024; 14:465. [PMID: 39330472 PMCID: PMC11434268 DOI: 10.3390/metabo14090465] [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: 07/19/2024] [Revised: 08/09/2024] [Accepted: 08/10/2024] [Indexed: 09/28/2024] Open
Abstract
Poor maternal nutrition during gestation negatively affects offspring growth and metabolism. To evaluate the impact of maternal nutrient restriction and realimentation on metabolism in the fetal liver, skeletal muscle, and circulation, on day 50 of gestation, ewes (n = 48) pregnant with singletons were fed 100% (CON) or 60% (RES) of requirements until day 90 of gestation, when a subset of ewes (n = 7/treatment) were euthanized, and fetal samples were collected. The remaining ewes were maintained on a current diet (CON-CON, n = 6; RES-RES, n = 7) or switched to an alternative diet (CON-RES, RES-CON; n = 7/treatment). On day 130 of gestation, the remaining ewes were euthanized, and fetal samples were collected. Fetal liver, longissimus dorsi (LD), and blood metabolites were analyzed using LC-MS/MS, and pathway enrichment analysis was conducted using MetaboAnalyst. Then, 600, 518, and 524 metabolites were identified in the liver, LD, and blood, respectively, including 345 metabolites that were present in all three. Nutrient restriction was associated with changes in amino acid, carbohydrate, lipid, and transulfuration/methionine metabolic pathways, some of which were alleviated by realimentation. Fetal age also affected metabolite abundance. The differential abundance of metabolites involved in amino acid, methionine, betaine, and bile acid metabolism could impact fetal epigenetic regulation, protein synthesis, lipid metabolism, and signaling associated with glucose and lipid metabolism.
Collapse
Affiliation(s)
- Brandon I. Smith
- Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA (S.A.R.)
| | - Manuel A. Vásquez-Hidalgo
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA; (M.A.V.-H.); (A.T.G.-B.); (K.C.S.)
| | - Xiaomeng Li
- Department of Statistics, University of Connecticut, Storrs, CT 06269, USA (T.E.M.)
| | - Kimberly A. Vonnahme
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA; (M.A.V.-H.); (A.T.G.-B.); (K.C.S.)
| | - Anna T. Grazul-Bilska
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA; (M.A.V.-H.); (A.T.G.-B.); (K.C.S.)
| | - Kendall C. Swanson
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58102, USA; (M.A.V.-H.); (A.T.G.-B.); (K.C.S.)
| | - Timothy E. Moore
- Department of Statistics, University of Connecticut, Storrs, CT 06269, USA (T.E.M.)
| | - Sarah A. Reed
- Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA (S.A.R.)
| | - Kristen E. Govoni
- Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA (S.A.R.)
| |
Collapse
|
11
|
Ouyang X, Wahlsten M, Pollari M, Delbaje E, Jokela J, Fewer DP. Identification of a homoarginine biosynthetic gene from a microcystin biosynthetic pathway in Fischerella sp. PCC 9339. Toxicon 2024; 243:107733. [PMID: 38670499 DOI: 10.1016/j.toxicon.2024.107733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/02/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Microcystins (MCs) are a family of chemically diverse toxins produced by numerous distantly related cyanobacteria. They are potent inhibitors of eukaryotic protein phosphatases 1 and 2A and are responsible for the toxicosis and death of wild and domestic animals around the world. Microcystins are synthesized on large enzyme complexes comprised of peptide synthetases, polyketide synthases, and additional modifying enzymes. Bioinformatic analysis identified the presence of an additional uncharacterized enzyme in the microcystin (mcy) biosynthetic gene cluster in Fischerella sp. PCC 9339, which we named McyK, that lacked a clearly defined role in the biosynthesis of microcystin. Further bioinformatic analysis suggested that McyK belongs to the inosamine-phosphate amidinotransferase family and could be involved in synthesizing homo amino acids. Quadrupole time-of-flight tandem mass spectrometry (Q-TOFMS/MS) analysis confirmed that Fischerella sp. PCC 9339 produces MC-Leucine2-Homoarginine4(MC-LHar) and [Aspartic acid3]MC-Leucine2-Homoarginine4 ([Asp3]MC-LHar) as the dominant chemical variants. We hypothesized that the McyK enzyme might be involved in the production of microcystin variants containing homoarginine (Har) in the strain. Heterologous expression of a codon-optimized mcyK gene in Escherichia coli confirmed that McyK is responsible for the synthesis of L-Har. These results confirm the production of MC-LHar, a novel microcystin chemical variant [Asp3]MC-LHar, and a new microcystin biosynthetic enzyme involved in supply of the rare homo-amino acid Har to the microcystin biosynthetic pathway in Fischerella sp. PCC 9339. This study provides new insights into the logic underpinning the biosynthesis of microcystin chemical variants and broadens our knowledge of structural diversity of the microcystin family of toxins.
Collapse
Affiliation(s)
- Xiaodan Ouyang
- Department of Microbiology, University of Helsinki, Viikinkaari 9, FI-00014, Helsinki, Finland
| | - Matti Wahlsten
- Department of Microbiology, University of Helsinki, Viikinkaari 9, FI-00014, Helsinki, Finland
| | - Maija Pollari
- Department of Agricultural Sciences, University of Helsinki, Latokartanonkaari 5, FI-00014, Helsinki, Finland
| | - Endrews Delbaje
- Departamento de Ciências Farmacêuticas, Universidade de São Paulo, Avenida do Café S/N, 14040-903, Ribeirão Preto, Brazil
| | - Jouni Jokela
- Department of Microbiology, University of Helsinki, Viikinkaari 9, FI-00014, Helsinki, Finland
| | - David P Fewer
- Department of Microbiology, University of Helsinki, Viikinkaari 9, FI-00014, Helsinki, Finland.
| |
Collapse
|
12
|
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: 5] [Impact Index Per Article: 5.0] [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
|
13
|
Isailă OM, Moroianu LA, Hostiuc S. Current Trends in Biohumoral Screening for the Risk of Sudden Cardiac Death: A Systematic Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:418. [PMID: 38541144 PMCID: PMC10972295 DOI: 10.3390/medicina60030418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 07/23/2024]
Abstract
Background and Objectives: Sudden cardiac death (SCD) represents a challenge to health systems globally and is met with increased frequency in the population. Over time, multiple screening methods have been proposed, including the analysis of various plasma biomarkers. This article aims to analyze for illustrative purposes the specialized literature in terms of current biomarkers and testing trends, in the case of cardiovascular diseases and implicitly sudden cardiac death. Materials and Methods: In this regard, we searched the PubMed database from 2010 to the present time using the keywords "sudden cardiac death" and "biomarkers". The inclusion criteria were clinical trials that analyzed the effectiveness of screening methods in terms of biomarkers used in stratifying the risk of cardiac distress and/or sudden cardiac death. We excluded reviews, meta-analyses, and studies looking at the effectiveness of treatments. Results: An extended approach was found, through studies that brought to the forefront both classical markers analyzed by new, more performant methods, markers for other pathologies that also determined cardiovascular impact, non-specific molecules with effects on the cardiovascular system, and state-of-the-art markers, such as microRNA. Some molecules were analyzed simultaneously in certain groups of patients. Conclusion: The observed current trend revealed the tendency to define the clinical-biological particularities of the person to be screened.
Collapse
Affiliation(s)
- Oana-Maria Isailă
- Department of Legal Medicine and Bioethics, Faculty of Dentistry, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Lavinia-Alexandra Moroianu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 47 Domneasca Street, 800008 Galati, Romania;
| | - Sorin Hostiuc
- Department of Legal Medicine and Bioethics, Faculty of Dentistry, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
14
|
Petersen KS, Chandra M, Chen See JR, Leister J, Jafari F, Tindall A, Kris-Etherton PM, Lamendella R. Walnut consumption and gut microbial metabolism: Results of an exploratory analysis from a randomized, crossover, controlled-feeding study. Clin Nutr 2023; 42:2258-2269. [PMID: 37826992 DOI: 10.1016/j.clnu.2023.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND & AIMS The effect of walnut-related modulation of gut microbiota composition on microbiota functionality is unknown. The aim was to characterize the effect of a walnut-enriched diet (WD), compared to a fatty acid-matched diet devoid of walnuts (WFMD) and a diet where oleic acid replaces alpha-linolenic acid (ORAD), on bacterial gene expression. METHODS A 3-period, randomized, crossover, controlled-feeding study was conducted. Participants were provided a 2-week run-in standard western diet (SWD; 50% kcal carbohydrate, 16% protein, 34% fat, 12% SFA). Following the SWD in random sequence order, participants were provided the WD, WFMD, and ORAD (48% carbohydrate; 17% protein; fat 35%; 7% SFA). The WD contained 18% of energy from walnuts (57 g/d/2100 kcal). The WFMD and ORAD were devoid of walnuts; liquid non-tropical plant oils were included in these diets. Metatranscriptomic analyses were performed as an exploratory outcome. RESULTS The analytical sample included 35 participants (40% female) with a mean ± SD age of 43 ± 10 y and BMI of 30.3 ± 4.9 kg/m2. The ⍺-diversity of taxa actively expressing genes, assessed by observed species (p = 0.27) and Pielou's Evenness (p = 0.09), did not differ among the diets. The ⍺-diversity of actively expressed genes was greater following the WD compared to the WFMD and ORAD as assessed by the observed genes and Pielou's Evenness metrics (p < 0.05). β-Diversity of the actively expressed genes differed following the WD compared to the WFMD (p = 0.001) and ORAD (p = 0.001); β-diversity did not differ between the WFMD and ORAD. Active composition analyses showed increased Gordonibacter (p < 0.001) activity following the WD vs. the ORAD. Greater expression of many genes was observed following the WD compared to the WFMD and ORAD. Following the WD, greater expression of metabolism-related genes encoding glycine amidinotransferase (GATM; K00613) and arginine deiminase (K01478) was observed compared to the WFMD. Greater expression of glycine amidinotransferase (GATM; K00613) by Gordonibacter was also observed following the WD vs. the WFMD and ORAD. CONCLUSION Our results suggest walnut intake may increase endogenous production of homoarginine through gut microbiota-mediated upregulation of GATM, which is a novel mechanism by which walnuts may lower cardiovascular disease risk. However, given the exploratory nature replication is needed. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov (NCT02210767).
Collapse
Affiliation(s)
- Kristina S Petersen
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA.
| | - Mansi Chandra
- Department of Biology, Juniata College, Huntingdon, PA, USA; Wright Labs, LLC, Huntingdon, PA, USA
| | - Jeremy R Chen See
- Department of Biology, Juniata College, Huntingdon, PA, USA; Wright Labs, LLC, Huntingdon, PA, USA
| | - Jillian Leister
- Department of Biology, Juniata College, Huntingdon, PA, USA; Wright Labs, LLC, Huntingdon, PA, USA
| | - Fatemeh Jafari
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Alyssa Tindall
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA; Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Regina Lamendella
- Department of Biology, Juniata College, Huntingdon, PA, USA; Wright Labs, LLC, Huntingdon, PA, USA
| |
Collapse
|
15
|
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
|
16
|
Reynolds KM, Horimoto ARVR, Lin BM, Zhang Y, Kurniansyah N, Yu B, Boerwinkle E, Qi Q, Kaplan R, Daviglus M, Hou L, Zhou LY, Cai J, Shaikh SR, Sofer T, Browning SR, Franceschini N. Ancestry-driven metabolite variation provides insights into disease states in admixed populations. Genome Med 2023; 15:52. [PMID: 37461045 PMCID: PMC10351197 DOI: 10.1186/s13073-023-01209-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 07/10/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Metabolic pathways are related to physiological functions and disease states and are influenced by genetic variation and environmental factors. Hispanics/Latino individuals have ancestry-derived genomic regions (local ancestry) from their recent admixture that have been less characterized for associations with metabolite abundance and disease risk. METHODS We performed admixture mapping of 640 circulating metabolites in 3887 Hispanic/Latino individuals from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). Metabolites were quantified in fasting serum through non-targeted mass spectrometry (MS) analysis using ultra-performance liquid chromatography-MS/MS. Replication was performed in 1856 nonoverlapping HCHS/SOL participants with metabolomic data. RESULTS By leveraging local ancestry, this study identified significant ancestry-enriched associations for 78 circulating metabolites at 484 independent regions, including 116 novel metabolite-genomic region associations that replicated in an independent sample. Among the main findings, we identified Native American enriched genomic regions at chromosomes 11 and 15, mapping to FADS1/FADS2 and LIPC, respectively, associated with reduced long-chain polyunsaturated fatty acid metabolites implicated in metabolic and inflammatory pathways. An African-derived genomic region at chromosome 2 was associated with N-acetylated amino acid metabolites. This region, mapped to ALMS1, is associated with chronic kidney disease, a disease that disproportionately burdens individuals of African descent. CONCLUSIONS Our findings provide important insights into differences in metabolite quantities related to ancestry in admixed populations including metabolites related to regulation of lipid polyunsaturated fatty acids and N-acetylated amino acids, which may have implications for common diseases in populations.
Collapse
Affiliation(s)
- Kaylia M Reynolds
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
- Department of Epidemiology, University of North Carolina, 123 W Franklin St, Suite 401, NC, NC 27516, Chapel Hill, USA
| | | | - Bridget M Lin
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Ying Zhang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Nuzulul Kurniansyah
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Bing Yu
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Martha Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Laura Y Zhou
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Saame Raza Shaikh
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Departments of Medicine and Biostatistics, Harvard University, Boston, MA, USA
| | - Sharon R Browning
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, 123 W Franklin St, Suite 401, NC, NC 27516, Chapel Hill, USA.
| |
Collapse
|
17
|
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 2023; 37:519-529. [PMID: 36509694 DOI: 10.1111/fcp.12858] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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
|
18
|
Watanabe K, Wilmanski T, Diener C, Earls JC, Zimmer A, Lincoln B, Hadlock JJ, Lovejoy JC, Gibbons SM, Magis AT, Hood L, Price ND, Rappaport N. Multiomic signatures of body mass index identify heterogeneous health phenotypes and responses to a lifestyle intervention. Nat Med 2023; 29:996-1008. [PMID: 36941332 PMCID: PMC10115644 DOI: 10.1038/s41591-023-02248-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 02/02/2023] [Indexed: 03/23/2023]
Abstract
Multiomic profiling can reveal population heterogeneity for both health and disease states. Obesity drives a myriad of metabolic perturbations and is a risk factor for multiple chronic diseases. Here we report an atlas of cross-sectional and longitudinal changes in 1,111 blood analytes associated with variation in body mass index (BMI), as well as multiomic associations with host polygenic risk scores and gut microbiome composition, from a cohort of 1,277 individuals enrolled in a wellness program (Arivale). Machine learning model predictions of BMI from blood multiomics captured heterogeneous phenotypic states of host metabolism and gut microbiome composition better than BMI, which was also validated in an external cohort (TwinsUK). Moreover, longitudinal analyses identified variable BMI trajectories for different omics measures in response to a healthy lifestyle intervention; metabolomics-inferred BMI decreased to a greater extent than actual BMI, whereas proteomics-inferred BMI exhibited greater resistance to change. Our analyses further identified blood analyte-analyte associations that were modified by metabolomics-inferred BMI and partially reversed in individuals with metabolic obesity during the intervention. Taken together, our findings provide a blood atlas of the molecular perturbations associated with changes in obesity status, serving as a resource to quantify metabolic health for predictive and preventive medicine.
Collapse
Affiliation(s)
| | | | | | - John C Earls
- Institute for Systems Biology, Seattle, WA, USA
- Thorne HealthTech, New York, NY, USA
| | - Anat Zimmer
- Institute for Systems Biology, Seattle, WA, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- eScience Institute, University of Washington, Seattle, WA, USA
| | | | - Leroy Hood
- Institute for Systems Biology, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Phenome Health, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Nathan D Price
- Institute for Systems Biology, Seattle, WA, USA
- Thorne HealthTech, New York, NY, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
19
|
Mecha E, Alves ML, Bento da Silva A, Pereira AB, Rubiales D, Vaz Patto MC, Bronze MR. High Inter- and Intra- Diversity of Amino Acid Content and Protein Digestibility Disclosed in Five Cool Season Legume Species with a Growing Market Demand. Foods 2023; 12:foods12071383. [PMID: 37048201 PMCID: PMC10093753 DOI: 10.3390/foods12071383] [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: 02/19/2023] [Revised: 03/15/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Legumes have been sought as alternative protein sources to ensure food security and environmental sustainability. Characterizing their protein content and quality, including in underutilized grain legumes, e.g., grass pea, gives value to the legumes' underexplored variability. To fill the gap of knowledge in legumes' protein quality, for the first time, five extensive collections of cool season grain legumes were cropped under the same environmental conditions and further analyzed. Multivariate analysis showed the existent intra- and inter-species variability. The legume species with the highest protein content, grass pea, Lathyrus sativus (LS), was not the one with the overall highest individual amino acids content and in vitro protein digestibility. With these last characteristics lentil, Lens culinaris (LC), was highlighted. The highest average values of arginine (Arg), glutamic acid (Glu), and threonine (Thr) were found in LS and Vicia faba (VF). Cicer arietinum (CA) stood out as the species with the highest values of Thr and methionine (Met). Regarding the in vitro protein digestibility (IVPD), LC, followed by Pisum sativum (PS) and LS, were the legume species with the highest values. Ultimately, this study bought to the fore legume species that are not commonly used in western diets but have high adaptability to the European agricultural systems.
Collapse
Affiliation(s)
- Elsa Mecha
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Av. da República, Apartado 12, 2781-901 Oeiras, Portugal
| | - Mara Lisa Alves
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Andreia Bento da Silva
- Faculdade de Farmácia, Universidade de Lisboa, Av. das Forças Armadas, 1649-019 Lisboa, Portugal
| | - Ana Bárbara Pereira
- iBET, Instituto de Biologia Experimental e Tecnológica, Av. da República, Apartado 12, 2781-901 Oeiras, Portugal
| | - Diego Rubiales
- Institute for Sustainable Agriculture, CSIC, Avda Menéndez Pidal s/n, 14004 Córdoba, Spain
| | - Maria Carlota Vaz Patto
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Maria Rosário Bronze
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Av. da República, Apartado 12, 2781-901 Oeiras, Portugal
- Faculdade de Farmácia, Universidade de Lisboa, Av. das Forças Armadas, 1649-019 Lisboa, Portugal
| |
Collapse
|
20
|
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. [PMID: 35697514 DOI: 10.1111/fcp.12808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022]
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.
Collapse
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
| | - Thomas J Vogl
- 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
|
21
|
Surendran P, Stewart ID, Au Yeung VPW, Pietzner M, Raffler J, Wörheide MA, Li C, Smith RF, Wittemans LBL, Bomba L, Menni C, Zierer J, Rossi N, Sheridan PA, Watkins NA, Mangino M, Hysi PG, Di Angelantonio E, Falchi M, Spector TD, Soranzo N, Michelotti GA, Arlt W, Lotta LA, Denaxas S, Hemingway H, Gamazon ER, Howson JMM, Wood AM, Danesh J, Wareham NJ, Kastenmüller G, Fauman EB, Suhre K, Butterworth AS, Langenberg C. Rare and common genetic determinants of metabolic individuality and their effects on human health. Nat Med 2022; 28:2321-2332. [PMID: 36357675 PMCID: PMC9671801 DOI: 10.1038/s41591-022-02046-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 09/16/2022] [Indexed: 11/12/2022]
Abstract
Garrod's concept of 'chemical individuality' has contributed to comprehension of the molecular origins of human diseases. Untargeted high-throughput metabolomic technologies provide an in-depth snapshot of human metabolism at scale. We studied the genetic architecture of the human plasma metabolome using 913 metabolites assayed in 19,994 individuals and identified 2,599 variant-metabolite associations (P < 1.25 × 10-11) within 330 genomic regions, with rare variants (minor allele frequency ≤ 1%) explaining 9.4% of associations. Jointly modeling metabolites in each region, we identified 423 regional, co-regulated, variant-metabolite clusters called genetically influenced metabotypes. We assigned causal genes for 62.4% of these genetically influenced metabotypes, providing new insights into fundamental metabolite physiology and clinical relevance, including metabolite-guided discovery of potential adverse drug effects (DPYD and SRD5A2). We show strong enrichment of inborn errors of metabolism-causing genes, with examples of metabolite associations and clinical phenotypes of non-pathogenic variant carriers matching characteristics of the inborn errors of metabolism. Systematic, phenotypic follow-up of metabolite-specific genetic scores revealed multiple potential etiological relationships.
Collapse
Affiliation(s)
- Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | | | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Raffler
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Digital Medicine, University Hospital of Augsburg, Augsburg, Germany
| | - Maria A Wörheide
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Chen Li
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Rebecca F Smith
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Laura B L Wittemans
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Big Data Institute, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Lorenzo Bomba
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Open Targets, Wellcome Genome Campus, Hinxton, UK
| | - Cristina Menni
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Jonas Zierer
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Niccolò Rossi
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | | | | | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
- NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, UK
| | - Pirro G Hysi
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- Health Data Science Research Centre, Human Technopole, Milan, Italy
| | - Mario Falchi
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Tim D Spector
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Nicole Soranzo
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Open Targets, Wellcome Genome Campus, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | | | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Luca A Lotta
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Spiros Denaxas
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK, London, UK
- British Heart Foundation Data Science Centre, London, UK
| | - Harry Hemingway
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK, London, UK
| | - Eric R Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Clare Hall & MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Joanna M M Howson
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Angela M Wood
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
- The Alan Turing Institute, London, UK
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
| | - Nicholas J Wareham
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Eric B Fauman
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Karsten Suhre
- Department of Biophysics and Physiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK.
- NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK.
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK.
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
22
|
Nitz K, Lacy M, Bianchini M, Wichapong K, Kücükgöze IA, Bonfiglio CA, Migheli R, Wu Y, Burger C, Li Y, Forné I, Ammar C, Janjic A, Mohanta S, Duchene J, Heemskerk JWM, Megens RTA, Schwedhelm E, Huveneers S, Lygate CA, Santovito D, Zimmer R, Imhof A, Weber C, Lutgens E, Atzler D. The Amino Acid Homoarginine Inhibits Atherogenesis by Modulating T-Cell Function. Circ Res 2022; 131:701-712. [PMID: 36102188 DOI: 10.1161/circresaha.122.321094] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Amino acid metabolism is crucial for inflammatory processes during atherogenesis. The endogenous amino acid homoarginine is a robust biomarker for cardiovascular outcome and mortality with high levels being protective. However, the underlying mechanisms remain elusive. We investigated the effect of homoarginine supplementation on atherosclerotic plaque development with a particular focus on inflammation. METHODS Female ApoE-deficient mice were supplemented with homoarginine (14 mg/L) in drinking water starting 2 weeks before and continuing throughout a 6-week period of Western-type diet feeding. Control mice received normal drinking water. Immunohistochemistry and flow cytometry were used for plaque- and immunological phenotyping. T cells were characterized using mass spectrometry-based proteomics, by functional in vitro approaches, for example, proliferation and migration/chemotaxis assays as well as by super-resolution microscopy. RESULTS Homoarginine supplementation led to a 2-fold increase in circulating homoarginine concentrations. Homoarginine-treated mice exhibited reduced atherosclerosis in the aortic root and brachiocephalic trunk. A substantial decrease in CD3+ T cells in the atherosclerotic lesions suggested a T-cell-related effect of homoarginine supplementation, which was mainly attributed to CD4+ T cells. Macrophages, dendritic cells, and B cells were not affected. CD4+ T-cell proteomics and subsequent pathway analysis together with in vitro studies demonstrated that homoarginine profoundly modulated the spatial organization of the T-cell actin cytoskeleton and increased filopodia formation via inhibition of Myh9 (myosin heavy chain 9). Further mechanistic studies revealed an inhibition of T-cell proliferation as well as a striking impairment of the migratory capacities of T cells in response to relevant chemokines by homoarginine, all of which likely contribute to its atheroprotective effects. CONCLUSIONS Our study unravels a novel mechanism by which the amino acid homoarginine reduces atherosclerosis, establishing that homoarginine modulates the T-cell cytoskeleton and thereby mitigates T-cell functions important during atherogenesis. These findings provide a molecular explanation for the beneficial effects of homoarginine in atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Katrin Nitz
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (K.N., M.L., C.A.B., J.D., D.S., C.W., E.L., D.A.)
| | - Michael Lacy
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (K.N., M.L., C.A.B., J.D., D.S., C.W., E.L., D.A.).,Department of Medical Laboratory Sciences, Virginia Commonwealth University, Richmond (M.L.)
| | - Mariaelvy Bianchini
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Kanin Wichapong
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (K.W., J.W.M.H., C.W.)
| | - Irem Avcilar Kücükgöze
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Cecilia A Bonfiglio
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (K.N., M.L., C.A.B., J.D., D.S., C.W., E.L., D.A.)
| | - Roberta Migheli
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Yuting Wu
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Carina Burger
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Yuanfang Li
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Ignasi Forné
- Biomedical Center Munich, Department of Molecular Biology (I.F., A.I.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Constantin Ammar
- Institute of Bioinformatics, Department of Informatics (C.A., R.Z.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Aleksandar Janjic
- Anthropology & Human Genomics, Department of Biology II (A.J.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Sarajo Mohanta
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Johan Duchene
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (K.N., M.L., C.A.B., J.D., D.S., C.W., E.L., D.A.)
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (K.W., J.W.M.H., C.W.)
| | - Remco T A Megens
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany.,Department of Biomedical Engineering, CARIM, Maastricht University, Maastricht, the Netherlands (R.T.A.M.)
| | - Edzard Schwedhelm
- Department of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Germany (E.S.).,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Hamburg/Kiel/Lübeck, Germany (E.S.)
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam University Medical Centre, Amsterdam Cardiovascular Sciences, the Netherlands (S.H.)
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and the BHF Centre of Research Excellence, University of Oxford, United Kingdom (C.A.L.)
| | - Donato Santovito
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (K.N., M.L., C.A.B., J.D., D.S., C.W., E.L., D.A.)
| | - Ralf Zimmer
- Institute of Bioinformatics, Department of Informatics (C.A., R.Z.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Axel Imhof
- Biomedical Center Munich, Department of Molecular Biology (I.F., A.I.), Ludwig-Maximilians-Universität, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (K.N., M.L., C.A.B., J.D., D.S., C.W., E.L., D.A.).,Department of Medical Laboratory Sciences, Virginia Commonwealth University, Richmond (M.L.)
| | - Esther Lutgens
- DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (K.N., M.L., C.A.B., J.D., D.S., C.W., E.L., D.A.).,Department of Cardiovascular Medicine, Experimental Cardiovascular Immunology Laboratory, Mayo Clinic, Rochester, MN (E.L.)
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention (K.N., M.L., M.B., I.A.K., C.A.B., R.M., Y.W., C.B., Y.L., S.M., J.D., R.T.A.M., D.S., C.W., E.L., D.A.), Ludwig-Maximilians-Universität, Munich, Germany.,Walther Straub Institute of Pharmacology and Toxicology (D.A.), Ludwig-Maximilians-Universität, Munich, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Munich Heart Alliance, Munich, Germany (K.N., M.L., C.A.B., J.D., D.S., C.W., E.L., D.A.)
| |
Collapse
|
23
|
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
|
24
|
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: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
| | - Miguel Ruiz-Canela
- 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
| |
Collapse
|
25
|
Onmaz DE, Tezcan D, Abusoglu S, Yilmaz S, Kuzu M, Abusoglu G, H Yerlikaya F, Unlu A. Raised total methylated arginine load in patients with gout. Biomark Med 2022; 16:993-1004. [PMID: 36052727 DOI: 10.2217/bmm-2022-0368] [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: 11/21/2022] Open
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.
Collapse
Affiliation(s)
- Duygu Eryavuz Onmaz
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Dilek Tezcan
- Department of Internal Medicine, Division of Rheumatology, Gülhane Faculty of Medicine, University of Health Sciences Turkey, Ankara, Turkey
| | - Sedat Abusoglu
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Sema Yilmaz
- Department of Internal Medicine, Division of Rheumatology, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Menekse Kuzu
- Department of Biochemistry, Faculty of Pharmacy, Biruni University, Istanbul, Turkey
| | - Gulsum Abusoglu
- Department of Medical Laboratory Techniques, Selcuk University Vocational School of Health, Konya, Turkey
| | - Fatma H Yerlikaya
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Ali Unlu
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| |
Collapse
|
26
|
Fulghum K, Collins HE, Jones SP, Hill BG. Influence of biological sex and exercise on murine cardiac metabolism. JOURNAL OF SPORT AND HEALTH SCIENCE 2022; 11:479-494. [PMID: 35688382 PMCID: PMC9338340 DOI: 10.1016/j.jshs.2022.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/07/2022] [Accepted: 04/27/2022] [Indexed: 05/03/2023]
Abstract
Although the structural and functional effects of exercise on the heart are well established, the metabolic changes that occur in the heart during and after exercise remain unclear. In this study, we used metabolomics to assess time-dependent changes in the murine cardiac metabolome following 1 session of treadmill exercise. After the exercise bout, we also recorded blood lactate, glucose, and ketone body levels and measured cardiac mitochondrial respiration. In both male and female mice, moderate- and high-intensity exercise acutely increased blood lactate levels. In both sexes, low- and moderate-intensity exercise augmented circulating 3-hydroxybutryrate levels immediately after the exercise bout; however, only in female mice did high-intensity exercise increase 3-hydroxybutyrate levels, with significant increases occurring 1 h after the exercise session. Untargeted metabolomics analyses of sedentary female and male hearts suggest considerable sex-dependent differences in basal cardiac metabolite levels, with female hearts characterized by higher levels of pantothenate, pyridoxamine, homoarginine, tryptophan, and several glycerophospholipid and sphingomyelin species and lower levels of numerous metabolites, including acetyl coenzyme A, glucuronate, gulonate, hydroxyproline, prolyl-hydroxyproline, carnosine, anserine, and carnitinylated and glycinated species, as compared with male hearts. Immediately after a bout of treadmill exercise, both male and female hearts had higher levels of corticosterone; however, female mice showed more extensive exercise-induced changes in the cardiac metabolome, characterized by significant, time-dependent changes in amino acids (e.g., serine, alanine, tyrosine, tryptophan, branched-chain amino acids) and the ketone body 3-hydroxybutyrate. Results from experiments using isolated cardiac mitochondria suggest that high-intensity treadmill exercise does not acutely affect respiration or mitochondrial coupling; however, female cardiac mitochondria demonstrate generally higher adenosine diphosphate sensitivity compared with male cardiac mitochondria. Collectively, these findings in mice reveal key sex-dependent differences in cardiac metabolism and suggest that the metabolic network in the female heart is more responsive to physiological stress caused by exercise.
Collapse
Affiliation(s)
- Kyle Fulghum
- Diabetes and Obesity Center, Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA; Department of Physiology, University of Louisville, Louisville, KY 40202, USA
| | - Helen E Collins
- Diabetes and Obesity Center, Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| | - Steven P Jones
- Diabetes and Obesity Center, Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| | - Bradford G Hill
- Diabetes and Obesity Center, Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
27
|
Yuan X, Cai L, Hu F, Xie L, Chen X, Wu J, Li Q. Evaluation of the predictive values of elevated serum L-homoarginine and dimethylarginines in preeclampsia. Amino Acids 2022; 54:1215-1227. [PMID: 35752997 PMCID: PMC9365731 DOI: 10.1007/s00726-022-03177-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/28/2022] [Indexed: 01/14/2023]
Abstract
L-homoarginine (hARG) is involved in nitric oxide biosynthesis, but its role and concentration in preeclampsia (PE) have not been fully revealed. The purpose of this study was to develop and validate a feasible clinical assay to quantify serum hARG, arginine (ARG), asymmetric (ADMA) and symmetric dimethylarginines (SDMA) levels by LC-MS/MS and investigate their differences at different stages of pregnancy with or without preeclampsia. Serum samples were collected from 84 pregnant women without complications (controls), 84 with mild preeclampsia (MPE), and 81 with severe preeclampsia (SPE) at various gestation stages (before the 20th week, during the 20th-28th week or after the 28th week of gestation). No significant difference in ARG levels was observed between PE and controls at any stage (P > 0.05). The serum hARG levels and hARG/ADMA ratios of MPE before the 20th week were higher than those of controls (P < 0.001). ADMA levels of MPE were higher than those of controls during the 20th-28th week (P < 0.01). SDMA levels of SPE were higher than those of MPE (P < 0.01) and controls (P < 0.05) after the 28th week. Elevated serum hARG before the 20th week was identified as an independent predictor for PE (OR = 1.478, 95% CI 1.120-1.950). ROC curve analysis showed serum hARG before the 20th week had a good potential to predict MPE (AUC = 0.875, 95% CI 0.759-0.948). In conclusion, our study indicated that elevated serum hARG and dimethylarginine levels detected by LC-MS/MS might serve as potential biomarkers for the early prediction of PE.
Collapse
Affiliation(s)
- Xiangmei Yuan
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Leiming Cai
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Fengmei Hu
- Shanghai AB Sciex Analytical Instrument Trading Co., Ltd., Shanghai, 200050, China
| | - Li Xie
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Xiong Chen
- Department of Gynecology and Obstetrics, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Jingjing Wu
- Department of Gynecology and Obstetrics, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Qian Li
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China.
| |
Collapse
|
28
|
Saleh S, George J, Kott KA, Meikle PJ, Figtree GA. The Translation and Commercialisation of Biomarkers for Cardiovascular Disease—A Review. Front Cardiovasc Med 2022; 9:897106. [PMID: 35722087 PMCID: PMC9201254 DOI: 10.3389/fcvm.2022.897106] [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: 03/15/2022] [Accepted: 05/11/2022] [Indexed: 11/19/2022] Open
Abstract
As a leading cause of mortality and morbidity worldwide, cardiovascular disease and its diagnosis, quantification, and stratification remain significant health issues. Increasingly, patients present with cardiovascular disease in the absence of known risk factors, suggesting the presence of yet unrecognized pathological processes and disease predispositions. Fortunately, a host of emerging cardiovascular biomarkers characterizing and quantifying ischaemic heart disease have shown great promise in both laboratory settings and clinical trials. These have demonstrated improved predictive value additional to widely accepted biomarkers as well as providing insight into molecular phenotypes beneath the broad umbrella of cardiovascular disease that may allow for further personalized treatment regimens. However, the process of translation into clinical practice – particularly navigating the legal and commercial landscape – poses a number of challenges. Practical and legal barriers to the biomarker translational pipeline must be further considered to develop strategies to bring novel biomarkers into the clinical sphere and apply these advances at the patient bedside. Here we review the progress of emerging biomarkers in the cardiovascular space, with particular focus on those relevant to the unmet needs in ischaemic heart disease.
Collapse
Affiliation(s)
- Soloman Saleh
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Jacob George
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Katharine A. Kott
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Peter J. Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Gemma A. Figtree
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- *Correspondence: Gemma A. Figtree
| |
Collapse
|
29
|
Targeting Arginine in COVID-19-Induced Immunopathology and Vasculopathy. Metabolites 2022; 12:metabo12030240. [PMID: 35323682 PMCID: PMC8953281 DOI: 10.3390/metabo12030240] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) represents a major public health crisis that has caused the death of nearly six million people worldwide. Emerging data have identified a deficiency of circulating arginine in patients with COVID-19. Arginine is a semi-essential amino acid that serves as key regulator of immune and vascular cell function. Arginine is metabolized by nitric oxide (NO) synthase to NO which plays a pivotal role in host defense and vascular health, whereas the catabolism of arginine by arginase to ornithine contributes to immune suppression and vascular disease. Notably, arginase activity is upregulated in COVID-19 patients in a disease-dependent fashion, favoring the production of ornithine and its metabolites from arginine over the synthesis of NO. This rewiring of arginine metabolism in COVID-19 promotes immune and endothelial cell dysfunction, vascular smooth muscle cell proliferation and migration, inflammation, vasoconstriction, thrombosis, and arterial thickening, fibrosis, and stiffening, which can lead to vascular occlusion, muti-organ failure, and death. Strategies that restore the plasma concentration of arginine, inhibit arginase activity, and/or enhance the bioavailability and potency of NO represent promising therapeutic approaches that may preserve immune function and prevent the development of severe vascular disease in patients with COVID-19.
Collapse
|
30
|
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: 0.3] [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]
|
31
|
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
|
32
|
Koch V, Weber C, Riffel JH, Buchner K, Buss SJ, Hein S, Mereles D, Hagenmueller M, Erbel C, März W, Booz C, Albrecht MH, Vogl TJ, Frey N, Hardt SE, Ochs M. Impact of Homoarginine on Myocardial Function and Remodeling in a Rat Model of Chronic Renal Failure. J Cardiovasc Pharmacol Ther 2022; 27:10742484211054620. [PMID: 34994208 DOI: 10.1177/10742484211054620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE Low plasma concentrations of the amino acid homoarginine (HA) have been shown to correlate with adverse cardiovascular outcome, particularly in patients with chronic kidney disease. The present study sought to investigate the effect of HA treatment on cardiac remodeling in rats undergoing artificially induced renal insufficiency by 5/6 nephrectomy (5/6 Nx). METHODS A total of 33 male Wistar rats were randomly divided into sham and 5/6 Nx groups, receiving either placebo treatment or 400 mg·kg-1·day-1 HA over a 4-week period. RESULTS 5/6 Nx per se resulted in adverse myocardial remodeling with aggravated cardiac function and associated cardiac overload as the most obvious alteration (-23% ejection fraction, P < 0.0001), as well as increased myocardial fibrosis (+80%, P = 0.0005) compared to placebo treated sham animals. HA treatment of 5/6 Nx rats has led to an improvement of ejection fraction (+24%, P = 0.0003) and fractional shortening (+21%, P = 0.0126), as well as a decrease of collagen deposition (-32%, P = 0.0041), left ventricular weight (-14%, P = 0.0468), and myocyte cross-sectional area (-12%, P < 0.0001). These changes were accompanied by a downregulation of atrial natriuretic factor (-65% P < 0.0001) and collagen type V alpha 1 chain (-44%, P = 0.0006). Sham animals revealed no significant changes in cardiac function, myocardial fibrosis, or any of the aforementioned molecular changes after drug treatment. CONCLUSION Dietary HA supplementation appears to have the potential of preventing cardiac remodeling and improving heart function in the setting of chronic kidney disease. Our findings shed new light on HA as a possible new therapeutic agent for patients at high cardiovascular risk.
Collapse
Affiliation(s)
- Vitali Koch
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Christophe Weber
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Johannes H Riffel
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Kristina Buchner
- Institute of Human Genetics, Section for Developmental Genetics, 27178University of Heidelberg, Heidelberg, Germany
| | - Sebastian J Buss
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Selina Hein
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Derliz Mereles
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Marco Hagenmueller
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Erbel
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Winfried März
- Synlab Academy, Synlab Holding Deutschland GmbH, Augsburg, Germany
| | - Christian Booz
- 9173Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Thomas J Vogl
- 9173Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan E Hardt
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Marco Ochs
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
33
|
Eryavuz Onmaz D, Abusoglu S, Yaglioglu H, Abusoglu G, Unlu A. Developing a robust, fast and reliable measurement method for the analysis of methylarginine derivatives and related metabolites. J Mass Spectrom Adv Clin Lab 2021; 19:34-45. [PMID: 34820664 PMCID: PMC8601011 DOI: 10.1016/j.jmsacl.2021.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 11/21/2022] Open
Abstract
Background Nitric oxide (NO) plays an important role in endothelial homeostasis. Asymmetric dimethyl arginine (ADMA), L-N monomethyl arginine (L-NMMA) and symmetric dimethyl arginine (SDMA), which are derivatives of methylarginine, directly or indirectly reduce NO production. Therefore, these metabolites are an important risk factor for various diseases, including cardiovascular diseases. Numerous methods have been developed for the measurement of methylarginine derivatives, but various difficulties have been encountered. This study aimed to develop a reliable, fast and cost-effective method for the analysis and measurement of methylarginine derivatives (ADMA, SDMA, L-NMMA) and related metabolites (arginine, citrulline, homoarginine, ornithine), and to validate this method according to Clinical and Laboratory Standards Institute (CLSI) protocols. Methods For the analysis of ADMA, SDMA, L-NMMA, arginine, homoarginine, citrulline, ornithine, 200 µl of serum were precipitated with methanol, and subsequently derivatized with a butanol solution containing 5% acetyl chloride. Butyl derivatives were separated using a C18 reverse phase column with a 5 min run time. Detection of analytes was achieved by utilising the specific fragmentation patterns identified through tandem mass spectrometry. Results The method was linear for ADMA, SDMA, L-NMMA, ornithine, arginine, homoarginine and citrulline in the ranges of 0.023–6.0, 0.021–5.5, 0.019–5.0, 0.015–250, 0.015–250, 0.019–5 and 0.015–250 µM, respectively. The inter-assay CV% values for all analytes was less than 9.8%. Conclusions Data obtained from method validation studies shows that the developed method is highly sensitive, precise and accurate. Short analysis time, cost-effectiveness, and multiplexed analysis of these metabolites, with the same pretreatment steps, are the main advantages of the method.
Collapse
Key Words
- ADMA
- ADMA, asymmetric dimethyl arginine
- CE, capillary electrophoresis
- CE, collision energy
- CLSI, The Clinical & Laboratory Standards Institute
- CXP, collision cell exit potential
- DDAH, dimethylaminohydrolase
- DP, declustering potential
- EP, enterance potential
- FDA, Food and Drug Administration
- GC–MS, gas chromatography–mass spectrometry
- HPLC, high performance liquid chromatography
- L-NMMA, L-N monomethyl arginine
- LC-MS, liquid chromatography–mass spectrometry
- LC-MS/MS, liquid chromatography tandem-mass spectrometry
- MRM, multiple reaction monitoring
- Methylarginines
- NO, nitric oxide
- NOS, nitric oxide synthase
- PRMTs, protein arginine methyltransferases
- SDMA, symmetric dimethyl arginine
- Tandem mass spectrometry
- Validation
Collapse
Affiliation(s)
- Duygu Eryavuz Onmaz
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Sedat Abusoglu
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Havva Yaglioglu
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Gulsum Abusoglu
- Department of Medical Laboratory Techniques, Selcuk University Vocational School of Health, Konya, Turkey
| | - Ali Unlu
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| |
Collapse
|
34
|
Bajaj JS, Garcia-Tsao G, Reddy KR, O’Leary JG, Vargas HE, Lai JC, Kamath PS, Tandon P, Subramanian RM, Thuluvath P, Fagan A, Sehrawat T, de la Rosa Rodriguez R, Thacker LR, Wong F. Admission Urinary and Serum Metabolites Predict Renal Outcomes in Hospitalized Patients With Cirrhosis. Hepatology 2021; 74:2699-2713. [PMID: 34002868 PMCID: PMC9338693 DOI: 10.1002/hep.31907] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Acute kidney injury (AKI) has a poor prognosis in cirrhosis. Given the variability of creatinine, the prediction of AKI and dialysis by other markers is needed. The aim of this study is to determine the role of serum and urine metabolomics in the prediction of AKI and dialysis in an inpatient cirrhosis cohort. APPROACH AND RESULTS Inpatients with cirrhosis from 11 North American Consortium of End-stage Liver Disease centers who provided admission serum/urine when they were AKI and dialysis-free were included. Analysis of covariance adjusted for demographics, infection, and cirrhosis severity was performed to identify metabolites that differed among patients (1) who developed AKI or not; (2) required dialysis or not; and/pr (3) within AKI subgroups who needed dialysis or not. We performed random forest and AUC analyses to identify specific metabolite(s) associated with outcomes. Logistic regression with clinical variables with/without metabolites was performed. A total of 602 patients gave serum (218 developed AKI, 80 needed dialysis) and 435 gave urine (164 developed AKI, 61 needed dialysis). For AKI prediction, clinical factor-adjusted AUC was 0.91 for serum and 0.88 for urine. Major metabolites such as uremic toxins (2,3-dihydroxy-5-methylthio-4-pentenoic acid [DMTPA], N2N2dimethylguanosine, uridine/pseudouridine) and tryptophan/tyrosine metabolites (kynunerate, 8-methoxykyunerate, quinolinate) were higher in patients who developed AKI. For dialysis prediction, clinical factor-adjusted AUC was 0.93 for serum and 0.91 for urine. Similar metabolites as AKI were altered here. For dialysis prediction in those with AKI, the AUC was 0.81 and 0.79 for serum/urine. Lower branched-chain amino-acid (BCAA) metabolites but higher cysteine, tryptophan, glutamate, and DMTPA were seen in patients with AKI needing dialysis. Serum/urine metabolites were additive to clinical variables for all outcomes. CONCLUSIONS Specific admission urinary and serum metabolites were significantly additive to clinical variables to predict AKI development and dialysis initiation in inpatients with cirrhosis. These observations can potentially facilitate earlier initiation of renoprotective measures.
Collapse
Affiliation(s)
- Jasmohan S. Bajaj
- Virginia Commonwealth University and Central Virginia Veterans Healthcare System, Richmond, VA
| | | | | | | | | | | | | | | | | | | | - Andrew Fagan
- Virginia Commonwealth University and Central Virginia Veterans Healthcare System, Richmond, VA
| | | | | | - Leroy R. Thacker
- Virginia Commonwealth University and Central Virginia Veterans Healthcare System, Richmond, VA
| | | |
Collapse
|
35
|
Onmaz DE, Isik K, Sivrikaya A, Abusoglu S, Gezer İA, Abusoglu G, Yerlikaya FH, Unlu A. Determination of serum methylarginine levels by tandem mass spectrometric method in patients with ankylosing spondylitis. Amino Acids 2021; 53:1329-1338. [PMID: 34273021 DOI: 10.1007/s00726-021-03046-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/05/2021] [Indexed: 10/20/2022]
Abstract
Our aim in this study was to measure serum levels of methylarginines and related metabolites in patients with ankylosing spondylitis (AS), moreover, to investigate the relationship between these parameters and various clinical and laboratory parameters of patients with AS. The study included 60 patients with AS and 60 healthy volunteers. Serum asymmetric dimethylarginine (ADMA), L-N monomethylarginine (L-NMMA), symmetric dimethylarginine (SDMA), arginine (Arg), homoarginine (hArg), ornithine, and citrulline concentrations were measured with tandem mass spectrometry. In addition, participants were divided into three groups according to the treatment regimen: TNF-α inhibitor group (n = 25), conventional therapy group (n = 35), and control group (n = 60). These groups were compared in terms of serum levels of methylarginine pathway metabolites and various biochemical parameters. It was found that total methylated arginine load significantly increased in patients with AS (p < 0.001), and the Arg/ADMA ratio was positively correlated with HDL levels and negatively correlated with glucose, ESR, total cholesterol, triglyceride, and LDL levels. In addition, serum ADMA, SDMA, total methylated arginine load, and CRP levels were lower (p < 0.05) in the TNF-α group compared to the conventional treatment group. To the best of our knowledge, this is the first study to comprehensively investigate serum methylarginine levels in patients with AS. Elevated total methylated arginine load and decreased global arginine bioavailability ratio (GABR) indicate that NO metabolism is impaired in patients with AS. Therefore, the increased cardiovascular risk in patients with AS may be related to the decreased NO production or bioavailability due to the elevated total methylarginine load.
Collapse
Affiliation(s)
- Duygu Eryavuz Onmaz
- Department of Biochemistry, Selcuk University Faculty of Medicine Alaaddin Keykubat Campus, 42075, Selcuklu, Konya, Turkey.
| | - Kevser Isik
- Department of Physical Medicine and Rehabilitation, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Abdullah Sivrikaya
- Department of Biochemistry, Selcuk University Faculty of Medicine Alaaddin Keykubat Campus, 42075, Selcuklu, Konya, Turkey
| | - Sedat Abusoglu
- Department of Biochemistry, Selcuk University Faculty of Medicine Alaaddin Keykubat Campus, 42075, Selcuklu, Konya, Turkey
| | - İlknur Albayrak Gezer
- Department of Physical Medicine and Rehabilitation, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Gulsum Abusoglu
- Department of Medical Laboratory Techniques, Selcuk University Vocational School of Health, Konya, Turkey
| | - Fatma Humeyra Yerlikaya
- Department of Biochemistry, Selcuk University Faculty of Medicine Alaaddin Keykubat Campus, 42075, Selcuklu, Konya, Turkey
| | - Ali Unlu
- Department of Biochemistry, Selcuk University Faculty of Medicine Alaaddin Keykubat Campus, 42075, Selcuklu, Konya, Turkey
| |
Collapse
|
36
|
Homoarginine and methylarginines independently predict long-term outcome in patients presenting with suspicion of venous thromboembolism. Sci Rep 2021; 11:9569. [PMID: 33953241 PMCID: PMC8100302 DOI: 10.1038/s41598-021-88986-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/12/2021] [Indexed: 11/09/2022] Open
Abstract
Endogenous arginine derivatives homoarginine, asymmetric dimethylarginine (ADMA) and symmetric dimethyarginine (SDMA) are independent mortality predictors in atherosclerotic cardiovascular disease (CVD). Our study reports the first analysis, whether homoarginine, ADMA and SDMA predict venous thromboembolism (VTE) recurrence and overall mortality in patients with suspected acute VTE. We assessed serum levels of homoarginine, ADMA and SDMA by LC-MS/MS in 865 individuals from a prospective consecutive cohort of patients with clinical suspicion of VTE. The median follow-up time for mortality was 1196 days. VTE was confirmed by imaging in 418 patients and excluded in 447 patients. Low levels of homoarginine and high levels of ADMA or SDMA independently predicted all-cause mortality after adjustment for sex, age, oral anticoagulants, body mass index, arterial hypertension, diabetes mellitus, smoking, dyslipidemia, chronic heart failure, history of stroke, creatinine and cancer both in patients with VTE and without VTE. Interestingly, none of those parameters was predictive for VTE recurrence. We provide the first report that low circulating levels of homoarginine and high circulating levels of ADMA and SDMA independently predict all-cause mortality in patients with suspected VTE. These parameters might serve as markers of "frailty" and should be considered for future risk stratification approaches in this clinical population. Taking into account that homoarginine supplementation is protective in animal models of CVD and safe in healthy human volunteers, our study provides the basis for future homoarginine supplementation studies in patients with suspected VTE to investigate possible direct protective effects of homoarginine in this population.
Collapse
|
37
|
Mokhaneli MC, Botha-Le Roux S, Fourie CMT, Böger R, Schwedhelm E, Mels CMC. L-homoarginine is associated with decreased cardiovascular- and all-cause mortality. Eur J Clin Invest 2021; 51:e13472. [PMID: 33320332 DOI: 10.1111/eci.13472] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Increasing evidence suggests that L-homoarginine, an endogenous analogue of the amino acid L-arginine, may have beneficial effects on vascular homeostasis. We examined whether L-homoarginine is associated with 10-year risk of all-cause and cardiovascular mortality in a black South African population. METHODS We included 669 black South African participants (mean age 59.5 years), 143 of whom died during the 10-year follow-up period. Mortality data were acquired via verbal autopsy. Plasma L-homoarginine (and other related markers) were analysed with liquid chromatography-tandem mass spectrometry. RESULTS Survivors had higher L-homoarginine levels compared with nonsurvivors (1.25 µM vs. 0.89 µM; P < .001). Multivariable Cox regression analyses revealed that higher plasma L-homoarginine predicted a reduction in 10-year cardiovascular (hazard ratio [HR] per SD increment, 0.61; 95% CI 0.50 to 0.75) and all-cause (hazard ratio [HR] per SD increment, 0.59; 95% CI 0.41 to 0.84) mortality risk. CONCLUSION Higher L-homoarginine levels are associated with reduced risk of 10-year cardiovascular and all-cause mortality. Regulation of L-homoarginine levels as a therapeutic target in the management of cardiovascular disease should be investigated.
Collapse
Affiliation(s)
- Maserame Cleopatra Mokhaneli
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Shani Botha-Le Roux
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Carla Maria Theresia Fourie
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Rainer Böger
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Deutsches Zentrum fuer Herz-Kreislauf-Forschung E.V. (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Edzard Schwedhelm
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Deutsches Zentrum fuer Herz-Kreislauf-Forschung E.V. (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Catharina Martha Cornelia Mels
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| |
Collapse
|
38
|
Malle O, Trummer C, Theiler-Schwetz V, Meinitzer A, Keppel MH, Grübler MR, Tomaschitz A, Voelkl J, März W, Pilz S. NO Synthesis Markers are Not Significantly Associated with Blood Pressure and Endothelial Dysfunction in Patients with Arterial Hypertension: A Cross-Sectional Study. J Clin Med 2020; 9:E3895. [PMID: 33266290 PMCID: PMC7760204 DOI: 10.3390/jcm9123895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/17/2022] Open
Abstract
Nitric oxide (NO) synthesis markers, comprising L-homoarginine, asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA), are significantly associated with cardiovascular events and mortality. Being involved in NO pathways, they may be of high importance regulating vascular tone and arterial hypertension, but data on this topic are sparse and controversial. In this study, we evaluated whether these NO synthesis markers are associated with blood pressure values and pulse wave velocity (PWV). This analysis was based on the data of the Styrian Vitamin D Hypertension Trial, which included adults with arterial hypertension. We analyzed correlations of NO synthesis markers with 24 h ambulatory blood pressure values and PWV (primary outcomes), as well as with anthropometric and laboratory data. A total of 509 patients were included in the present analysis. The mean age was 61.2 ± 10.5 years, mean PWV was 8.6 ± 2.4 m/s, mean 24 h systolic blood pressure was 127.5 ± 13.8 mmHg and mean 24 h diastolic blood pressure was 76.4 ± 9.5 mmHg. In bivariate analyses, there was a significant positive correlation between homoarginine and 24 h diastolic blood pressure (r = 0.1; p = 0.02), which was revealed to be no longer significant after adjustment for age, gender and glomerular filtration rate (GFR) in multivariate regression analysis. No other significant correlations of any NO synthesis markers with blood pressure or PWV were observed. In line with previous studies, there were inverse associations between homoarginine and age and between ADMA or SDMA and GFR (p < 0.05 for all). This study did not reveal a significant association between homoarginine, ADMA or SDMA and blood pressure or PWV in hypertensive adults. These results suggested that the associations of these parameters with adverse outcome may not be mediated by hypertension and/or endothelial dysfunction.
Collapse
Affiliation(s)
- Oliver Malle
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria;
| | - Christian Trummer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria;
| | - Verena Theiler-Schwetz
- Department of Laboratory Medicine, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria;
| | - Andreas Meinitzer
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrinology Lab Platform, Medical University of Graz, 8036 Graz, Austria;
| | - Martin H. Keppel
- Health Center Trofaiach-Gössgrabenstrasse, 8793 Trofaiach, Austria;
| | - Martin R. Grübler
- Institute for Physiology and Pathophysiology, Johannes Kepler University, 4040 Linz, Austria;
| | - Andreas Tomaschitz
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany;
| | - Jakob Voelkl
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Winfried März
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany;
| | - Stefan Pilz
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria;
| |
Collapse
|
39
|
Zaric BL, Radovanovic JN, Gluvic Z, Stewart AJ, Essack M, Motwalli O, Gojobori T, Isenovic ER. Atherosclerosis Linked to Aberrant Amino Acid Metabolism and Immunosuppressive Amino Acid Catabolizing Enzymes. Front Immunol 2020; 11:551758. [PMID: 33117340 PMCID: PMC7549398 DOI: 10.3389/fimmu.2020.551758] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/25/2020] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular disease is the leading global health concern and responsible for more deaths worldwide than any other type of disorder. Atherosclerosis is a chronic inflammatory disease in the arterial wall, which underpins several types of cardiovascular disease. It has emerged that a strong relationship exists between alterations in amino acid (AA) metabolism and the development of atherosclerosis. Recent studies have reported positive correlations between levels of branched-chain amino acids (BCAAs) such as leucine, valine, and isoleucine in plasma and the occurrence of metabolic disturbances. Elevated serum levels of BCAAs indicate a high cardiometabolic risk. Thus, BCAAs may also impact atherosclerosis prevention and offer a novel therapeutic strategy for specific individuals at risk of coronary events. The metabolism of AAs, such as L-arginine, homoarginine, and L-tryptophan, is recognized as a critical regulator of vascular homeostasis. Dietary intake of homoarginine, taurine, and glycine can improve atherosclerosis by endothelium remodeling. Available data also suggest that the regulation of AA metabolism by indoleamine 2,3-dioxygenase (IDO) and arginases 1 and 2 are mediated through various immunological signals and that immunosuppressive AA metabolizing enzymes are promising therapeutic targets against atherosclerosis. Further clinical studies and basic studies that make use of animal models are required. Here we review recent data examining links between AA metabolism and the development of atherosclerosis.
Collapse
Affiliation(s)
- Bozidarka L. Zaric
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena N. Radovanovic
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Zoran Gluvic
- Department of Endocrinology and Diabetes, Faculty of Medicine, University Clinical-Hospital Centre Zemun-Belgrade, University of Belgrade, Belgrade, Serbia
| | - Alan J. Stewart
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - Magbubah Essack
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center, Computer (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Olaa Motwalli
- College of Computing and Informatics, Saudi Electronic University (SEU), Medina, Saudi Arabia
| | - Takashi Gojobori
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center, Computer (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Esma R. Isenovic
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
40
|
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: 1.4] [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
|
41
|
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: 1.2] [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
|
42
|
Grosse GM, Schwedhelm E, Worthmann H, Choe CU. Arginine Derivatives in Cerebrovascular Diseases: Mechanisms and Clinical Implications. Int J Mol Sci 2020; 21:ijms21051798. [PMID: 32150996 PMCID: PMC7084464 DOI: 10.3390/ijms21051798] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 02/07/2023] Open
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.
Collapse
Affiliation(s)
- Gerrit M. Grosse
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany;
- Correspondence:
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20249 Hamburg, Germany;
- DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Hamburg/Kiel/Lübeck, 20249 Hamburg, Germany
| | - Hans Worthmann
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany;
| | - Chi-un Choe
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20249 Hamburg, Germany;
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Because arginine is the substrate for nitric oxide synthesis, which is pivotal to vascular homeostasis and linked to the insulin response, it has long been posited that supplemental arginine could benefit cardiometabolic health. RECENT FINDINGS Recent data have supported the view that supplemental arginine could alleviate the initiation and development of endothelial dysfunction and also shown that it may reduce the risk of type 2 diabetes. One important finding is that these effects may indeed vary as a function of the amount of arginine, its form and notably the metabolic status of the population. Some studies have shown that low doses of slow-release arginine are better used for nitric oxide synthesis and beneficial in individuals with abnormal arginine metabolism/bioavailability. Pathophysiological data in rodents have emphasized the importance of arginase activation during the development of cardiometabolic risk, which lends credence to a potential benefit for arginine supplements. Likewise, epidemiological evidence suggests that alterations to arginine bioavailability are important regarding the cardiometabolic risk. However, other metabolic mechanisms linked to the multiple pathways of arginine metabolism may also play a role. SUMMARY Further studies are needed to confirm and analyze how and when supplemental arginine is beneficial to cardiometabolic health.
Collapse
Affiliation(s)
- François Mariotti
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay, 75005, Paris, France
| |
Collapse
|
44
|
Synthesis of homoagmatine and GC–MS analysis of tissue homoagmatine and agmatine: evidence that homoagmatine but not agmatine is a metabolite of pharmacological L-homoarginine in the anesthetized rat. Amino Acids 2019; 52:235-245. [DOI: 10.1007/s00726-019-02808-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023]
|
45
|
Choe CU, Lezius S, Cordts K, Gerloff C, Böger RH, Schwedhelm E, Grant PJ. Low homoarginine/SDMA ratio is associated with poor short- and long-term outcome after stroke in two prospective studies. Neurol Sci 2019; 41:149-153. [PMID: 31482247 DOI: 10.1007/s10072-019-04058-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 08/28/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Guanidino compounds, including asymmetric dimethylarginine (ADMA), symmetric dimethylarginine (SDMA), and L-homoarginine (hArg), have been associated with cardio- and cerebrovascular events and risk. We aimed to study if low hArg/ADMA and hArg/SDMA ratios are associated with mortality and outcome after stroke. METHODS In two prospective cohorts of acute stroke patients from Germany and the UK, we analyzed hArg, ADMA, and SDMA to determine hArg/ADMA and hArg/SDMA ratios. The guanidino compound levels were associated with mortality, adverse events, and neurological impairment, i.e., National Institutes of Health Stroke Scale (NIHSS) and modified Rankin Scale (mRS). RESULTS During 7.4 years, high hArg/ADMA and hArg/SDMA ratios were both associated with a reduction in all-cause mortality in patients with ischemic stroke in a UK stroke cohort (hArg/ADMA: hazard ratio (HR) 0.75 [95% confidence interval (CI) 0.62-0.92]; n = 394; P = 0.006; hArg/SDMA: HR 0.68 [0.54-0.85]; n = 394; P = 0.001). In a German stroke cohort, patients with high hArg/SDMA ratio experienced fewer adverse events compared with those with low hArg/SDMA ratios within 30 days after stroke (HR 0.73 [0.57-0.92]; n = 135; P = 0.009), whereas hArg/ADMA was not predictive. Furthermore, hArg/SDMA ratios inversely correlated with the degree of neurological impairment (NIHSS) (r = - 0.27; P = 0.001; n = 138). Lower hArg/SDMA ratios were also found in dependent (mRS 3-6) compared with independent patients (mRS < 3; P = 0.007; n = 138), whereas hArg/ADMA did not. CONCLUSION These results from two prospective stroke studies reveal that hArg/SDMA ratio could prove a valuable blood-based biomarker to discriminate patients with poor short- and long-term outcome, increased neurological impairment, and severe disability after stroke.
Collapse
Affiliation(s)
- Chi-Un Choe
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Susanne Lezius
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kathrin Cordts
- Institute of Clinical Pharmacology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Christian Gerloff
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Rainer H Böger
- Institute of Clinical Pharmacology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Peter J Grant
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, UK
| |
Collapse
|
46
|
Wetzel MD, Gao T, Venkatachalam M, Morris SM, Awad AS. l-Homoarginine supplementation prevents diabetic kidney damage. Physiol Rep 2019; 7:e14235. [PMID: 31552707 PMCID: PMC6759505 DOI: 10.14814/phy2.14235] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
l-homoarginine is an endogenous, non-proteinogenic amino acid that has emerged as a new player in health and disease. Specifically, low l-homoarginine levels are associated with cardiovascular diseases, stroke, and reduced kidney function. However, the role of l-homoarginine in the pathogenesis of diabetic nephropathy (DN) is not known. Experiments were conducted in 6-week-old Ins2Akita mice supplemented with l-homoarginine via drinking water or mini osmotic pump for 12 weeks. Both plasma and kidney l-homoarginine levels were significantly reduced in diabetic mice compared to nondiabetic controls. Untreated Ins2Akita mice showed significant increases in urinary albumin excretion, histological changes, glomerular macrophage recruitment, the inflammatory cytokine KC-GRO/CXCL1, and urinary thiobarbituric acid reactive substances (TBARS) excretion as an indicator of oxidative stress, along with a significant reduction in kidney nitrate + nitrite levels compared to control mice at 18 weeks of age. In contrast, l-homoarginine supplementation for 12 weeks in Ins2Akita mice, via either drinking water or mini osmotic pump, significantly reduced albuminuria, renal histological changes, glomerular macrophage recruitment, KC-GRO/CXCL1 levels, urinary TBARS excretion, and largely restored kidney nitrate + nitrite levels. These data demonstrate that l-homoarginine supplementation attenuates specific features of DN in mice and could be a potential new therapeutic tool for treating diabetic patients.
Collapse
Affiliation(s)
- Michael D. Wetzel
- Department of MedicineUniversity of Texas Health Science Center at San AntonioSan AntonioTexas
| | - Ting Gao
- Department of MedicinePenn State University College of MedicineHersheyPennsylvania
| | - Manjeri Venkatachalam
- Department of PathologyUniversity of Texas Health Science Center at San AntonioSan AntonioTexas
| | - Sidney M. Morris
- Department of Microbiology & Molecular GeneticsUniversity of PittsburghPittsburghPennsylvania
| | - Alaa S. Awad
- Department of MedicineUniversity of Texas Health Science Center at San AntonioSan AntonioTexas
- Department of MedicinePenn State University College of MedicineHersheyPennsylvania
| |
Collapse
|