1
|
Hannemann J, Wasser K, Mileva Y, Kleinsang F, Schubert M, Schwedhelm E, Guan K, Wachter R, Böger R. Symmetric Dimethylarginine Predicts Previously Undetected Atrial Fibrillation in Patients With Ischemic Stroke. J Am Heart Assoc 2024; 13:e034994. [PMID: 39190577 DOI: 10.1161/jaha.124.034994] [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: 02/20/2024] [Accepted: 05/01/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Atrial fibrillation (AF) is a stroke risk factor that often remains undetected at hospital admission. Long-term Holter monitoring helps to identify patients with previously unrecognized AF. Asymmetric (ADMA) and symmetric dimethylarginine (SDMA) are elevated in AF in cross-sectional studies. We analyzed ADMA, SDMA, and other L-arginine metabolites to assess their association with AF in the Find-AF trial. METHODS AND RESULTS We included 280 patients presenting with acute cerebral ischemia. Patients presenting in sinus rhythm received 7-day Holter-ECG. Biomarkers were quantified by ultra-performance liquid chromatography-tandem mass spectrometry. We also analyzed protein methylation and L-arginine-related metabolites in human induced pluripotent stem cell-derived cardiomyocytes in vitro. ADMA and SDMA were elevated in 44 patients who presented with AF. SDMA, but not ADMA, was significantly elevated in patients newly diagnosed with AF in Holter-ECG as compared with those in sinus rhythm. SDMA plasma concentration >0.571 μmol/L significantly predicted presence of AF in Holter-ECG (area under the curve=0.676 [0.530-0.822]; P=0.029; sensitivity 0.786, specificity 0.572). SDMA levels further increased in patients with AF during the first 24 hours in hospital, and ADMA levels remained stable. In vitro, induced pluripotent stem cell-derived cardiomyocytes showed increased symmetric protein methylation and elevated SDMA during rapid pacing (2.0 Hz versus 0.5 Hz), whereas asymmetric protein methylation and ADMA were unchanged. CONCLUSIONS SDMA at admission was significantly elevated in stroke patients presenting with AF and showed a moderate but significant prospective association with previously unrecognized AF. Thus, stroke patients with elevated SDMA concentration at admission may specifically benefit from extended Holter-ECG monitoring.
Collapse
Affiliation(s)
- Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf Hamburg Germany
| | - Katrin Wasser
- Clinic for Neurology University of Göttingen Germany
| | - Yoana Mileva
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf Hamburg Germany
| | - Fiona Kleinsang
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf Hamburg Germany
| | - Mario Schubert
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus Technical University of Dresden Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf Hamburg Germany
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus Technical University of Dresden Germany
| | - Rolf Wachter
- Department of Cardiology University Hospital Leipzig Leipzig Germany
- Clinic for Cardiology and Pneumology University Medicine Göttingen Göttingen Germany
- German Cardiovascular Research Center (DZHK), partner site Göttingen Germany
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf Hamburg Germany
- German Cardiovascular Research Center (DZHK), partner site Hamburg-Lübeck-Kiel Hamburg Germany
| |
Collapse
|
2
|
Hannemann J, Zink A, Mileva Y, Balfanz P, Dahl E, Volland S, Illig T, Schwedhelm E, Kurth F, Stege A, Aepfelbacher M, Hoffmann A, Böger R. A multicenter study of asymmetric and symmetric dimethylarginine as predictors of mortality risk in hospitalized COVID-19 patients. Sci Rep 2024; 14:15739. [PMID: 38977837 PMCID: PMC11231343 DOI: 10.1038/s41598-024-66288-3] [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: 02/28/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
Mortality of patients hospitalized with COVID-19 has remained high during the consecutive SARS-CoV-2 pandemic waves. Early discrimination of patients at high mortality risk is crucial for optimal patient care. Symmetric (SDMA) and asymmetric dimethylarginine (ADMA) have been proposed as possible biomarkers to improve risk prediction of COVID-19 patients. We measured SDMA, ADMA, and other L-arginine-related metabolites in 180 patients admitted with COVID-19 in four German university hospitals as compared to 127 healthy controls. Patients were treated according to accepted clinical guidelines and followed-up until death or hospital discharge. Classical inflammatory markers (leukocytes, CRP, PCT), renal function (eGFR), and clinical scores (SOFA) were taken from hospital records. In a small subgroup of 23 COVID-19 patients, sequential blood samples were available and analyzed for biomarker trends over time until 14 days after admission. Patients had significantly elevated SDMA, ADMA, and L-ornithine and lower L-citrulline concentrations than controls. Within COVID-19 patients, SDMA and ADMA were significantly higher in non-survivors (n = 41, 22.8%) than in survivors. In ROC analysis, the optimal cut-off to discriminate non-survivors from survivors was 0.579 µmol/L for SDMA and 0.599 µmol/L for ADMA (both p < 0.001). High SDMA and ADMA were associated with odds ratios for death of 11.45 (3.37-38.87) and 5.95 (2.63-13.45), respectively. Analysis of SDMA and ADMA allowed discrimination of a high-risk (mortality, 43.7%), medium-risk (15.1%), and low-risk group (3.6%); risk prediction was significantly improved over classical laboratory markers. We conclude that analysis of ADMA and SDMA after hospital admission significantly improves risk prediction in COVID-19.
Collapse
Affiliation(s)
- Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Zink
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yoana Mileva
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Balfanz
- Department of Cardiology, Angiology and Intensive Care Medicine, Medical Clinic I, University Hospital Aachen, Aachen, Germany
- Institute of Pathology and Central Biobank, University Hospital Aachen, Aachen, Germany
| | - Edgar Dahl
- Hannover Unified Biobank, Medizinische Hochschule Hannover, Hannover, Germany
| | - Sonja Volland
- Department of Physiology, Henri Mondor Hospital, FHU-SENEC, INSERM U955, Université Paris-Est Créteil (UPEC), AP-HP, Créteil, France
| | - Thomas Illig
- Department of Physiology, Henri Mondor Hospital, FHU-SENEC, INSERM U955, Université Paris-Est Créteil (UPEC), AP-HP, Créteil, France
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Florian Kurth
- Department of Infectious Diseases and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Alexandra Stege
- Central Biobank Charité, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Aepfelbacher
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Armin Hoffmann
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
3
|
Sudová V, Prokop P, Trefil L, Racek J, Rajdl D. Comparison of two methods for dimethylarginines quantification. Pract Lab Med 2024; 39:e00359. [PMID: 38313812 PMCID: PMC10831080 DOI: 10.1016/j.plabm.2024.e00359] [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: 01/30/2023] [Revised: 07/10/2023] [Accepted: 01/15/2024] [Indexed: 02/06/2024] Open
Abstract
Objectives Both dimethylarginines are widely bound to chronic kidney disease (CKD). This study was focused to validate published LC-MS/MS method and compared the measured data with an immunoassay. Design and methods The analysis was performed on a Dionex UltiMate 3000 UHPLC-Standard (Thermo Fisher Scientific, Waltham, Massachusetts, USA) with an amaZon SL ion trap (Bruker, Billerica, Massachusetts, USA). Comparison was evaluated by using Passing Bablok regression and Bland Altman plot. Healthy volunteers (n = 40) were used for validation and as control group to patients group (n = 40) with different stages of CKD. Results The results in healthy controls determined by the LC-MS/MS (ELISA) method were 0.52 ± 0.0892 with 95 % CI: 0.49-0.55 (0.61 ± 0.1213 with 95 % CI: 0.57-0.64) μmol/L for AD MA and 0.56 ± 0.0810 with 95 % CI: 0.53-0.58 (0.62 ± 0.0752 with 95 % CI: 0.57-0.65) μmol/L for SDMA. In the same way, the patient group values determined by the LC-MS/MS (ELISA) method were 0.82 ± 0.1604 with 95 % CI: 0.75-0.88 (1.06 ± 0.3002 with 95 % CI: 0.94-1.19) μmol/L and 2.14 ± 0.8778 with 95 % CI: 1.47-2.58 (1.65 ± 0.5160 with 95 % CI: 1.40-1.98) μmol/L for ADMA and SDMA, respectively. The correlation between the methods, expressed as the Spearman correlation coefficient (R), was 0.858 (0.8059) for ADMA (p < 0.0001) and 0.895 (0.9607) for SDMA (p < 0.0001). Conclusions ADMA levels determined by the immunoassay were almost 30 % overestimated, in contrast to SDMA levels, which were 3 % underestimated. According to our findings, a better correlation could be obtained by simple sample dilution.
Collapse
Affiliation(s)
- Vendula Sudová
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, Pilsen, 32300, Czech Republic
- Department of Clinical Biochemistry and Haematology, Charles University and University Hospital, Pilsen, Czech Republic
| | - Pavel Prokop
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, Pilsen, 32300, Czech Republic
- Department of Clinical Biochemistry and Haematology, Charles University and University Hospital, Pilsen, Czech Republic
| | - Ladislav Trefil
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, Pilsen, 32300, Czech Republic
- Department of Clinical Biochemistry and Haematology, Charles University and University Hospital, Pilsen, Czech Republic
| | - Jaroslav Racek
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, Pilsen, 32300, Czech Republic
- Department of Clinical Biochemistry and Haematology, Charles University and University Hospital, Pilsen, Czech Republic
| | - Daniel Rajdl
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, Pilsen, 32300, Czech Republic
- Department of Clinical Biochemistry and Haematology, Charles University and University Hospital, Pilsen, Czech Republic
| |
Collapse
|
4
|
Winkler MS, Bahls M, Böger RH, Ittermann T, Dörr M, Friedrich N, Schwedhelm E. Association of Asymmetric and Symmetric Dimethylarginine with Inflammation in the Population-Based Study of Health in Pomerania. Biomolecules 2023; 13:1612. [PMID: 38002294 PMCID: PMC10669713 DOI: 10.3390/biom13111612] [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: 08/21/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
The amino acids arginine (Arg), asymmetric (ADMA) and symmetric dimethylarginine (SDMA) are related to nitric oxide (NO) metabolism and potential markers of two different disease entities: cardiovascular disease such as atherosclerosis and systemic inflammation in critically ill patients with sepsis. Although very different in their pathophysiological genesis, both entities involve the functional integrity of blood vessels. In this context, large population-based data associating NO metabolites with proinflammatory markers, e.g., white blood cell count (WBC), high-sensitivity C-reactive protein (hsCRP), and fibrinogen, or cytokines are sparse. We investigated the association of Arg, ADMA and SDMA with WBC, hsCRP, and fibrinogen in 3556 participants of the Study of Health in Pomerania (SHIP)-TREND study. Furthermore, in a subcohort of 456 subjects, 31 inflammatory markers and cytokines were analyzed. We identified Arg and SDMA to be positively associated with hsCRP (β coefficient 0.010, standard error (SE) 0.002 and 0.298, 0.137, respectively) as well as fibrinogen (β 5.23 × 10-3, SE 4.75 × 10-4 and 0.083, 0.031, respectively). ADMA was not associated with WBC, hsCRP, or fibrinogen. Furthermore, in the subcohort, Arg was inversely related to a proliferation-inducing ligand (APRIL). SDMA was positively associated with osteocalcin, tumor necrosis factor receptor 1 and 2, and soluble cluster of differentiation 30. Our findings provide new insights into the involvement of Arg, ADMA, and SDMA in subclinical inflammation in the general population.
Collapse
Affiliation(s)
- Martin Sebastian Winkler
- Department of Anesthesiology, Emergency and Intensive Care Medicine, University of Göttingen, 37075 Göttingen, Germany
| | - Martin Bahls
- Department of Internal Medicine B, University Medicine Greifswald, 17475 Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, 17475 Greifswald, Germany
| | - Rainer H. Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany (E.S.)
| | - Till Ittermann
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, 17475 Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Marcus Dörr
- Department of Internal Medicine B, University Medicine Greifswald, 17475 Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, 17475 Greifswald, Germany
| | - Nele Friedrich
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, 17475 Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany (E.S.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| |
Collapse
|
5
|
Hannemann J, Skene DJ, Middleton B, Schwedhelm E, Laing A, Böger R. Diurnal Variation of L-Arginine and the Cardiovascular Risk Markers Asymmetric and Symmetric Dimethylarginine and Homoarginine in Rotating Night Shift Workers and Controls. Biomolecules 2023; 13:1282. [PMID: 37759682 PMCID: PMC10526524 DOI: 10.3390/biom13091282] [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: 07/11/2023] [Revised: 08/17/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
Asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA) interfere with nitric oxide (NO) formation from L-arginine via different mechanisms. ADMA is a biomarker of cardiovascular disease and mortality, whilst SDMA is a biomarker of mortality after ischemic stroke. Homoarginine, another L-arginine-derived amino acid, is associated with stroke and congestive heart failure. Acute ischemic events like myocardial infarction show a time-of-day variation in the timing of their onset, as do NO-mediated vascular function and blood pressure. We studied whether the plasma concentrations of L-arginine-related amino acid metabolites show diurnal variation in a clinical study comparing 12 non-night shift workers with 60 rotating night shift workers. The plasma concentrations of L-arginine-related biomarkers, melatonin, and cortisol were measured every 3 h during a 24-h period. In addition, 24-h blood pressure recordings were performed. In non-night shift workers, L-arginine and homoarginine plasma concentrations showed diurnal variation with a 12-h period, which were both attenuated in night shift workers. ADMA and SDMA showed a 24-h rhythmicity with no significant differences in phase between night shift and non-night shift workers. The plasma profiles of melatonin and cortisol were not significantly different between both groups, suggesting that the rotating night shift work does not have a major influence on central suprachiasmatic nuclei clock timing. In addition, systolic and diastolic blood pressure patterns were similar between both groups. Our data show diurnal variation of dimethylarginines with the timing of their acrophases corresponding to the published timing of the peak incidence of cardiac ischemic events.
Collapse
Affiliation(s)
- Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.H.); (E.S.); (A.L.)
| | - Debra J. Skene
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK; (D.J.S.); (B.M.)
| | - Benita Middleton
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK; (D.J.S.); (B.M.)
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.H.); (E.S.); (A.L.)
| | - Anika Laing
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.H.); (E.S.); (A.L.)
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.H.); (E.S.); (A.L.)
| |
Collapse
|
6
|
Hannemann J, Thorarinnsdottir EH, Amaral AFS, Schwedhelm E, Schmidt-Hutten L, Stang H, Benediktsdottir B, Gunnarsdóttir I, Gislason T, Böger R. Biomarkers of the L-Arginine/Dimethylarginine/Nitric Oxide Pathway in People with Chronic Airflow Obstruction and Obstructive Sleep Apnoea. J Clin Med 2023; 12:5230. [PMID: 37629272 PMCID: PMC10455103 DOI: 10.3390/jcm12165230] [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: 07/05/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) and obstructive sleep apnoea (OSA) are common chronic diseases that are associated with chronic and intermittent hypoxemia, respectively. Patients affected by the overlap of COPD and OSA have a particularly unfavourable prognosis. The L-arginine/nitric oxide (NO) pathway plays an important role in regulating pulmonary vascular function. Asymmetric (ADMA) and symmetric dimethylarginine (SDMA) interfere with NO production. METHODS We analysed the serum concentrations of ADMA, SDMA, L-arginine, L-citrulline, and L-ornithine in a large sample of the Icelandic general population together with chronic airflow obstruction (CAO), a key physiological marker of COPD that was assessed by post-bronchodilator spirometry (FEV1/FVC < LLN). OSA risk was determined by the multivariable apnoea prediction (MAP) index. RESULTS 713 individuals were analysed, of whom 78 (10.9%) showed CAO and 215 (30%) had MAP > 0.5. SDMA was significantly higher in individuals with CAO (0.518 [0.461-0.616] vs. 0.494 [0.441-0.565] µmol/L; p = 0.005), but ADMA was not. However, ADMA was significantly associated with decreasing FEV1 percent predicted among those with CAO (p = 0.002). ADMA was 0.50 (0.44-0.56) µmol/L in MAP ≤ 0.5 versus 0.52 (0.46-0.58) µmol/L in MAP > 0.5 (p = 0.008). SDMA was 0.49 (0.44-0.56) µmol/L versus 0.51 (0.46-0.60) µmol/L, respectively (p = 0.004). The highest values for ADMA and SDMA were observed in individuals with overlap of CAO and MAP > 0.5, which was accompanied by lower L-citrulline levels. CONCLUSIONS The plasma concentrations of ADMA and SDMA are elevated in COPD patients with concomitant intermittent hypoxaemia. This may account for impaired pulmonary NO production, enhanced pulmonary vasoconstriction, and disease progression.
Collapse
Affiliation(s)
- Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.H.); (E.S.); (L.S.-H.); (H.S.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20246 Hamburg, Germany
| | - Elin H. Thorarinnsdottir
- Primary Health Care of the Capital Area, 103 Reykjavik, Iceland;
- Faculty of Medicine, University of Iceland, 102 Reykjavik, Iceland; (B.B.); (T.G.)
| | - André F. S. Amaral
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK;
- NIHR Imperial Biomedical Research Centre, London W2 1NY, UK
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.H.); (E.S.); (L.S.-H.); (H.S.)
| | - Lena Schmidt-Hutten
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.H.); (E.S.); (L.S.-H.); (H.S.)
| | - Heike Stang
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.H.); (E.S.); (L.S.-H.); (H.S.)
| | - Bryndis Benediktsdottir
- Faculty of Medicine, University of Iceland, 102 Reykjavik, Iceland; (B.B.); (T.G.)
- Sleep Department, Landspitali University Hospital of Iceland, 105 Reykjavik, Iceland
| | - Ingibjörg Gunnarsdóttir
- Unit for Nutrition Research, Landspitali University Hospital & Faculty of Food Science and Nutrition, University of Iceland, 102 Reykjavik, Iceland;
| | - Thórarinn Gislason
- Faculty of Medicine, University of Iceland, 102 Reykjavik, Iceland; (B.B.); (T.G.)
- Sleep Department, Landspitali University Hospital of Iceland, 105 Reykjavik, Iceland
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.H.); (E.S.); (L.S.-H.); (H.S.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20246 Hamburg, Germany
| |
Collapse
|
7
|
Xie Y, Zhao F, Freitag N, Borowski S, Wang Y, Harms C, Pang PC, Desforges J, Wen T, Schwedhelm E, Singh M, Dechend R, Dell A, Haslam SM, Dveksler G, Garcia MG, Blois SM. Maternal-derived galectin-1 shapes the placenta niche through Sda terminal glycosylation: Implication for preeclampsia. PNAS NEXUS 2023; 2:pgad247. [PMID: 37575671 PMCID: PMC10416815 DOI: 10.1093/pnasnexus/pgad247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023]
Abstract
Placental abnormalities cause impaired fetal growth and poor pregnancy outcome (e.g. preeclampsia [PE]) with long-lasting consequences for the mother and offspring. The molecular dialogue between the maternal niche and the developing placenta is critical for the function of this organ. Galectin-1 (gal-1), a highly expressed glycan-binding protein at the maternal-fetal interface, orchestrates the maternal adaptation to pregnancy and placenta development. Down-regulation or deficiency of gal-1 during pregnancy is associated with the development of PE; however, the maternal- and placental-derived gal-1 contributions to the disease onset are largely unknown. We demonstrate that lack of gal-1 imposes a risk for PE development in a niche-specific manner, and this is accompanied by a placental dysfunction highly influenced by the absence of maternal-derived gal-1. Notably, differential placental glycosylation through the Sda-capped N-glycans dominates the invasive trophoblast capacity triggered by maternal-derived gal-1. Our findings show that gal-1 derived from the maternal niche is essential for healthy placenta development and indicate that impairment of the gal-1 signaling pathway within the maternal niche could be a molecular cause for maternal cardiovascular maladaptation during pregnancy.
Collapse
Affiliation(s)
- Yiran Xie
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Fangqi Zhao
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Nancy Freitag
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max-Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin, 13125 Berlin, Germany
| | - Sophia Borowski
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max-Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin, 13125 Berlin, Germany
| | - Yiru Wang
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Charlotte Harms
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Poh-Choo Pang
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Juliette Desforges
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Tianyu Wen
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf and German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20249 Hamburg, Germany
| | - Manvendra Singh
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max-Delbrück Center for Molecular Medicine (MDC) and Charité-Universitätsmedizin, 13125 Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS-Klinikum, 13125 Berlin, Germany
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Gabriela Dveksler
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Mariana G Garcia
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
8
|
Sozio E, Hannemann J, Fabris M, Cifù A, Ripoli A, Sbrana F, Cescutti D, Vetrugno L, Fapranzi S, Bassi F, Sponza M, Curcio F, Tascini C, Böger R. The role of asymmetric dimethylarginine (ADMA) in COVID-19: association with respiratory failure and predictive role for outcome. Sci Rep 2023; 13:9811. [PMID: 37330534 PMCID: PMC10276836 DOI: 10.1038/s41598-023-36954-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: 10/12/2022] [Accepted: 06/13/2023] [Indexed: 06/19/2023] Open
Abstract
We aimed to assess the potential role of Asymmetric dimethylarginine (ADMA) in conditioning respiratory function and pulmonary vasoregulation during Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) infection. Within 72 h from admission, samples from 90 COVID-19 patients were assessed for ADMA, SDMA, L-arginine concentrations. In addition to classical statistics, patients were also clustered by a machine learning approach according to similar features. Multivariable analysis showed that C-reactive protein (OR 1.012), serum ADMA (OR 4.652), white blood cells (OR = 1.118) and SOFA (OR = 1.495) were significantly associated with negative outcomes. Machine learning-based clustering showed three distinct clusters: (1) patients with low severity not requiring invasive mechanical ventilation (IMV), (2) patients with moderate severity and respiratory failure whilst not requiring IMV, and (3) patients with highest severity requiring IMV. Serum ADMA concentration was significantly associated with disease severity and need for IMV although less pulmonary vasodilation was observed by CT scan. High serum levels of ADMA are indicative of high disease severity and requirement of mechanical ventilation. Serum ADMA at the time of hospital admission may therefore help to identify COVID-19 patients at high risk of deterioration and negative outcome.
Collapse
Affiliation(s)
- Emanuela Sozio
- Infectious Diseases Clinic, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy
| | - Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, Hamburg, Germany
| | - Martina Fabris
- Istituto di Patologia Clinica, Azienda Sanitaria Universitaria Friuli Centrale - Udine (ASUFC), Udine, Italy
| | - Adriana Cifù
- Department of Medical Area (DAME), University of Udine, Udine, Italy
| | - Andrea Ripoli
- Bioengineering Department, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Francesco Sbrana
- Lipoapheresis Unit - Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana "Gabriele Monasterio", Via Moruzzi 1, 56124, Pisa, Italy
| | - Demetrio Cescutti
- Infectious Diseases Clinic, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy
| | - Luigi Vetrugno
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Stefano Fapranzi
- Emergency Radiology Department - Azienda Sanitaria, Universitaria del Friuli Centrale (ASUFC), Udine, Italia
| | - Flavio Bassi
- Department of Anesthesia and Intensive Care Medicine, Azienda Sanitaria Universitaria Friuli Centrale - Udine (ASUFC), Udine, Italy
| | - Massimo Sponza
- Emergency Radiology Department - Azienda Sanitaria, Universitaria del Friuli Centrale (ASUFC), Udine, Italia
| | - Francesco Curcio
- Istituto di Patologia Clinica, Azienda Sanitaria Universitaria Friuli Centrale - Udine (ASUFC), Udine, Italy
- Department of Medical Area (DAME), University of Udine, Udine, Italy
| | - Carlo Tascini
- Infectious Diseases Clinic, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy.
- Department of Medical Area (DAME), University of Udine, Udine, Italy.
- U.O. Malattie Infettive, Azienda Sanitaria Universitaria Integrata di Udine, Via Pozzuolo, 330, 33100, Udine, Italy.
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, Hamburg, Germany
| |
Collapse
|
9
|
Tian Q, Mitchell BA, Moaddel R, Zoccali C, Bandinelli S, Ferrucci L. Metabolomic markers mediate erythrocyte anisocytosis in older adults: Results from three independent aging cohorts. J Intern Med 2023; 293:589-599. [PMID: 36739565 PMCID: PMC11353686 DOI: 10.1111/joim.13612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Anisocytosis reflects unequal-sized red blood cells and is quantified using red blood cell distribution width (RDW). RDW increases with age and has been consistently associated with adverse health outcomes, such as cardiovascular disease and mortality. Why RDW increases with age is not understood. We aimed to identify plasma metabolomic markers mediating anisocytosis with aging. METHODS We performed mediation analyses of plasma metabolomics on the association between age and RDW using resampling techniques after covariate adjustment. We analyzed data from adults aged 70 or older from the main discovery cohort of the Baltimore Longitudinal Study of Aging (BLSA, n = 477, 46% women) and validation cohorts of the Health, Aging and Body Composition Study (Health ABC, n = 620, 52% women) and Invecchiare in Chianti, Aging in the Chianti Area (InCHIANTI) study (n = 735, 57% women). Plasma metabolomics was assayed using the Biocrates MxP Quant 500 kit in BLSA and Health ABC and liquid chromatography with tandem mass spectrometry in InCHIANTI. RESULTS In all three cohorts, symmetric dimethylarginine (SDMA) significantly mediated the association between age and RDW. Asymmetric dimethylarginine (ADMA) and 1-methylhistidine were also significant mediators in the discovery cohort and one validation cohort. In the discovery cohort, we also found choline, homoarginine, and several long-chain triglycerides significantly mediated the association between age and RDW. CONCLUSIONS AND RELEVANCE This metabolomics study of three independent aging cohorts identified a specific set of metabolites mediating anisocytosis with aging. Whether SDMA, ADMA, and 1-methylhistidine are released by the damaged erythrocytes with high RDW or they affect the physiology of erythrocytes causing high RDW should be further investigated.
Collapse
Affiliation(s)
- Qu Tian
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, USA
| | - Brendan A. Mitchell
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, USA
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, Maryland, USA
| | - Carmine Zoccali
- Institute of Biology and Molecular Genetics (BIOGEM), Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia e Trapianto Renale (IPNET), Reggio Calabria, Italy
| | | | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Parthasarathy S, Soundararajan P, Sakthivelu M, Karuppiah KM, Velusamy P, Gopinath SC, Pachaiappan R. The role of prognostic biomarkers and their implications in early detection of preeclampsia: A systematic review. Process Biochem 2023. [DOI: 10.1016/j.procbio.2023.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
11
|
Piatek K, Feuerstein A, Zach V, Rozados da Conceicao C, Beblo A, Belyavskiy E, Pieske‐Kraigher E, Krannich A, Schwedhelm E, Hinz S, Pieske B, Edelmann F. Nitric oxide metabolites: associations with cardiovascular biomarkers and clinical parameters in patients with HFpEF. ESC Heart Fail 2022; 9:3961-3972. [PMID: 35979962 PMCID: PMC9773705 DOI: 10.1002/ehf2.14116] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/06/2022] [Accepted: 08/04/2022] [Indexed: 01/19/2023] Open
Abstract
AIMS Heart failure with preserved ejection fraction (HFpEF) is one of the most rapidly growing cardiovascular health burden worldwide, but there is still a lack of understanding about the HFpEF pathophysiology. The nitric oxide (NO) signalling pathway has been identified as a potential key element. The aim of our study was to investigate markers of NO metabolism [l-arginine (l-Arg), homoarginine (hArg), and asymmetric and symmetric dimethylarginine (ADMA and SDMA)], additional biomarkers [N-terminal pro-B-type natriuretic peptide (NT-proBNP), endothelin-1 (ET-1), mid-regional pro-adrenomedullin (MR-proADM), copeptin, and high-sensitivity C-reactive protein (hsCRP)], and the endothelial function in an integrated approach focusing on associations with clinical characteristics in patients with HFpEF. METHODS AND RESULTS Seventy-three patients, prospectively enrolled in the 'German HFpEF Registry', were analysed. Inclusion criteria were left ventricular ejection fraction (LVEF) ≥ 50%; New York Heart Association functional class ≥ II; elevated levels of NT-proBNP > 125 pg/mL; and at least one additional criterion for structural heart disease or diastolic dysfunction. All patients underwent transthoracic echocardiography, cardiopulmonary exercise testing, and pulse amplitude tonometry (EndoPAT™). Patients were categorized in two groups based on their retrospectively calculated HFA-PEFF score. Serum concentrations of l-Arg, hArg, ADMA, SDMA, NT-proBNP, ET-1, MR-proADM, copeptin, and hsCRP were determined. Patients had a median age of 74 years, 47% were female, and median LVEF was 57%. Fifty-two patients (71%) had an HFA-PEFF score ≥ 5 (definitive HFpEF), and 21 patients (29%) a score of 3 to 4 (risk for HFpEF). Overall biomarker concentrations were 126 ± 32 μmol/L for l-Arg, 1.67 ± 0.55 μmol/L for hArg, 0.74 (0.60;0.85) μmol/L for SDMA, and 0.61 ± 0.10 μmol/L for ADMA. The median reactive hyperaemia index (RHI) was 1.55 (1.38;1.87). SDMA correlated with NT-proBNP (r = 0.291; P = 0.013), ET-1 (r = 0.233; P = 0.047), and copeptin (r = 0.381; P = 0.001). ADMA correlated with ET-1 (r = 0.250; P = 0.033) and hsCRP (r = 0.303; P = 0.009). SDMA was associated with the left atrial volume index (β = 0.332; P = 0.004), also after adjustment for age, sex, and comorbidities. Biomarkers were non-associated with the RHI. A principal component analysis revealed two contrary clusters of biomarkers. CONCLUSIONS Our findings suggest an impaired NO metabolism as one possible key pathogenic determinant in at least a subgroup of patients with HFpEF. We argue for further evaluation of NO-based therapies. Upcoming studies should clarify whether subgroups of HFpEF patients can take more benefit from therapies that are targeting NO metabolism and pathway.
Collapse
Affiliation(s)
- Karsten Piatek
- Department of Internal Medicine and CardiologyCharité – Universitätsmedizin Berlin, Campus Virchow KlinikumAugustenburger Platz 1Berlin13353Germany
| | - Anna Feuerstein
- Department of Internal Medicine and CardiologyCharité – Universitätsmedizin Berlin, Campus Virchow KlinikumAugustenburger Platz 1Berlin13353Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
| | - Veronika Zach
- Department of Internal Medicine and CardiologyCharité – Universitätsmedizin Berlin, Campus Virchow KlinikumAugustenburger Platz 1Berlin13353Germany
| | - Cristina Rozados da Conceicao
- Department of Internal Medicine and CardiologyCharité – Universitätsmedizin Berlin, Campus Virchow KlinikumAugustenburger Platz 1Berlin13353Germany
| | - Amelie Beblo
- Department of Internal Medicine and CardiologyCharité – Universitätsmedizin Berlin, Campus Virchow KlinikumAugustenburger Platz 1Berlin13353Germany
| | - Evgeny Belyavskiy
- Department of Internal Medicine and CardiologyCharité – Universitätsmedizin Berlin, Campus Virchow KlinikumAugustenburger Platz 1Berlin13353Germany
| | - Elisabeth Pieske‐Kraigher
- Department of Internal Medicine and CardiologyCharité – Universitätsmedizin Berlin, Campus Virchow KlinikumAugustenburger Platz 1Berlin13353Germany
| | - Alexander Krannich
- Experimental and Clinical Research Center (ECRC)Charité – Universitätsmedizin BerlinBerlinGermany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and ToxicologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/LübeckHamburgGermany
| | - Sarah Hinz
- Institute of Clinical Pharmacology and ToxicologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Burkert Pieske
- Department of Internal Medicine and CardiologyCharité – Universitätsmedizin Berlin, Campus Virchow KlinikumAugustenburger Platz 1Berlin13353Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Department of CardiologyDeutsches Herzzentrum Berlin (DHZB)BerlinGermany
| | - Frank Edelmann
- Department of Internal Medicine and CardiologyCharité – Universitätsmedizin Berlin, Campus Virchow KlinikumAugustenburger Platz 1Berlin13353Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
| |
Collapse
|
12
|
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: 11] [Impact Index Per Article: 5.5] [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
|
13
|
Low-Dose Tacrolimus Promotes the Migration and Invasion and Nitric Oxide Production in the Human-Derived First Trimester Extravillous Trophoblast Cells In Vitro. Int J Mol Sci 2022; 23:ijms23158426. [PMID: 35955565 PMCID: PMC9369346 DOI: 10.3390/ijms23158426] [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: 08/16/2021] [Revised: 07/12/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Placentation is one of the most important determinants for a successful pregnancy, and this is dependent on the process of trophoblast migration and invasion. Progesterone receptors (PGR) are critical effectors of progesterone (P4) signaling that is required for trophoblast migration and invasion conducive to a successful gestation. In immune complicated pregnancies, evidence has shown that abnormal placentation occurs because of aberrant expression of PGR. Therapeutic intervention with tacrolimus (FK506) was able to restore PGR expression and improve pregnancy outcomes in immune-complicated gestations; however, the exact mode of action of tacrolimus in assisting placentation is not clear. Here, we attempt to uncover the mode of action of tacrolimus by examining its effects on trophoblast invasion and migration in the human-derived extravillous trophoblast (EVT) cell line, the HTR-8/SVneo cells. Using a variety of functional assays, we demonstrated that low-dose tacrolimus (10 ng/mL) was sufficient to significantly (p < 0.001) stimulate the migration and invasion of the HTR-8/SVneo cells, inducing their cytosolic/nuclear progesterone receptor expression and activation, and modulating their Nitric Oxide (NO) production. Moreover, tacrolimus abrogated the suppressive effect of the NOS inhibitor Nω- Nitro-L-Arginine Methyl Ester (L-NAME) on these vital processes critically involved in the establishment of human pregnancy. Collectively, our data suggest an immune-independent mode of action of tacrolimus in positively influencing placentation in complicated gestations, at least in part, through promoting the migration and invasion of the first trimester extravillous trophoblast cells by modulating their NO production and activating their cytosolic/nuclear progesterone-receptors. To our knowledge, this is the first report to show that the mode of action of tacrolimus as a monotherapy for implantation failure is plausibly PGR-dependent.
Collapse
|
14
|
Bonnitcha P, Sullivan D, Fitzpatrick M, Ireland A, Nguyen VL, Koay YC, O'Sullivan J. Design and validation of an LC-MS/MS method for simultaneous quantification of asymmetric dimethylguanidino valeric acid, asymmetric dimethylarginine and symmetric dimethylarginine in human plasma. Pathology 2022; 54:591-598. [DOI: 10.1016/j.pathol.2022.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 10/18/2022]
|
15
|
Homoarginine and blood pressure: a 10-year prospective relationship in normotensives. J Hum Hypertens 2022; 36:135-143. [PMID: 34040153 DOI: 10.1038/s41371-020-00449-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/23/2020] [Accepted: 11/08/2020] [Indexed: 02/06/2023]
Abstract
Nitric oxide plays a major role in the regulation of blood pressure, and impaired nitric oxide bioavailability contributes to the development of hypertension (HT). Various factors may contribute to nitric oxide bioavailability-including availability of the substrate for nitric oxide synthesis, L-arginine and its homolog L-homoarginine. We investigated whether blood pressure after 10 years associates with baseline L-homoarginine in participants who remained normotensive (NT) or developed HT, respectively. Data from the South African leg of the Prospective Urban and Rural Epidemiology study, performed in the North-West Province, were used. We investigated participants who either remained NT (N = 166) or who developed HT (N = 166) over 10 years. Blood pressure was measured with validated OMRON devices and serum L-homoarginine was analyzed with liquid chromatography-tandem mass spectrometry. L-homoarginine levels were similar at baseline (p = 0.39) and follow-up (p = 0.93) between NT and hypertensive groups. In the group that remained NT after 10 years, baseline L-homoarginine correlated positively with follow-up brachial systolic blood pressure (adj.R2 = 0.13; β = 0.33; p = 0.001), brachial pulse pressure (adj.R2 = 0.15 β = 0.40; p = 0.001), and central pulse pressure (adj.R2 = 0.20; β = 0.30; p = 0.003). No significant associations were found in the group that developed HT after 10 years. We found a positive, independent association between blood pressure and L-homoarginine in a group that remained NT, but not in a group that developed HT after 10 years. This may suggest a protective role for L-homoarginine to maintain normal blood pressure, but only to a certain level. Once HT develops other factors may overshadow the protective effects of L-homoarginine.
Collapse
|
16
|
Hannemann J, Siques P, Schmidt-Hutten L, Zummack J, Brito J, Böger R. Association of Genes of the NO Pathway with Altitude Disease and Hypoxic Pulmonary Hypertension. J Clin Med 2021; 10:jcm10245761. [PMID: 34945057 PMCID: PMC8704804 DOI: 10.3390/jcm10245761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 01/03/2023] Open
Abstract
Chronic intermittent hypoxia leads to high-altitude pulmonary hypertension, which is associated with high asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthesis. Therefore, we aimed to understand the relation of single nucleotide polymorphisms in this pathway to high-altitude pulmonary hypertension (HAPH). We genotyped 69 healthy male Chileans subjected to chronic intermittent hypoxia. Acclimatization to altitude was determined using the Lake Louise Score and the presence of acute mountain sickness. Echocardiography was performed after six months in 24 individuals to estimate pulmonary arterial pressure. The minor allele of dimethylarginine dimethylaminohydrolase (DDAH)1 rs233112 was associated with high-baseline plasma ADMA concentration, while individuals homozygous for the major allele of DDAH2 rs805304 had a significantly greater increase in ADMA during chronic intermittent hypoxia. The major allele of alanine glyoxylate aminotransferase-2 (AGXT2) rs37369 was associated with a greater reduction of plasma symmetric dimethylarginine (SDMA). Several genes were associated with high-altitude pulmonary hypertension, and the nitric oxide synthase (NOS)3 and DDAH2 genes were related to acute mountain sickness. In conclusion, DDAH1 determines baseline plasma ADMA, while DDAH2 modulates ADMA increase in hypoxia. AGXT2 may be up-regulated in hypoxia. Genomic variation in the dimethylarginine pathway affects the development of HAPH and altitude acclimatization.
Collapse
Affiliation(s)
- Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.-H.); (J.Z.); (R.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20246 Hamburg, Germany and Iquique 1100000, Chile; (P.S.); (J.B.)
- Correspondence:
| | - Patricia Siques
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20246 Hamburg, Germany and Iquique 1100000, Chile; (P.S.); (J.B.)
- Institute of Health Studies, Universidad Arturo Prat, Iquique 1100000, Chile
| | - Lena Schmidt-Hutten
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.-H.); (J.Z.); (R.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20246 Hamburg, Germany and Iquique 1100000, Chile; (P.S.); (J.B.)
| | - Julia Zummack
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.-H.); (J.Z.); (R.B.)
| | - Julio Brito
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20246 Hamburg, Germany and Iquique 1100000, Chile; (P.S.); (J.B.)
- Institute of Health Studies, Universidad Arturo Prat, Iquique 1100000, Chile
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.-H.); (J.Z.); (R.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20246 Hamburg, Germany and Iquique 1100000, Chile; (P.S.); (J.B.)
| |
Collapse
|
17
|
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
|
18
|
Arginine metabolism and nitric oxide turnover in the ZSF1 animal model for heart failure with preserved ejection fraction. Sci Rep 2021; 11:20684. [PMID: 34667218 PMCID: PMC8526609 DOI: 10.1038/s41598-021-00216-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/07/2021] [Indexed: 12/21/2022] Open
Abstract
Endothelial dysfunction and altered nitric oxide (NO) metabolism are considered causal factors in heart failure with preserved ejection fraction (HFpEF). NO synthase activity depends on the availability of arginine and its derivatives. Thus, we analyzed arginine, associated metabolites, arginine-metabolizing enzymes and NO turnover in 20-week-old female healthy lean (L-ZSF1) and obese ZSF1 rats (O-ZSF1) with HFpEF. Serum, urine and lysates of liver, kidney and heart were analyzed. There were significantly lower lysine (− 28%), arginine (− 31%), homoarginine (− 72%) and nitrite (− 32%) levels in serum of O-ZSF1 rats. Ornithine (+ 60%) and citrulline (+ 20%) levels were higher. Similar results were found in the heart. Expression of arginine consuming enzymes in liver and kidney was unchanged. Instead, we observed a 5.8-fold higher arginase 1 expression, presumably of granulocyte origin, in serum and > fourfold increased cardiac macrophage invasion in O-ZSF1. We conclude that inflammatory cells in blood and heart consume arginine and probably homoarginine via arginase 1 and inducible NO synthase and release ornithine and citrulline. In combination with evidence for decreased NO turnover in O-ZSF1 rats, we assume lower arginine bioavailability to endothelial NO synthase.
Collapse
|
19
|
Yola IM, Moser C, Duncan MS, Schwedhelm E, Atzler D, Maas R, Hannemann J, Böger RH, Vasan RS, Xanthakis V. Associations of circulating dimethylarginines with the metabolic syndrome in the Framingham Offspring study. PLoS One 2021; 16:e0254577. [PMID: 34492019 PMCID: PMC8423279 DOI: 10.1371/journal.pone.0254577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/29/2021] [Indexed: 01/04/2023] Open
Abstract
Background Circulating levels of the endogenous inhibitor of nitric oxide synthase, asymmetric dimethylarginine (ADMA), are positively associated with the prevalence of metabolic syndrome (MetS) in cross-sectional investigations. It is unclear if circulating ADMA and other methylarginines are associated with incident MetS prospectively. Methods We related circulating ADMA, symmetric dimethylarginine (SDMA), L-arginine (ARG) concentrations (measured with a validated tandem mass spectrometry assay) and the ARG/ADMA ratio to MetS and its components in 2914 (cross-sectional analysis, logistic regression; mean age 58 years, 55% women) and 1656 (prospective analysis, Cox regression; mean age 56 years, 59% women) individuals from the Framingham Offspring Study who attended a routine examination. Results Adjusting for age, sex, smoking, and eGFR, we observed significant associations of ADMA (direct) and ARG/ADMA (inverse) with odds of MetS (N = 1461 prevalent cases; Odds Ratio [OR] per SD increment 1.13, 95%CI 1.04–1.22; and 0.89, 95%CI 0.82–0.97 for ADMA and ARG/ADMA, respectively). Upon further adjustment for waist circumference, systolic and diastolic blood pressure, glucose, high-density lipoprotein cholesterol, and triglycerides, we observed a positive relation between SDMA and MetS (OR per SD increment 1.15, 95% CI 1.01–1.30) but the other associations were rendered statistically non-significant. We did not observe statistically significant associations between any of the methylarginines and the risk of new-onset MetS (752 incident events) over a median follow-up of 11 years. Conclusion It is unclear whether dimethylarginines play an important role in the incidence of cardiometabolic risk in the community, notwithstanding cross-sectional associations. Further studies of larger samples are needed to replicate our findings.
Collapse
Affiliation(s)
- Ibrahim Musa Yola
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, United States of America
| | - Carlee Moser
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| | - Meredith S. Duncan
- Department of Biostatistics, University of Kentucky, Lexington, KY, United States of America
| | - Edzard Schwedhelm
- Department 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
| | - Dorothee Atzler
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), Partner Site Munich Heart Alliance, Munich, Germany
| | - Renke Maas
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Juliane Hannemann
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rainer H. Böger
- Department 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
| | - Ramachandran S. Vasan
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, United States of America
- Framingham Heart Study, Framingham, MA, United States of America
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States of America
- Boston University Center for Computing and Data Sciences, Boston, MA, United States of America
| | - Vanessa Xanthakis
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, United States of America
- Framingham Heart Study, Framingham, MA, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
20
|
Mader MM, Böger R, Appel D, Schwedhelm E, Haddad M, Mohme M, Lamszus K, Westphal M, Czorlich P, Hannemann J. Intrathecal and systemic alterations of L-arginine metabolism in patients after intracerebral hemorrhage. J Cereb Blood Flow Metab 2021; 41:1964-1977. [PMID: 33461409 PMCID: PMC8327100 DOI: 10.1177/0271678x20983216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Alterations in the concentration of nitric oxide (NO) and L-arginine metabolites have been associated with the pathophysiology of different vascular diseases. Here, we describe striking changes in L-arginine metabolism after hemorrhagic stroke. Blood and cerebrospinal fluid (CSF) samples of patients with intracerebral hemorrhage (ICH) and/or intraventricular hemorrhage were collected over a ten-day period. Liquid chromatography-tandem mass spectrometry was used to quantify key substrates and products of L-arginine metabolizing enzymes as well as asymmetric (ADMA) and symmetric dimethylarginine (SDMA). Changes in the plasma were limited to early reductions in L-ornithine, L-lysine, and L-citrulline concentrations. Intrathecally, we observed signs of early NO synthase (NOS) upregulation followed by a decrease back to baseline accompanied by a rise in the level of its endogenous NOS-inhibitor ADMA. SDMA demonstrated increased levels throughout the observation period. For arginase, a pattern of persistently elevated activity was measured and arginine:glycine amidinotransferase (AGAT) appeared to be reduced in its activity at later time points. An early reduction in CSF L-arginine concentration was an independent risk factor for poor outcome. Together, these findings further elucidate pathophysiological mechanisms after ICH potentially involved in secondary brain injury and may reveal novel therapeutic targets.
Collapse
Affiliation(s)
- Marius M Mader
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel Appel
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Munif Haddad
- Department of Clinical Chemistry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Mohme
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Westphal
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Patrick Czorlich
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
21
|
Appel D, Böger R, Windolph J, Heinze G, Goetz AE, Hannemann J. Asymmetric dimethylarginine predicts perioperative cardiovascular complications in patients undergoing medium-to-high risk non-cardiac surgery. J Int Med Res 2021; 48:300060520940450. [PMID: 32842812 PMCID: PMC7453459 DOI: 10.1177/0300060520940450] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Objectives Perioperative cardiovascular events remain an important factor that affects surgery outcome. We assessed if asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthesis, predicts perioperative risk, and if pre-operative supplementation with L-arginine/L-citrulline improves the plasma L-arginine/ADMA ratio. Methods In this prospective study, planned thoracic and/or abdominal surgery patients were randomized to receive L-arginine/L-citrulline (5 g/day) or placebo 1 to 5 days before surgery. We measured perioperative plasma ADMA and L-arginine levels. The primary outcome was a 30-day combined cardiovascular endpoint. Results Among 269 patients, 23 (8.6%) experienced a major adverse cardiovascular event. ADMA and C-reactive protein were significantly associated with the incidence of cardiovascular complications in the multivariable-adjusted analysis. The L-arginine plasma concentration was significantly higher on the day of surgery with L-arginine/L-citrulline supplementation compared with placebo. In patients with high pre-operative ADMA, there was a non-significant trend towards reduced incidence of the primary endpoint with L-arginine/L-citrulline supplementation (six vs. nine events). Conclusions ADMA is a predictor of major adverse cardiovascular complications in the perioperative period for patients who are undergoing major abdominal and/or thoracic surgery. Supplementation with L-arginine/L-citrulline increased the L-arginine plasma concentration, enhanced the L-arginine/ADMA ratio, and induced a trend towards fewer perioperative events.
Collapse
Affiliation(s)
- Daniel Appel
- University Medical Center Hamburg-Eppendorf, Department of Anesthesiology, Hamburg, DE, Germany
| | - Rainer Böger
- University Medical Center Hamburg-Eppendorf, Institute of Clinical Pharmacology and Toxicology, Hamburg, DE, Germany
| | - Julia Windolph
- University Medical Center Hamburg-Eppendorf, Institute of Clinical Pharmacology and Toxicology, Hamburg, DE, Germany
| | - Gina Heinze
- University Medical Center Hamburg-Eppendorf, Department of Anesthesiology, Hamburg, DE, Germany
| | - Alwin E Goetz
- University Medical Center Hamburg-Eppendorf, Department of Anesthesiology, Hamburg, DE, Germany
| | - Juliane Hannemann
- University Medical Center Hamburg-Eppendorf, Institute of Clinical Pharmacology and Toxicology, Hamburg, DE, Germany
| |
Collapse
|
22
|
Castro-Diehl C, Ehrbar R, Obas V, Oh A, Vasan RS, Xanthakis V. Biomarkers representing key aging-related biological pathways are associated with subclinical atherosclerosis and all-cause mortality: The Framingham Study. PLoS One 2021; 16:e0251308. [PMID: 33989340 PMCID: PMC8121535 DOI: 10.1371/journal.pone.0251308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/24/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Increased oxidative stress, leukocyte telomere length (LTL) shortening, endothelial dysfunction, and lower insulin-like growth factor (IGF)-1 concentrations reflect key molecular mechanisms of aging. We hypothesized that biomarkers representing these pathways are associated with measures of subclinical atherosclerosis and all-cause mortality. METHODS AND RESULTS We evaluated up to 2,314 Framingham Offspring Study participants (mean age 61 years, 55% women) with available biomarkers of aging: LTL, circulating concentrations of IGF-1, asymmetrical dimethylarginine (ADMA), and urinary F2-Isoprostanes indexed to urinary creatinine. We evaluated the association of each biomarker with coronary artery calcium [ln (CAC+1)] and carotid intima-media thickness (IMT). In multivariable-adjusted linear regression models, higher ADMA levels were associated with higher CAC values (βADMA per 1-SD increase 0.25; 95% confidence interval [CI] [0.11, 0.39]). Additionally, shorter LTL and lower IGF-1 values were associated with higher IMT values (βLTL -0.08, 95%CI -0.14, -0.02, and βIGF-1 -0.04, 95%CI -0.08, -0.01, respectively). During a median follow-up of 15.5 years, 593 subjects died. In multivariable-adjusted Cox regression models, LTL and IGF-1 values were inversely associated with all-cause mortality (hazard ratios [HR] per SD increase in biomarker, 0.85, 95% CI 0.74-0.99, and 0.90, 95% CI 0.82-0.98 for LTL and IGF-1, respectively). F2-Isoprostanes and ADMA values were positively associated with all-cause mortality (HR per SD increase in biomarker, 1.15, 95% CI, 1.10-1.22, and 1.10, 95% CI, 1.02-1.20, respectively). CONCLUSION In our prospective community-based study, aging-related biomarkers were associated with measures of subclinical atherosclerosis cross-sectionally and with all-cause mortality prospectively, supporting the concept that these biomarkers may reflect the aging process in community-dwelling adults.
Collapse
Affiliation(s)
- Cecilia Castro-Diehl
- Department of Medicine, Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Rachel Ehrbar
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Vanesa Obas
- Department of Medicine, Internal Medicine Residency Program, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Albin Oh
- Department of Medicine, Internal Medicine Residency Program, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Ramachandran S. Vasan
- Department of Medicine, Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Lung, and Blood Institute’s Framingham Heart Study, Boston University’s and National Heart, Framingham, Massachusetts, United States of America
- Department of Medicine, Section of Cardiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Vanessa Xanthakis
- Department of Medicine, Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- Lung, and Blood Institute’s Framingham Heart Study, Boston University’s and National Heart, Framingham, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
23
|
Hannemann J, Balfanz P, Schwedhelm E, Hartmann B, Ule J, Müller-Wieland D, Dahl E, Dreher M, Marx N, Rainer Böger. Elevated serum SDMA and ADMA at hospital admission predict in-hospital mortality of COVID-19 patients. Sci Rep 2021; 11:9895. [PMID: 33972591 PMCID: PMC8110746 DOI: 10.1038/s41598-021-89180-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
COVID-19 is a disease with a variable clinical course ranging from mild symptoms to critical illness, organ failure, and death. Prospective biomarkers may help to predict the severity of an individual’s clinical course and mortality risk. We analyzed asymmetric (ADMA) and symmetric dimethylarginine (SDMA) in blood samples from 31 patients hospitalized for COVID-19. We calculated associations of ADMA and SDMA with mortality and organ failure, and we developed a predictive algorithm based upon these biomarkers to predict mortality risk. Nine patients (29%) experienced in-hospital death. SDMA and ADMA serum concentrations were significantly higher at admission in COVID-19 patients who died than in survivors. Cut-offs of 0.90 µmol/L for SDMA (AUC, 0.904, p = 0.0005) and 0.66 µmol/L for ADMA (AUC, 0.874, p = 0.0013) were found in ROC analyses to best discriminate both subgroups of patients. Hazard ratio for in-hospital mortality was 12.2 (95% CI: 2.2–31.2) for SDMA and 6.3 (1.1–14.7) for ADMA above cut-off. Sequential analysis of both biomarkers allowed discriminating a high-risk group (87.5% mortality) from an intermediate-risk group (25% mortality) and a low-risk group (0% mortality). Elevated circulating concentrations of SDMA and ADMA may help to better identify COVID-19 patients with a high risk of in-hospital mortality.
Collapse
Affiliation(s)
- Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany. .,Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, Hamburg, Germany.
| | - Paul Balfanz
- Department of Cardiology, Angiology and Intensive Care Medicine, Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Bojan Hartmann
- Department of Cardiology, Angiology and Intensive Care Medicine, Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Johanna Ule
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Dirk Müller-Wieland
- Department of Cardiology, Angiology and Intensive Care Medicine, Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Edgar Dahl
- Institute of Pathology, University Hospital Aachen, Aachen, Germany.,RWTH centralized Biomaterial Bank (RWTH cBMB), Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Michael Dreher
- Department of Pneumology and Intensive Care Medicine, Medical Clinic V, University Hospital Aachen, Aachen, Germany
| | - Nikolaus Marx
- Department of Cardiology, Angiology and Intensive Care Medicine, Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
24
|
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: 2.0] [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
|
25
|
Tůma P, Sommerová B, Koval D, Couderc F. Electrophoretic Determination of Symmetric and Asymmetric Dimethylarginine in Human Blood Plasma with Whole Capillary Sample Injection. Int J Mol Sci 2021; 22:2970. [PMID: 33804011 PMCID: PMC7998904 DOI: 10.3390/ijms22062970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/16/2022] Open
Abstract
Asymmetric and symmetric dimethylarginines are toxic non-coded amino acids. They are formed by post-translational modifications and play multifunctional roles in some human diseases. Their determination in human blood plasma is performed using capillary electrophoresis with contactless conductivity detection. The separations are performed in a capillary covered with covalently bonded PAMAPTAC polymer, which generates anionic electroosmotic flow and the separation takes place in the counter-current regime. The background electrolyte is a 750 mM aqueous solution of acetic acid with pH 2.45. The plasma samples for analysis are treated by the addition of acetonitrile and injected into the capillary in a large volume, reaching 94.5% of the total volume of the capillary, and subsequently subjected to electrophoretic stacking. The attained LODs are 16 nm for ADMA and 22 nM for SDMA. The electrophoretic resolution of both isomers has a value of 5.3. The developed method is sufficiently sensitive for the determination of plasmatic levels of ADMA and SDMA. The determination does not require derivatization and the individual steps in the electrophoretic stacking are fully automated. The determined plasmatic levels for healthy individuals vary in the range 0.36-0.62 µM for ADMA and 0.32-0.70 µM for SDMA.
Collapse
Affiliation(s)
- Petr Tůma
- Department of Hygiene, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague 10, Czech Republic;
| | - Blanka Sommerová
- Department of Hygiene, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague 10, Czech Republic;
| | - Dušan Koval
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague 6, Czech Republic;
| | - François Couderc
- Laboratoire des IMRCP UMR 5623, University Toulouse III-Paul Sabatier, 31062 Toulouse, France;
| |
Collapse
|
26
|
Hannemann J, Glatzel A, Hillig J, Zummack J, Schumacher U, Lüneburg N, Harbaum L, Böger R. Upregulation of DDAH2 Limits Pulmonary Hypertension and Right Ventricular Hypertrophy During Chronic Hypoxia in Ddah1 Knockout Mice. Front Physiol 2020; 11:597559. [PMID: 33281630 PMCID: PMC7689360 DOI: 10.3389/fphys.2020.597559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
Objective: Chronic hypoxia causes pulmonary vasoconstriction leading to pulmonary hypertension and right ventricular hypertrophy. Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide (NO) synthesis; its level increases in hypoxia (HX) concomitantly with reduced activity of dimethylarginine dimethylaminohydrolases (DDAH-1 and DDAH-2), enzymes metabolizing ADMA. Ddah1 knockout (KO) mice may therefore help to understand the pathophysiological roles of this enzyme and its substrate, ADMA, in the development of hypoxia-associated pulmonary hypertension. Methods: Ddah1 KO mice and their wild-type (WT) littermates were subjected to normoxia (NX) or for 21 days. We measured ADMA concentration in plasma and lungs, DDAH1 and DDAH2 mRNA and protein expression in the lungs, right ventricular systolic pressure (RVSP), right ventricular hypertrophy by the Fulton index, and cardiomyocyte hypertrophy by dystrophin staining of the heart. Results: Ddah1 KO mice had higher ADMA concentrations in plasma and in lung tissue than WT in NX (p < 0.05). ADMA significantly increased in WT-HX in plasma and lungs, while there were no significant differences in WT-HX vs. KO-HX. This finding was paralleled by a 38 ± 13% reduction in Ddah1 but not Ddah2 mRNA expression, and reduced DDAH1 protein expression but stable DDAH2 protein levels in WT mice. Ddah1 KO mice showed significant elevation of DDAH2 protein but not mRNA levels, which further increased in HX. HX led to increased RVSP and right ventricular hypertrophy in both, WT and KO mice, with no significant differences between both genotypes. Conclusions: Chronic hypoxia causes an elevation of ADMA, which may impair NO production and lead to endothelial dysfunction and vasoconstriction. Downregulation of DDAH1 expression and activity may be involved in this; however, knockout of the Ddah1 gene does not modify the hypoxia-induced pathophysiological changes of pulmonary blood pressure and right ventricular hypertrophy, possibly due to compensatory upregulation of DDAH2 protein.
Collapse
Affiliation(s)
- Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute DECIPHER (German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae), Hamburg, Germany
| | - Antonia Glatzel
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas Hillig
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute DECIPHER (German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae), Hamburg, Germany
| | - Julia Zummack
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute DECIPHER (German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae), Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicole Lüneburg
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Harbaum
- Department of Pulmonology, II. Medical Clinic and Policlinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute DECIPHER (German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae), Hamburg, Germany
| |
Collapse
|
27
|
LC-MS/MS quantification of asymmetric dimethyl arginine and symmetric dimethyl arginine in plasma using surrogate matrix and derivatization with fluorescamine. Bioanalysis 2020; 12:1607-1619. [PMID: 33151745 DOI: 10.4155/bio-2020-0223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aim: A novel LC-MS/MS method using a surrogate matrix and derivatization with fluorescamine was developed and validated for simultaneous quantification of asymmetric dimethyl arginine and symmetric dimethyl arginine. Methods & results: Asymmetric dimethyl arginine, symmetric dimethyl arginine and corresponding internal standards were extracted using protein precipitation and derivatization with fluorescamine followed by SPE. Derivatives were analyzed by turbo ion spray LC-MS/MS in the positive ion mode. Methodology was successfully transferred across multiple preclinical species and utilized in the support of several investigative studies. Conclusion: A new LC-MS/MS analytical methodology that utilizes a surrogate matrix and derivatization with fluorescamine was successfully developed and validated.
Collapse
|
28
|
Tun B, Ehrbar R, Short M, Cheng S, Vasan RS, Xanthakis V. Association of Exhaled Carbon Monoxide With Ideal Cardiovascular Health, Circulating Biomarkers, and Incidence of Heart Failure in the Framingham Offspring Study. J Am Heart Assoc 2020; 9:e016762. [PMID: 33100134 PMCID: PMC7763395 DOI: 10.1161/jaha.120.016762] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Exhaled carbon monoxide (eCO) is directly associated with traditional cardiovascular disease risk factors and incident cardiovascular disease. However, its relation with the cardiovascular health score and incidence of heart failure (HF) has not been investigated. Methods and Results We measured eCO in 3521 Framingham Heart Study Offspring participants attending examination cycle 6 (mean age 59 years, 53% women). We related the cardiovascular health score (composite of blood pressure, fasting plasma glucose, total cholesterol, body mass index, smoking, diet, and physical activity) to eCO adjusting for age, sex, and smoking. Higher cardiovascular health scores were associated with lower eCO (β=-0.02, P<0.0001), even among nonsmokers. Additionally, C-reactive protein, plasminogen activator inhibitor-1, fibrinogen, growth differentiation factor-15, homocysteine, and asymmetrical dimethylarginine were positively associated with eCO (P≤0.003 for all). The age- and sex-adjusted and multivariable-adjusted heritabilities of eCO were 49.5% and 31.4%, respectively. Over a median follow-up of 18 years, 309 participants (45% women) developed HF. After multivariable adjustment, higher eCO was associated with higher risk of HF (hazards ratio per SD increment: 1.39; 95% CI, 1.19-1.62 [P<0.001]) and with higher risk of HF with reduced ejection fraction (N=144 events; hazard ratio per SD increment in eCO: 1.43; 95% CI, 1.15-1.77 [P=0.001]). Conclusions In our community-based sample, higher levels of eCO were associated with lower cardiovascular health scores, an adverse cardiovascular biomarker profile, and a higher risk of HF, specifically HF with reduced ejection fraction. Our findings suggest that carbon monoxide may identify a novel pathway to HF development.
Collapse
Affiliation(s)
- Bradley Tun
- Department of Medicine Boston University School of Medicine and Boston Medical Center Boston MA
| | - Rachel Ehrbar
- Department of Biostatistics Boston University School of Public Health Boston MA
| | - Meghan Short
- Glenn Biggs Institute for Alzheimer's Disease & Neurodegenerative Diseases University of Texas Health Sciences Center San Antonio TX
| | - Susan Cheng
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Ramachandran S Vasan
- National Heart, Blood and Lung Institute Framingham Heart Study Framingham MA.,Sections of Preventive Medicine and Epidemiology and Cardiovascular Medicine Department of Medicine Boston University School of Medicine Boston MA.,Department of Epidemiology Boston University School of Public Health Boston MA
| | - Vanessa Xanthakis
- Department of Biostatistics Boston University School of Public Health Boston MA.,National Heart, Blood and Lung Institute Framingham Heart Study Framingham MA.,Sections of Preventive Medicine and Epidemiology and Cardiovascular Medicine Department of Medicine Boston University School of Medicine Boston MA
| |
Collapse
|
29
|
Du Q, Gao J, Lu R, Jin Y, Zou Y, Yu C, Yan Y. Asymmetric dimethylarginine compartmental behavior during high-flux hemodialysis. Ren Fail 2020; 42:760-766. [PMID: 32727241 PMCID: PMC7470094 DOI: 10.1080/0886022x.2020.1797790] [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: 04/07/2020] [Revised: 06/21/2020] [Accepted: 07/10/2020] [Indexed: 11/09/2022] Open
Abstract
AIM The accumulation of uremic toxins, such as asymmetric dimethylarginine (ADMA), has emerged as one of the major cardiovascular disease-related risk factors in patients with end-stage renal disease (ESRD). Based on the low molecular weight of ADMA, hemodialysis (HD) should theoretically effectively remove ADMA. In this study, we investigated the clearance behavior of ADMA during high-flux HD. METHODS Eight HD patients without residual renal function were included. Blood samples were collected at 0, 30, 60, 120 and 240 min after dialysis started, as well as 1 h and 48 h after dialysis. ADMA level was detected by HPLC-MS/MS. Herein, the ADMA level in blood cells and the ADMA protein binding rate were measured. Accordingly, the dialyzer extraction ratio was also determined. RESULTS The reduction ratio (RR) of ADMA (corrected for hemoconcentration) was significantly lower, at only 37.21 ± 6.44%, than that of urea and creatinine (p < .05). Interestingly, its clearance from plasma was precipitous early in dialysis and became slowly from 60 to 240 min. Additionally, a greater inlet erythrocyte than plasma concentration was found for ADMA. The dialyzer extraction ratio was comparable between ADMA and creatinine or urea (83 ± 5% for ADMA vs. 84 ± 3% and 88 ± 2% for creatinine and urea, respectively; both p>.05). Urea and creatinine had a slight rebound ratio of less than 10% at 1 h after the completion of HD. In contrast, considerable rebound of approximately 30% was detected in ADMA. CONCLUSION This study suggests that ADMA may present a multicompartmental distribution that cannot be representatively reflected by the urea kinetics model.
Collapse
Affiliation(s)
- Qiuna Du
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiayuan Gao
- Department of Nephrology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Renhua Lu
- Department of Nephrology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yun Jin
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanfang Zou
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yucheng Yan
- Department of Nephrology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Bergia RE, Campbell WW, Roseguini BT, Kim JE. A high-protein meal does not improve blood pressure or vasoactive biomarker responses to acute exercise in humans. Nutr Res 2020; 81:97-107. [PMID: 32949918 DOI: 10.1016/j.nutres.2020.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/09/2020] [Accepted: 08/05/2020] [Indexed: 11/19/2022]
Abstract
Blood pressure (BP) responses to exercise yield prognostic information beyond resting BP. While habitual higher dietary protein intake is associated with reduced resting BP, few studies have assessed the impact of high-protein meals on acute BP and vasoactive biomarker responses to exercise. To test the hypothesis that consuming a higher-protein, lower fat meal (HP; 30 g protein, 17 g fat, 52 g carbohydrate) would attenuate the BP response to exercise and result in a more robust post-exercise hypotensive response compared to a lower-protein, higher-fat meal (LP; 13 g protein, 25 g fat, 54 g carbohydrate), we recruited 31 pre-hypertensive subjects to complete this randomized, double-blind, cross-over acute feeding study. One hundred sixty-five minutes after consuming the test HP or LP meal, subjects exercised on a cycle ergometer at 70% VO2 max for 30 minutes. Blood pressure was measured prior to the meal and periodically before, during, and after exercise for a 315-minute period. Blood samples were periodically collected to quantify plasma arginine, arginine metabolites (asymmetric dimethylarginine, symmetric dimethylarginine; ADMA, SDMA), endothelin-1, nitrates, and nitrites in a subset of subjects (n = 15) as shown in Supplemental Table S1. Consuming the HP meal did not influence the BP responses to exercise, including the post-exercise return to baseline BP or systolic BP area under the curve. While the HP meal resulted in greater postprandial plasma arginine concentrations, ADMA, SDMA, endothelin-1, nitrates, and nitrites were unaltered. These results suggest that consuming a higher-protein, lower-fat meal does not influence BP or vasoactive biomarker responses to exercise compared to a lower-protein, higher-fat meal.
Collapse
Affiliation(s)
- Robert E Bergia
- Department of Nutrition Science, Purdue University, 700 West State St, West Lafayette, IN 47907
| | - Wayne W Campbell
- Department of Nutrition Science, Purdue University, 700 West State St, West Lafayette, IN 47907.
| | - Bruno T Roseguini
- Department of Health & Kinesiology, Purdue University, 800 W Stadium Ave, West Lafayette, IN 47907
| | - Jung Eun Kim
- Food Science and Technology Programme, c/o Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543.
| |
Collapse
|
31
|
Ramiro-Cortijo D, de la Calle M, Böger R, Hannemann J, Lüneburg N, López-Giménez MR, Rodríguez-Rodríguez P, Martín-Cabrejas MÁ, Benítez V, de Pablo ÁLL, González MDC, Arribas SM. Male fetal sex is associated with low maternal plasma anti-inflammatory cytokine profile in the first trimester of healthy pregnancies. Cytokine 2020; 136:155290. [PMID: 32956948 DOI: 10.1016/j.cyto.2020.155290] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/19/2020] [Accepted: 09/06/2020] [Indexed: 01/01/2023]
Abstract
Male fetal sex associates with higher rates of materno-fetal complications. Inflammation and inadequate vasoactive responses are mechanisms implicated in obstetric complications, and alterations in maternal plasma cytokine profile and nitric oxide (NO) metabolites are potential predictive biomarkers. We aimed to assess if these parameters are influenced by fetal sex. A prospective, observational study was carried out in 85 healthy pregnant women with singleton pregnancies in the first trimester of gestation. A blood sample was extracted at the tenth week of gestation. In plasma, we assessed: 1) cytokines (micro-array): pro-inflammatory (IL1α, IL1 β, IL6, TNFα), anti-inflammatory (IL4, IL10, IL13), and chemoattractant (IL8, MCP1, IFNγ), and 2) NO metabolites (liquid chromatography-tandem mass spectrometry and Griess reaction): L-arginine, ADMA, SDMA, nitrates (NOx). Women with a male fetus (n = 50) exhibited, compared with those with a female (n = 35): higher IL1β (OR = 1.09 with 95% CI: 0.97-1.28), and lower IL13 (OR = 0.93 with 95% CI: 0.87-0.99), and higher plasma NOx (OR = 1.14 with 95% CI: 1.03-1.31). Our data suggest that fetal sex influences maternal plasma cytokine profile and NO in early pregnancy. Women with a male fetus may have a worse capacity to counteract an inflammatory response. They may have better vasodilator capacity, but in the presence of an oxidative environment, a higher nitrosative damage may occur. These data reinforce the need to include sex as variable in predictive models.
Collapse
Affiliation(s)
- David Ramiro-Cortijo
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - María de la Calle
- Obstetrics and Gynecology Service, La Paz University Hospital, Madrid, Spain
| | - Rainer Böger
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Juliane Hannemann
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicole Lüneburg
- Hospital Pharmacy, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - María Rosario López-Giménez
- Department of Preventive Medicine, Public Health & Microbiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | - Vanesa Benítez
- Institute of Food Science Research (CIAL) & Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | - Silvia M Arribas
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
32
|
Gamil S, Erdmann J, Schwedhelm E, Bakheit KH, Abdalrahman IBB, Mohamed AO. Increased Serum Levels of Asymmetric Dimethylarginine and Symmetric Dimethylarginine and Decreased Levels of Arginine in Sudanese Patients with Essential Hypertension. Kidney Blood Press Res 2020; 45:727-736. [PMID: 32814314 DOI: 10.1159/000508695] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 05/14/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Essential hypertension (EH) is a disease caused by various environmental and genetic factors. Nitric oxide (NO) is important for the functional integrity of the endothelium. It is produced in endothelial cells by endothelial NO synthase (eNOS) that mediates the conversion of the amino acid arginine into NO and citrulline. Asymmetric dimethylarginine (ADMA) acts as an inhibitor of eNOS. In contrast, symmetric dimethylarginine (SDMA) has no direct effect on eNOS but plays an important role competing with arginine for transport across the amino acid transporter. ADMA and SDMA have been found to play a central role in the development of cardiovascular diseases. Serum ADMA levels may serve as a future diagnostic marker and a target of therapy in hypertensive patients in the Sudanese population. This study aimed to investigate the relation between serum arginine, ADMA, and SDMA levels with EH in the Sudanese population. METHODS Patients (n = 260) with established hypertension and controls (n = 144) with normal blood pressure were included in this case-control study. Serum blood samples were analyzed for arginine, ADMA, and SDMA, using high-performance liquid chromatography-tandem mass spectrometry. Other laboratory data were measured using routine methods. Mann-Whitney's U test and χ2 tests were used for continuous and categorical data, respectively. A multivariate logistic regression analysis was conducted to investigate the independent effect of multiple variables on the development of hypertension. RESULTS Serum arginine levels were significantly lower in the patient group than in the control group (p < 0.001). ADMA and SDMA levels were significantly higher in the patient group than the control group (p < 0.001, p = 0.001, respectively). Multivariate logistic regression analysis showed that only older age, being a male, and arginine levels are independent factors controlling the development of hypertension (p < 0.001, p < 0.001, and p = 0.046, respectively). ADMA and SDMA levels were not independent factors for the development of hypertension. CONCLUSIONS This study demonstrated increased serum levels of ADMA and SDMA and decreased arginine levels in Sudanese patients with EH. Lowering serum ADMA levels or increasing the arginine levels might be a novel therapeutic target in these individuals.
Collapse
Affiliation(s)
- Sahar Gamil
- Department of Biochemistry, Faculty of Medicine, University of Khartoum, Khartoum, Sudan,
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg/Kiel/Lübeck, Germany.,University Heart Center Lübeck, Lübeck, Germany
| | - Edzard Schwedhelm
- DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg/Kiel/Lübeck, Germany.,Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Ihab B B Abdalrahman
- Department of Medicine, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Abdelrahim O Mohamed
- Department of Biochemistry, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| |
Collapse
|
33
|
Arginine derivatives in atrial fibrillation progression phenotypes. J Mol Med (Berl) 2020; 98:999-1008. [PMID: 32504111 PMCID: PMC8556202 DOI: 10.1007/s00109-020-01932-9] [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: 03/02/2020] [Revised: 05/20/2020] [Accepted: 05/27/2020] [Indexed: 10/24/2022]
Abstract
Arginine, homoarginine (hArg), asymmetric dimethylarginine (ADMA), and symmetric dimethylarginine (SDMA) affect nitric oxide metabolism and altered concentrations are associated with cardiovascular morbidity and mortality. We analyzed these metabolites using liquid chromatography-tandem mass spectrometry in patients with atrial fibrillation (AF) (n = 241) with a focus on heart rhythm at blood withdrawal, AF progression phenotypes, and successful sinus rhythm (SR) restoration (n = 22). AF progression phenotypes were defined as paroxysmal AF with/without low voltage areas (LVA) and persistent AF with/without LVA. While arginine, ADMA, and hArg were within reference limits for healthy controls, SDMA was higher in the AF cohort (0.57 ± 0.12 vs. 0.53 μmol/L (97.5th percentile in reference cohort)). SR restoration in AF patients resulted in normalization of SDMA concentrations (0.465 ± 0.082 vs. 0.570 ± 0.134 μmol/L at baseline, p < 0.001). Patients with AF at the time of blood sampling had significantly lower hArg (1.65 ± 0.51 vs. 1.85 ± 0.60 μmol/L, p = 0.006) and higher ADMA concentrations (0.526 ± 0.08 vs. 0.477 ± 0.08 μmol/L, p < 0.001) compared with AF patients in SR. hArg concentrations were lower in patients with advanced AF progression phenotypes (persistent AF with LVA (p = 0.046)) independent of heart rhythm at blood sampling. Summarizing, arginine metabolism imbalance is associated with AF in general and AF progression and may contribute to associated risk. KEY MESSAGES: • Heart rhythm at blood withdrawal affects ADMA and hArg level in AF patients. • SDMA is higher in AF patients. • SDMA levels normalize after sinus rhythm restoration. • hArg levels decrease in advanced AF progression phenotypes.
Collapse
|
34
|
Sgarra L, Bortone AS, Potenza MA, Nacci C, De Salvia MA, Acquaviva T, De Cillis E, Ciccone MM, Grimaldi M, Montagnani M. Endothelial Dysfunction May Link Interatrial Septal Abnormalities and MTHFR-Inherited Defects to Cryptogenic Stroke Predisposition. Biomolecules 2020; 10:biom10060861. [PMID: 32512924 PMCID: PMC7355772 DOI: 10.3390/biom10060861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/22/2020] [Accepted: 06/02/2020] [Indexed: 12/21/2022] Open
Abstract
We explored the significance of the L-Arginine/asymmetric dimethylarginine (L-Arg/ADMA) ratio as a biomarker of endothelial dysfunction in stroke patients. To this aim, we evaluated the correlation, in terms of severity, between the degree of endothelial dysfunction (by L-Arg/ADMA ratio), the methylene tetrahydrofolate reductase (MTHFR) genotype, and the interatrial septum (IAS) phenotype in subject with a history of stroke. Methods and Results: L-Arg, ADMA, and MTHFR genotypes were evaluated; the IAS phenotype was assessed by transesophageal echocardiography. Patients were grouped according to the severity of IAS defects and the residual enzymatic activity of MTHFR-mutated variants, and values of L-Arg/ADMA ratio were measured in each subgroup. Of 57 patients, 10 had a septum integrum (SI), 38 a patent foramen ovale (PFO), and 9 an ostium secundum (OS). The L-Arg/ADMA ratio differed across septum phenotypes (p ≤ 0.01), and was higher in SI than in PFO or OS patients (p ≤ 0.05, p ≤ 0.01, respectively). In the PFO subgroup a negative correlation was found between the L-Arg/ADMA ratio and PFO tunnel length/height ratio (p ≤ 0.05; r = − 0.37; R2 = 0.14). Interestingly, the L-Arg/ADMA ratio varied across MTHFR genotypes (p ≤ 0.0001) and was lower in subgroups carrying the most impaired enzyme with respect to patients carrying the conservative MTHFR (p ≤ 0.0001, p ≤ 0.05, respectively). Consistently, OS patients carried the most dysfunctional MTHFR genotypes, whereas SI patients the least ones. Conclusions: A low L-Arg/ADMA ratio correlates with impaired activity of MTHFR and with the jeopardized IAS phenotype along a severity spectrum encompassing OS, PFO with long/tight tunnel, PFO with short/large tunnel, and SI. This infers that genetic MTHFR defects may underlie endothelial dysfunction-related IAS abnormalities, and predispose to a cryptogenic stroke. Our findings emphasize the role of the L-Arg/ADMA ratio as a reliable marker of stroke susceptibility in carriers of IAS abnormalities, and suggest its potential use both as a diagnostic tool and as a decision aid for therapy.
Collapse
Affiliation(s)
- Luca Sgarra
- Department of Biomedical Sciences and Human Oncology—Section of Pharmacology, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (L.S.); (M.A.P.); (C.N.); (M.A.D.S.)
| | - Alessandro Santo Bortone
- Department of Emergency and Organ Transplantation—Section of Cardiovascular Diseases, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.B.); (T.A.); (E.D.C.); (M.M.C.)
| | - Maria Assunta Potenza
- Department of Biomedical Sciences and Human Oncology—Section of Pharmacology, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (L.S.); (M.A.P.); (C.N.); (M.A.D.S.)
| | - Carmela Nacci
- Department of Biomedical Sciences and Human Oncology—Section of Pharmacology, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (L.S.); (M.A.P.); (C.N.); (M.A.D.S.)
| | - Maria Antonietta De Salvia
- Department of Biomedical Sciences and Human Oncology—Section of Pharmacology, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (L.S.); (M.A.P.); (C.N.); (M.A.D.S.)
| | - Tommaso Acquaviva
- Department of Emergency and Organ Transplantation—Section of Cardiovascular Diseases, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.B.); (T.A.); (E.D.C.); (M.M.C.)
| | - Emanuela De Cillis
- Department of Emergency and Organ Transplantation—Section of Cardiovascular Diseases, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.B.); (T.A.); (E.D.C.); (M.M.C.)
| | - Marco Matteo Ciccone
- Department of Emergency and Organ Transplantation—Section of Cardiovascular Diseases, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.B.); (T.A.); (E.D.C.); (M.M.C.)
| | - Massimo Grimaldi
- General Hospital “F. Miulli” Acquaviva delle Fonti, 70021 Bari, Italy;
| | - Monica Montagnani
- Department of Biomedical Sciences and Human Oncology—Section of Pharmacology, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (L.S.); (M.A.P.); (C.N.); (M.A.D.S.)
- Correspondence:
| |
Collapse
|
35
|
Myeloid-Derived Suppressor Cells Mediate Immunosuppression After Cardiopulmonary Bypass. Crit Care Med 2020; 47:e700-e709. [PMID: 31149961 DOI: 10.1097/ccm.0000000000003820] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Cardiopulmonary bypass is associated with severe immune dysfunctions. Particularly, a cardiopulmonary bypass-related long-lasting immunosuppressive state predisposes patients to a higher risk of postoperative complications, such as persistent bacterial infections. This study was conducted to elucidate mechanisms of post-cardiopulmonary bypass immunosuppression. DESIGN In vitro studies with human peripheral blood mononuclear cells. SETTING Cardiosurgical ICU, University Research Laboratory. PATIENTS Seventy-one patients undergoing cardiac surgery with cardiopulmonary bypass (enrolled May 2017 to August 2018). INTERVENTIONS Peripheral blood mononuclear cells before and after cardiopulmonary bypass were analyzed for the expression of immunomodulatory cell markers by real-time quantitative reverse transcription polymerase chain reaction. T cell effector functions were determined by enzyme-linked immunosorbent assay, carboxyfluorescein succinimidyl ester staining, and cytotoxicity assays. Expression of cell surface markers was assessed by flow cytometry. CD15 cells were depleted by microbead separation. Serum arginine was measured by mass spectrometry. Patient peripheral blood mononuclear cells were incubated in different arginine concentrations, and T cell functions were tested. MEASUREMENTS AND MAIN RESULTS After cardiopulmonary bypass, peripheral blood mononuclear cells exhibited significantly reduced levels of costimulatory receptors (inducible T-cell costimulator, interleukin 7 receptor), whereas inhibitory receptors (programmed cell death protein 1 and programmed cell death 1 ligand 1) were induced. T cell effector functions (interferon γ secretion, proliferation, and CD8-specific cell lysis) were markedly repressed. In 66 of 71 patients, a not yet described cell population was found, which could be characterized as myeloid-derived suppressor cells. Myeloid-derived suppressor cells are known to impair immune cell functions by expression of the arginine-degrading enzyme arginase-1. Accordingly, we found dramatically increased arginase-1 levels in post-cardiopulmonary bypass peripheral blood mononuclear cells, whereas serum arginine levels were significantly reduced. Depletion of myeloid-derived suppressor cells from post-cardiopulmonary bypass peripheral blood mononuclear cells remarkably improved T cell effector function in vitro. Additionally, in vitro supplementation of arginine enhanced T cell immunocompetence. CONCLUSIONS Cardiopulmonary bypass strongly impairs the adaptive immune system by triggering the accumulation of myeloid-derived suppressor cells. These myeloid-derived suppressor cells induce an immunosuppressive T cell phenotype by increasing serum arginine breakdown. Supplementation with L-arginine may be an effective measure to counteract the onset of immunoparalysis in the setting of cardiopulmonary bypass.
Collapse
|
36
|
Pre-Analytical and Clinical Validation of a Dried Blood Spot Assay for Asymmetric Dimethylarginine and L-Arginine. J Clin Med 2020; 9:jcm9041072. [PMID: 32283799 PMCID: PMC7230730 DOI: 10.3390/jcm9041072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 02/07/2023] Open
Abstract
Asymmetric dimethylarginine (ADMA) inhibits nitric oxide (NO) synthesis. It is a risk marker for cardiovascular events and mortality in patients with cardiometabolic diseases and in population-based studies. Plasma or serum analysis of ADMA may be hampered by pre-analytical sample handling. We validated a dried blood spot (DBS) assay for ADMA and L-arginine and show here that this assay has excellent variabilities and reproducibilities. Filter paper is impregnated with the arginase inhibitor nor-NOHA (Nω-hydroxy-nor-Arginine) to avoid L-arginine degradation. Clinical validation of this DBS assay confirms elevated ADMA concentration in hemodialysis patients as compared to healthy controls, higher ADMA concentrations in men versus women, and elevated L-arginine concentration in subjects supplemented with L-arginine. The DBS assay was used in a cohort study involving 100 primarily healthy subjects in the Andean region to assess the impact of chronic intermittent hypoxia on ADMA and L-arginine; ADMA DBS concentration at sea level was prospectively associated with pulmonary hypertension after six months of exposure to 3500 m. In a cohort of 753 individuals, L-arginine/ADMA ratio significantly decreased with increasing number of traditional cardiovascular risk factors. Analysis of ADMA and L-arginine in DBS is a reliable and reproducible method for quantitation of these markers in field studies.
Collapse
|
37
|
Vora A, de Lemos JA, Ayers C, Grodin JL, Lingvay I. Association of Galectin-3 With Diabetes Mellitus in the Dallas Heart Study. J Clin Endocrinol Metab 2019; 104:4449-4458. [PMID: 31162551 DOI: 10.1210/jc.2019-00398] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022]
Abstract
CONTEXT Galectin-3 is a biomarker associated with inflammation and fibrosis in cardiac, liver, and renal disease. Galectin-3 is higher in overweight and obese individuals; whether an association with diabetes exists independent of weight is unknown. OBJECTIVE To evaluate if galectin-3 is associated with diabetes mellitus. DESIGN We performed measurements of galectin-3 among participants in the Dallas Heart Study (DHS) Phases 1 and 2 (DHS-1 and DHS-2; n = 3392, and n = 3194, respectively). Of these, 1989 participants were evaluated longitudinally in both studies. Associations of galectin-3 with prevalent and incident type 2 diabetes were determined using logistic regression models. Associations of galectin-3 with relevant biomarkers and fat compartments were evaluated using Spearman correlation coefficients and multivariable linear regression models, respectively. SETTING AND PARTICIPANTS DHS is a population-based, single-site, multiethnic study conducted in Dallas County, Texas, with oversampling to comprise 50% blacks. RESULTS Galectin-3 levels were associated with diabetes prevalence in DHS-1 [OR 1.56 per SD change in log-galectin (95% CI 1.41 to 1.73)] and DHS-2 [OR 1.86 (95% CI 1.67 to 2.06)]. Galectin-3 levels in DHS-1 also associated with incident diabetes mellitus over the 7.1 (interquartile range 6.6 to 7.6)-year follow-up period [OR 1.34 (95% CI 1.14 to 1.58)]. These associations maintained significance in models adjusted for traditional metabolic risk factors (age, sex, race, body mass index, and hypertension) and renal function. Galectin-3 levels correlated with levels of biomarkers implicated in inflammation (high-sensitivity C-reactive peptide, IL-18, monocyte chemoattractant protein 1, soluble TNF receptor 1A, myeloperoxidase), insulin secretion (C-peptide and C-peptide/homeostatic model assessment for insulin resistance), and subcutaneous adiposity. CONCLUSIONS Galectin-3 is associated with diabetes prevalence and incidence, possibly through the inflammatory pathway contributing to β-cell fibrosis and impaired insulin secretion.
Collapse
Affiliation(s)
- Amy Vora
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Texas
| | - James A de Lemos
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Texas
| | - Colby Ayers
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Texas
| | - Justin L Grodin
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Texas
| | - Ildiko Lingvay
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Texas
- Department of Clinical Sciences, University of Texas Southwestern Medical Center at Dallas, Texas
| |
Collapse
|
38
|
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: 0.4] [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
|
39
|
Association of Asymmetric Dimethylarginine and Diastolic Dysfunction in Patients with Hypertrophic Cardiomyopathy. Biomolecules 2019; 9:biom9070277. [PMID: 31337005 PMCID: PMC6681289 DOI: 10.3390/biom9070277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/29/2022] Open
Abstract
Despite genetic heterogeneity, early manifestation of diastolic dysfunction (DD) is common in hypertrophic cardiomyopathy (HCM). Nitric oxide (NO) may contribute to myocardial relaxation. NO synthases (NOS) use l-arginine (Arg) as a substrate, as asymmetric dimethylarginine (ADMA) is a direct endogenous inhibitor of NOS. This study aimed to analyze the association of Arg and its derivates, i.e., l-homoarginine (hArg), ADMA and symmetric dimethylarginine (SDMA), with DD in HCM patients. In 215 HCM patients (mean age 54 ± 15 years, 58% male) transmitral and mitral annulus velocities were echocardiographically analyzed. Plasma concentrations of Arg derivatives were measured by liquid chromatography tandem-mass spectrometry. In 143 (70%) patients suffering from DD, ADMA showed the strongest association with DD (0.66 ± 0.16, 0.72 ± 0.24, and 0.76 ± 0.26 µmol/L, p < 0.01 for trend). In linear regression analyses, positive association per standard deviation increase of ADMA was found with E-wave (beta coefficient (95% confidence interval): 4.72 (0.43–9.01); p < 0.05) and mean E/E’ (1.76 (0.73–2.79) p < 0.001). Associations were adjusted for age, sex, body mass index (BMI), diabetes mellitus, coronary artery disease, and arterial hypertension. Elevated ADMA is associated with the severity of DD in HCM. Higher ADMA level might lead to decreased NO production and thus an impaired myocardial relaxation pattern.
Collapse
|
40
|
Siques P, Brito J, Schwedhelm E, Pena E, León-Velarde F, De La Cruz JJ, Böger RH, Hannemann J. Asymmetric Dimethylarginine at Sea Level Is a Predictive Marker of Hypoxic Pulmonary Arterial Hypertension at High Altitude. Front Physiol 2019; 10:651. [PMID: 31191349 PMCID: PMC6545974 DOI: 10.3389/fphys.2019.00651] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Prolonged exposure to altitude-associated chronic hypoxia (CH) may cause high-altitude pulmonary hypertension (HAPH). Chronic intermittent hypobaric hypoxia (CIH) occurs in individuals who commute between sea level and high altitude. CIH is associated with repetitive acute hypoxic acclimatization and conveys the long-term risk of HAPH. As nitric oxide (NO) regulates pulmonary vascular tone and asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of NO synthesis, we investigated whether ADMA concentration at sea level predicts HAPH among Chilean frontiers personnel exposed to 6 months of CIH. Methods: In this prospective study, 123 healthy army draftees were subjected to CIH (5 days at 3,550 m, 2 days at sea level) for 6 months. In 100 study participants with complete data, ADMA, symmetric dimethylarginine (SDMA), L-arginine, arterial oxygen saturation (SaO2), systemic blood pressure, and hematocrit were assessed at months 0 (sea level), 1, 4, and 6. Acclimatization to altitude was determined using the Lake Louise Score (LLS) and the presence of acute mountain sickness (AMS). Echocardiography was performed after 6 months of CIH in 43 individuals with either good (n = 23) or poor (n = 20) acclimatization. Results: SaO2 acutely decreased at altitude and plateaued at 90% thereafter. ADMA increased and SDMA decreased during the study course. The incidence of AMS and the LLS was high after the first ascent (53 and 3.1 ± 2.4) and at 1 month of CIH (47 and 3.0 ± 2.6), but decreased to 20 and 1.4 ± 2.0 at month 6 (both p < 0.001). Eighteen participants (42%) showed a mean pulmonary arterial pressure (mPAP) >25 mm Hg, out of which 9 (21%) were classified as HAPH (mPAP ≥ 30 mm Hg). ADMA at sea level was significantly associated with mPAP at high altitude in month 6 (R = 0.413; p = 0.007). In ROC analysis, a cutoff for baseline ADMA of 0.665 μmol/L was determined to predict HAPH (mPAP > 30 mm Hg) with a sensitivity of 100% and a specificity of 63.6%. Conclusions: ADMA concentration increases during CIH. ADMA at sea level is an independent predictive biomarker of HAPH. SDMA concentration decreases during CIH and shows no association with HAPH. Our data support a role of impaired NO-mediated pulmonary vasodilation in the pathogenesis of HAPH.
Collapse
Affiliation(s)
- Patricia Siques
- Institute of Health Studies, Universidad Arturo Prat, Iquique, Chile.,Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and its Health Sequelae, Hamburg, Germany and Iquique, Chile
| | - Julio Brito
- Institute of Health Studies, Universidad Arturo Prat, Iquique, Chile.,Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and its Health Sequelae, Hamburg, Germany and Iquique, Chile
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eduardo Pena
- Institute of Health Studies, Universidad Arturo Prat, Iquique, Chile.,Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and its Health Sequelae, Hamburg, Germany and Iquique, Chile
| | - Fabiola León-Velarde
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and its Health Sequelae, Hamburg, Germany and Iquique, Chile.,Department of Biological and Physiological Sciences, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Juan José De La Cruz
- Department of Preventive Medicine and Public Health, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rainer H Böger
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and its Health Sequelae, Hamburg, Germany and Iquique, Chile.,Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Juliane Hannemann
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and its Health Sequelae, Hamburg, Germany and Iquique, Chile.,Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
41
|
Quintana-Villamandos B, González MDC, Delgado-Martos MJ, Gutiérrez-Arzapalo PY, Böger RH, Lüneburg N, Muñoz D, Delgado-Baeza E. The protective effect of dronedarone on the structure and mechanical properties of the aorta in hypertensive rats by decreasing the concentration of symmetric dimethylarginine (SDMA). PLoS One 2019; 14:e0216820. [PMID: 31112581 PMCID: PMC6529158 DOI: 10.1371/journal.pone.0216820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND AIMS Dronedarone is a new multichannel-blocking antiarrhythmic for the treatment of patients with atrial fibrillation. Our group has demonstrated that dronedarone produces regression of cardiac remodeling; however, its effect on the remodeling of the elastic arteries has not yet been reported. We aim to assess the effects of dronedarone on the regression of thoracic aortic remodeling in spontaneously hypertensive rats (SHRs). METHOD Ten-month-old male SHRs were randomly assigned to an intervention group (SHR-D), where the animals received dronedarone treatment (100 mg/kg), to a control group (SHR) where rats were given vehicle, or to a group (SHR-A) where they were given amiodarone. A fourth group of normotensive control rats (Wistar-Kyoto rats, WKY) was also added. After two weeks of treatment, we studied the structure, the elastic fiber content of the thoracic aorta using histological techniques and confocal microscopy, and the vascular mechanical properties using an organ bath and isometric tension analysis. A mass spectrometric determination of symmetric dimethylarginine (SDMA) concentrations was performed. RESULTS SHR group developed the classic remodeling expected from the experimental model: outward hypertrophic remodeling, increased elastic fiber content and wall stiffness. However, the SHR-D group showed statistically significantly lower values for aortic tunica media thickness, wall to lumen ratio, external diameter, cross-sectional area, volume density of the elastic fibers, wall stiffness, and aortic SDMA concentration when compared to the SHR group. These parameters were similar in the SHR and SHR-A groups. Interestingly, the values for tunica media thickness, volume density of the elastic fibers, wall stiffness, and SDMA concentration obtained from the SHR-D group were similar to those measured in the WKY group. CONCLUSION These results suggest that dronedarone improves the structure and passive mechanical properties of the thoracic aorta in hypertensive rats, and that this protective effect could be associated with a reduction in the concentration of aortic SDMA.
Collapse
Affiliation(s)
- Begoña Quintana-Villamandos
- Department of Anesthesiology, Hospital Gregorio Marañón, Madrid, Spain
- Departament of Pharmacology and Toxicology, Faculty of Medicine, University Complutense de Madrid, Madrid, Spain
- * E-mail:
| | | | - María Jesús Delgado-Martos
- Molecular Biology Laboratory, Department Experimental Medicine and Surgery, Hospital Gregorio Marañón, Madrid, Spain
| | | | - Rainer H. Böger
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicole Lüneburg
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David Muñoz
- Department of Experimental Surgery, University Autónoma of Madrid, Madrid, Spain
| | - Emilio Delgado-Baeza
- Molecular Biology Laboratory, Department Experimental Medicine and Surgery, Hospital Gregorio Marañón, Madrid, Spain
| |
Collapse
|
42
|
Dronedarone induces regression of coronary artery remodeling related to better global antioxidant status. Hypertens Res 2019; 42:1485-1494. [PMID: 30992541 DOI: 10.1038/s41440-019-0257-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 02/05/2019] [Accepted: 03/12/2019] [Indexed: 01/23/2023]
Abstract
Our group previously demonstrated that dronedarone induces regression of left ventricular hypertrophy in spontaneously hypertensive rats (SHRs). We assessed changes in vascular remodeling and oxidative stress following short-term use of this agent. The coronary artery was isolated from 10-month-old male SHRs treated with 100 mg kg-1 dronedarone once daily for 14 days (SHR-D group), and age-matched untreated SHRs were used as hypertensive controls. We analyzed the geometry and composition of the artery and constructed dose-response curves for acetylcholine and serotonin (5-HT). We calculated a global score (OXY-SCORE) from plasma biomarkers of oxidative status: carbonyl levels, thiol levels, reduced glutathione levels, total antioxidant capacity, and superoxide anion scavenging activity. Finally, we analyzed asymmetric dimethylarginine (ADMA) concentrations in plasma. Dronedarone significantly decreased wall thickness (medial and adventitial layer thickness and cell count) and the cross-sectional area of the artery. Dronedarone significantly improved endothelium-dependent relaxation and reduced the contraction induced by 5-HT. The OXY-SCORE was negative in the SHR model group (suggesting an enhanced oxidative status) and was positive in the SHR-D group (suggesting enhanced antioxidant defense). Dronedarone significantly decreased the concentrations of ADMA. We conclude that dronedarone improves coronary artery remodeling in SHRs. The better global antioxidant status after treatment with dronedarone and decreased plasma ADMA levels could contribute to the cardiovascular protective effect of dronedarone.
Collapse
|
43
|
Schwemer TF, Deutscher N, Diermann N, Böger R, Schwedhelm E, Blankenberg S, Friedrich FW. Effect of ranolazine on plasma arginine derivatives and urinary isoprostane 8-iso-PGF 2α in patients with myocardial infarction in the randomized RIMINI-Trial. Sci Rep 2019; 9:5708. [PMID: 30952941 PMCID: PMC6450888 DOI: 10.1038/s41598-019-42239-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 03/13/2019] [Indexed: 12/20/2022] Open
Abstract
The purpose of the present study was to assess whether 6-week ranolazine application on top of guideline-based treatment impacts on the arginine/NO pathway and urinary isoprostane 8-iso-PGF2α as marker of oxidative stress in patients directly after a myocardial infarction. 20 patients with unstable angina pectoris and proof of acute cardiac ischemia entered the study. 10 subjects received the study drug ranolazine in addition to standard treatment, the others received only standard treatment. Urine and venous blood were collected before and after treatment. At the end of the study and compared to baseline, homoarginine levels had increased in the control group. This was not the case in ranolazine-patients. Interestingly, in ranolazine-treated-patients arginine plasma levels were significantly higher at the end of the study than at baseline (difference +26 µmol/L, 95% CI 8.6 to 44 µmol/L). ADMA and SDMA levels were not different. Urine levels of the oxidative stress marker 8-iso-PGF2α tended to be lower in ranolazine-treated patients (−144 pmol/mg creatinine). Findings of this hypothesis-driven study give evidence that ranolazine treatment enhances arginine plasma levels and lowers oxidative stress.
Collapse
Affiliation(s)
| | | | | | - Rainer Böger
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.,Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edzard Schwedhelm
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.,Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Blankenberg
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.,University Heart Center Hamburg, Hamburg, Germany
| | - Felix W Friedrich
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
44
|
Guanidino compound ratios are associated with stroke etiology, internal carotid artery stenosis and CHA2DS2-VASc score in three cross-sectional studies. J Neurol Sci 2019; 397:156-161. [DOI: 10.1016/j.jns.2018.12.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/23/2018] [Accepted: 12/31/2018] [Indexed: 02/07/2023]
|
45
|
Winkler MS, Nierhaus A, Rösler G, Lezius S, Harlandt O, Schwedhelm E, Böger RH, Kluge S. Symmetrical (SDMA) and asymmetrical dimethylarginine (ADMA) in sepsis: high plasma levels as combined risk markers for sepsis survival. Crit Care 2018; 22:216. [PMID: 30231905 PMCID: PMC6145330 DOI: 10.1186/s13054-018-2090-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 06/07/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Nitric oxide (NO) regulates processes involved in sepsis progression, including vascular and immune function. NO is generated by nitric oxide synthases (NOS) from L-arginine. Cellular L-arginine uptake is inhibited by symmetric dimethylarginine (SDMA) and asymmetric dimethylarginine (ADMA) is a competitive inhibitor of NOS. Increased inhibitor blood concentrations lead to reduce NO bioavailability. The aim of this study was to determine whether plasma concentrations of SDMA and ADMA are markers for sepsis survival. METHOD This prospective, single center study involved 120 ICU patients with sepsis. Plasma SDMA and ADMA were measured on admission (day 1), day 3 and day 7 by mass spectrometry together with other laboratory markers. The sequential organ failure assessment (SOFA) score was used to evaluate sepsis severity. Survival was documented until day 28. Groups were compared using the Mann-Whitney U test, chi-squared test or non-parametric analysis of variance (ANOVA). Mortality was assessed using Kaplan-Meier curves and compared using the log-rank test. Specific risk groups were identified using a decision tree algorithm. RESULTS Median plasma SDMA and ADMA levels were significantly higher in non-survivors than in survivors of sepsis: SDMA 1.14 vs. 0.82 μmol/L (P = 0.002) and ADMA 0.93 vs. 0.73 μmol/L (P = 0.016). ANOVA showed that increased plasma SDMA and ADMA concentrations were significantly associated with SOFA scores. The 28-day mortality was compared by chi-square test: for SDMA the mortality was 12% in the lower, 25% in the intermediate and 43% in the 75th percentile (P = 0.018); for ADMA the mortality was 18-20% in the lower and intermediate but 48% in the 75th percentile (P = 0.006). The highest mortality (61%) was found in patients with plasma SDMA > 1.34 together with ADMA levels > 0.97 μmol/L. CONCLUSIONS Increased plasma concentrations of SDMA and ADMA are associated with sepsis severity. Therefore, our findings suggest reduced NO bioavailability in non-survivors of sepsis. One may use individual SDMA and ADMA levels to identify patients at risk. In view of the pathophysiological role of NO we conclude that the vascular system and immune response are most severely affected when SDMA and ADMA levels are high.
Collapse
Affiliation(s)
- Martin Sebastian Winkler
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
- Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52 20246 Hamburg, Germany
| | - Axel Nierhaus
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Gilbert Rösler
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Susanne Lezius
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Olaf Harlandt
- Department of Internal Medicine II, Asklepios Klinik Nord-Heidberg, Tangstedter Landstr. 400, 22417 Hamburg, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Rainer H. Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| |
Collapse
|
46
|
Loso J, Lund N, Avanesov M, Muschol N, Lezius S, Cordts K, Schwedhelm E, Patten M. Serum Biomarkers of Endothelial Dysfunction in Fabry Associated Cardiomyopathy. Front Cardiovasc Med 2018; 5:108. [PMID: 30159316 PMCID: PMC6104487 DOI: 10.3389/fcvm.2018.00108] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/17/2018] [Indexed: 12/13/2022] Open
Abstract
Background: Fabry disease (FD) is characterized by early development of vasculopathy and endothelial dysfunction. However, it is unclear whether these findings also play a pivotal role in cardiac manifestation. As Fabry cardiomyopathy (FC) is the leading cause of death in FD, we aimed to gather a better insight in pathological mechanisms of the disease. Methods: Serum samples were obtained from 17 healthy controls, 15 FD patients with and 7 without FC. FC was defined by LV wall thickening of >12 mm in cardiac magnetic resonance imaging and serum level of proBNP, high sensitive Troponin T (hsT), and globotriaosylsphingosine (lyso-GB3) were obtained. A multiplex ELISA-Assay for 23 different angiogenesis markers was performed in pooled samples. Markers showing significant differences among groups were further analyzed in single samples using specific Elisa antibody assays. L-homoarginine (hArg), L-arginine, asymmetric (ADMA), and symmetric Dimethylarginine (SDMA) were quantified by liquid chromatography—mass spectrometry. Results: Angiostatin and matrix metalloproteinase 9 (MMP-9) were elevated in FD patients compared to controls independently of the presence of FC (angiostatin: 98 ± 25 vs. 75 ± 15 ng/mL; p = 0.001; MMP-9: 8.0 ± 3.4 vs. 5.0 ± 2.4 μg/mL; p = 0.002). SDMA concentrations were highest in patients with FC (0.90 ± 0.64 μmol/l) compared to patients without (0.57 ± 0.10 μmol/l; p = 0.027) and vs. controls (0.58 ± 0.12 μmol/l; p = 0.006) and was positively correlated with indexed LV-mass (r = 0.61; p = 0.003), hsT (r = 0.56, p = 0.008), and lyso-Gb3 (r = 0.53, p = 0.013). Accordingly, the ratio of L-homoarginine to SDMA (hArg/SDMA) was lowest in patients with FC (2.63 ± 1.78) compared to controls (4.16 ± 1.44; p = 0.005). For L-arginine, hArg and ADMA no significant differences among groups could be detected, although a trend toward higher ADMA and lower hArg levels could be observed in the FC group. Furthermore, a significant relationship between kidney and cardiac function could be revealed (p = 0.045). Conclusion: Elevated MMP-9 and angiostatin levels suggest an increased extracellular matrix turnover in FD patients. Furthermore, endothelial dysfunction may also be involved in FC, as SDMA and hArg/SDMA are altered in these patients.
Collapse
Affiliation(s)
- Jefferson Loso
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Natalie Lund
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Maxim Avanesov
- Department of Diagnostic and Interventional Radiology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Nicole Muschol
- Department of Pediatrics, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Susanne Lezius
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Kathrin Cordts
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research e.V.), Hamburg, Germany
| | - Edzard Schwedhelm
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg- Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research e.V.), Hamburg, Germany
| | - Monica Patten
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research e.V.), Hamburg, Germany
| |
Collapse
|
47
|
Baldassarri F, Schwedhelm E, Atzler D, Böger RH, Cordts K, Haller B, Pressler A, Müller S, Suchy C, Wachter R, Düngen HD, Hasenfuss G, Pieske B, Halle M, Edelmann F, Duvinage A. Relationship between exercise intervention and NO pathway in patients with heart failure with preserved ejection fraction. Biomarkers 2018; 23:540-550. [DOI: 10.1080/1354750x.2018.1460762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Flavia Baldassarri
- Department of Prevention, Rehabilitation and Sports Medicine, Technische Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Dorothee Atzler
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Rainer H. Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Kathrin Cordts
- Institute of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Bernhard Haller
- Institute of Medical Statistics and Epidemiology, Technische Universität München, Munich, Germany
| | - Axel Pressler
- Department of Prevention, Rehabilitation and Sports Medicine, Technische Universität München, Munich, Germany
| | - Stephan Müller
- Department of Prevention, Rehabilitation and Sports Medicine, Technische Universität München, Munich, Germany
| | - Christiane Suchy
- Department of Prevention, Rehabilitation and Sports Medicine, Technische Universität München, Munich, Germany
| | - Rolf Wachter
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Hans-Dirk Düngen
- Department of Cardiology, Charite´ – Universita¨tsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Burkert Pieske
- Department of Cardiology, Charite´ – Universita¨tsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Department of Cardiology, Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany
| | - Martin Halle
- Department of Prevention, Rehabilitation and Sports Medicine, Technische Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Frank Edelmann
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Department of Cardiology, Charite´ – Universita¨tsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - André Duvinage
- Department of Prevention, Rehabilitation and Sports Medicine, Technische Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
48
|
Cardiomyocyte dimethylarginine dimethylaminohydrolase-1 (DDAH1) plays an important role in attenuating ventricular hypertrophy and dysfunction. Basic Res Cardiol 2017; 112:55. [PMID: 28819685 DOI: 10.1007/s00395-017-0644-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 08/01/2017] [Indexed: 12/17/2022]
Abstract
Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthases that limits nitric oxide bioavailability. Dimethylarginine dimethylaminohydrolase-1 (DDAH1) exerts a critical role for ADMA degradation and plays an important role in NO signaling. In the heart, DDAH1 is observed in endothelial cells and in the sarcolemma of cardiomyocytes. While NO signaling is important for cardiac adaptation to stress, DDAH1 impact on cardiomyocyte homeostasis is not clear. Here we used the MerCreMer-LoxP model to specifically disrupt cardiomyocyte DDAH1 expression in adult mice to determine the physiological impact of cardiomyocyte DDAH1 under basal conditions and during hypertrophic stress imposed by transverse aortic constriction (TAC). Under control conditions, cardiomyocyte-specific DDAH1 knockout (cDDAH KO) had no detectable effect on plasma ADMA and left ventricular (LV) hypertrophy or function in adult or aging mice. In response to TAC, DDAH1 levels were elevated 2.5-fold in WT mice, which exhibited no change in LV or plasma ADMA content and moderate LV hypertrophy and LV dysfunction. In contrast, cDDAH1 KO mice exposed to TAC showed no increase in LV DDAH1 expression, slightly increased LV tissue ADMA levels, no increase in plasma ADMA, but significantly exacerbated LV hypertrophy, fibrosis, nitrotyrosine production, and LV dysfunction. These findings indicate cardiomyocyte DDAH1 activity is dispensable for cardiac function under basal conditions, but plays an important role in attenuating cardiac hypertrophy and ventricular remodeling under stress conditions, possibly through locally confined regulation of subcellular ADMA and NO signaling.
Collapse
|
49
|
Winkler MS, Kluge S, Holzmann M, Moritz E, Robbe L, Bauer A, Zahrte C, Priefler M, Schwedhelm E, Böger RH, Goetz AE, Nierhaus A, Zoellner C. Markers of nitric oxide are associated with sepsis severity: an observational study. Crit Care 2017; 21:189. [PMID: 28709458 PMCID: PMC5513203 DOI: 10.1186/s13054-017-1782-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 06/29/2017] [Indexed: 02/23/2023] Open
Abstract
Background Nitric oxide (NO) regulates processes involved in sepsis progression, including vascular function and pathogen defense. Direct NO measurement in patients is unfeasible because of its short half-life. Surrogate markers for NO bioavailability are substrates of NO generating synthase (NOS): L-arginine (lArg) and homoarginine (hArg) together with the inhibitory competitive substrate asymmetric dimethylarginine (ADMA). In immune cells ADMA is cleaved by dimethylarginine-dimethylaminohydrolase-2 (DDAH2). The aim of this study was to investigate whether concentrations of surrogate markers for NO bioavailability are associated with sepsis severity. Method This single-center, prospective study involved 25 controls and 100 patients with surgical trauma (n = 20), sepsis (n = 63), or septic shock (n = 17) according to the Sepsis-3 definition. Plasma lArg, hArg, and ADMA concentrations were measured by mass spectrometry and peripheral blood mononuclear cells (PBMCs) were analyzed for DDAH2 expression. Results lArg concentrations did not differ between groups. Median (IQR) hArg concentrations were significantly lower in patient groups than controls, being 1.89 (1.30–2.29) μmol/L (P < 0.01), with the greatest difference in the septic shock group, being 0.74 (0.36–1.44) μmol/L. In contrast median ADMA concentrations were significantly higher in patient groups compared to controls, being 0.57 (0.46–0.65) μmol/L (P < 0.01), with the highest levels in the septic shock group, being 0.89 (0.56–1.39) μmol/L. The ratio of hArg:ADMA was inversely correlated with disease severity as determined by the Sequential Organ Failure Assessment (SOFA) score. Receiver-operating characteristic analysis for the presence or absence of septic shock revealed equally high sensitivity and specificity for the hArg:ADMA ratio compared to the SOFA score. DDAH2 expression was lower in patients than controls and lowest in the subgroup of patients with increasing SOFA. Conclusions In patients with sepsis, plasma hArg concentrations are decreased and ADMA concentrations are increased. Both metabolites affect NO metabolism and our findings suggest reduced NO bioavailability in sepsis. In addition, reduced expression of DDAH2 in immune cells was observed and may not only contribute to blunted NO signaling but also to subsequent impaired pathogen defense.
Collapse
Affiliation(s)
- Martin Sebastian Winkler
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany. .,Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Maximilian Holzmann
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Eileen Moritz
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Linda Robbe
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Antonia Bauer
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Corinne Zahrte
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Marion Priefler
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Rainer H Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Alwin E Goetz
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Axel Nierhaus
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Christian Zoellner
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,Center for Anesthesiology and Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| |
Collapse
|
50
|
Wiśniewski J, Fleszar MG, Piechowicz J, Krzystek-Korpacka M, Chachaj A, Szuba A, Lorenc-Kukula K, Masłowski L, Witkiewicz W, Gamian A. A novel mass spectrometry-based method for simultaneous determination of asymmetric and symmetric dimethylarginine, l
-arginine and l
-citrulline optimized for LC-MS-TOF and LC-MS/MS. Biomed Chromatogr 2017; 31. [DOI: 10.1002/bmc.3994] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 03/20/2017] [Accepted: 04/19/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Jerzy Wiśniewski
- Department of Medical Biochemistry; Wroclaw Medical University; Wroclaw Poland
| | - Mariusz G. Fleszar
- Department of Medical Biochemistry; Wroclaw Medical University; Wroclaw Poland
| | - Joanna Piechowicz
- Department of Medical Biochemistry; Wroclaw Medical University; Wroclaw Poland
| | | | - Angelika Chachaj
- Department of Angiology; Wroclaw Medical University; Wroclaw Poland
- Department of Internal Medicine; 4th Military Hospital; Wroclaw Poland
| | - Andrzej Szuba
- Department of Angiology; Wroclaw Medical University; Wroclaw Poland
- Department of Internal Medicine; 4th Military Hospital; Wroclaw Poland
| | - Katarzyna Lorenc-Kukula
- Shimadzu Center For Advanced Analytical Chemistry; The University of Texas at Arlington; TX USA
| | - Leszek Masłowski
- Department of Vascular Surgery; Regional Specialist Hospital; Wroclaw Poland
| | - Wojciech Witkiewicz
- Department of Surgical Oncology; Regional Specialist Hospital; Wroclaw Poland
- Research and Development Center at Regional Specialist Hospital; Wroclaw Poland
| | - Andrzej Gamian
- Department of Medical Biochemistry; Wroclaw Medical University; Wroclaw Poland
- Wroclaw Research Center EIT+; Wroclaw Poland
| |
Collapse
|